Casein kinase 2 inhibition modulates the DNA damage response but fails to radiosensitize malignant glioma cells

  • Authors:
    • Jérôme Kroonen
    • Maria Artesi
    • Valérie Capraro
    • Minh-Tuan Nguyen-Khac
    • Marie Willems
    • Arnab Chakravarti
    • Vincent Bours
    • Pierre A. Robe
  • View Affiliations

  • Published online on: May 18, 2012     https://doi.org/10.3892/ijo.2012.1489
  • Pages: 776-782
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Inhibitors of casein kinase 2 (CK2), a regulator of cell proliferation and mediator of the DNA damage response, are being evaluated in clinical trials for the treatment of cancers. Apigenin was capable of inhibiting the activation of CK2 following γ irradiation in LN18 and U87 malignant glioma cells. Apigenin and siRNA-mediated CK2 protein depletion further inhibited NF-κB activation and altered the Tyr68 phosphorylation of Chk2 kinase, a DNA damage response checkpoint kinase, following irradiation. However, CK2 inhibition did not decrease the ability of these glioma cells to repair double-strand DNA breaks, as assessed by COMET assays and γ-H2Ax staining. Likewise, apigenin and siRNA-induced depletion of CK2 failed to sensitize glioma cells to the cytotoxic effect of 2 to 10 G-rays of γ irradiation, as assessed by clonogenic assays. These results contrast with those found in other cancer types, and urge to prudence regarding the inclusion of malignant glioma patients in clinical trials that assess the radiosensitizing role of CK2 inhibitors in solid cancers.

Introduction

Malignant astrocytomas represent the most frequent type of malignant brain tumors and are characterized by a strong resistance to therapies and a dismal prognosis (1). Among the factors that determine this resistance to treatment, anti-apoptotic signaling, for instance through constitutive NF-κB pathway activation (2,3), and altered DNA-damage response (4), are believed to play major roles.

Casein kinase 2 (CK2), an ubiquitous serine threonine kinase, consists of tetramers of 2 catalytic and 2 regulatory subunits. It has recently gained interest in the field of cancer research as both a regulator of proliferation and survival pathways and a modulator of the DNA-repair machinery (5,6). CK2 was thus shown to regulate NF-κB and STAT3 activation, P53 function (7), PTEN activity, Akt-dependant signaling, mTOR stability and SIRT-dependent protein acetylation (6,813). CK2 also regulates the function of several enzymes of the DNA-repair and DNA-damage sensing machinery, such as XRCC1 and 4, Rad9 and DNA-PK (1416). As a corollary, pre-clinical studies have shown that CK2 inhibitors elicit anti-tumoral effects against leukemias, prostate carcinomas, breast cancers, and some PTEN or TP53 mutated malignant gliomas (17,18). Based on these reports, CK2 inhibitors entered the field of clinical trials (8,1922). Among them, apigenin is a naturally occurring plant flavonoid and a specific inhibitor of the catalytic subunits of CK2 (8,23,24). It was shown to reduce the proliferation and to induce apoptosis in several carcinoma cells (2527) as well as in some human glioma cell lines (28), and has recently been used in a phase II trial for the prevention of colorectal cancer recurrence (NCT00609310).

Given this growing interest of clinicians and the industry for CK2 inhibitors, and in view of the fundamental yet disappointing role of radiation therapy for the treatment of malignant gliomas (1), we investigated whether CK2 inhibition would alter the radiation-induced DNA repair response and whether these tumors could be radiosensitized.

Materials and methods

Cell cultures, reagents and siRNA

Cell lines U87 and LN18 were obtained from the American Type Culture Collection (ATCC) and grown in DMEM (Dulbecco’s modified Eagle’s medium, Gibco, Gent, Belgium) containing 10% of fetal bovine serum (FBS, Gibco) and penicillin. Cultures were maintained at 37°C under a humidified atmosphere containing 5% carbon dioxide.

Apigenin was purchased from Sigma (Bornem, Belgium), dissolved in dimethylsulfoxide (DMSO) and used at final concentration of 40 μM (stock solution, 100 mM). Control cells were treated with a similar final concentration of DMSO as the apigenin-treated cells. Irradiations of cell lines were conducted with a research irradiator (Gammacell 40 Exactor, Theratronics, Stockley Park, UK).

Subconfluent cultured cells were transfected with 50 nmol/l ON-TARGETplus non-targeting pool or SMARTpool human CSNK2A1 siRNA from Dharmacon (Fisher Scientific, Tournai, Belgium) using oligofectamine (Invitrogen, Gent, Belgium) according to the manufacturer’s instructions. Cells were harvested and assayed 48 h after transfection. CK2 depletion was controlled using western blot analysis of the expression of CK2-α.

CK2 and IKK-β kinase assays

Cells were lysed using RIPA buffer extraction kit (Santa Cruz Biotechnology) and 300 μg of protein were taken for immunoprecipitation. After a pre-cleared step, supernatant were incubated with an anti-CK2 antibody (clone 1AD9, Millipore, Overijse, Belgium) under rotary agitation for 4 h at 4°C. GammaBind G Sephorase beads (25 μl/samples, GE Healthcare, Diegem, Belgium) were then added to the sample and incubated on a rotating system overnight at 4°C. After three washes, immunoprecipitated proteins were processed with the CK2 assay kit (Upstate, Millipore) or the IKK-β kinase assay kit (Cell Signaling, Bioke, Leiden, The Netherlands), according to the manufacturer’s instructions.

NF-κB transcription assay

Cells were co-transfected by using TransIT-2020 transfection reagent (Mirus, Eke, Belgium) with: i) a luciferase-coupled reporter gene for NF-κB and ii) a Renilla luciferase reporter driven by a constitutive promoter. Radiation (10 Gy) and apigenin treatment (40 μM) effects on NF-κB transcriptional activity were assessed 24 h later. Briefly, cells were lysed and luciferase activities were measured according to the manufacturer’s instructions for the Dual Luciferase Assay System (Promega, Leiden, The Netherlands) and using a Victor luminometer (PerkinElmer, Zaventem, Belgium). The relative NF-κB luciferase activity was normalized to the one of the Renilla.

Western blot analysis

10% polyacrylamide precast gels (Mini Protean TGX, Bio-Rad, Nazareth Eke, Belgium) were run for 30 min at 200 volts with nuclear extract (20 μg/well) obtained from irradiated and previously apigenin or DMSO treated cells. Protein extracts were obtained using conventional RIPA buffer and phosphatase inhibitors. After transfer to a PVDF membrane (Roche, Vilvoorde, Belgium) for 2 h at 300 mA and blocking with Tris buffered saline containing 0.2% Tween plus 5% dry milk powder, membranes were incubated overnight at 4°C in the presence of primary antibody directed against phospho(Thr68)-Chk2 (Cell Signaling, Bioké, Leiden, The Netherlands). A horseradish peroxidase-coupled secondary antibody was then incubated, and peroxidase activity was evidenced with the Super Signal West Pico Chemiluminescent substrate (Thermo Fisher Scientific, Aalst, Belgium) and the ImageQuant LAS 4000 Mini Biomolecular Imager (GE Healthcare).

Cell survival assays

Cell survival in response to apigenin treatment and radiation was assayed using clonogenic assays and MTS tests (One Solution Cell Proliferation Assay, Promega). Clonogenic assays were performed on cells plated at low-density as described previously (29) and as recommended by the manufacturer’s protocol for MTS assays.

DNA repair assays

Single-cell gel electrophoresis under alkaline conditions and flow cytometry measurement of phosphorylated γ-histone 2Ax (γ-H2Ax) foci were used to identify ds-DNA breaks and associated repair mechanisms, respectively.

ds-DNA breaks following apigenin (or DMSO) treatment and radiation were detected by using the CometAssay HT kit (Trevigen, Sanbio, Uden, The Netherlands). Briefly, single cells embedded in agarose were lysed to remove proteins and where then submitted to electrophorese. Staining was performed with SYBR green I (Trevigen) for 15 min. The slides were examined under a fluorescent microscope (Zeiss Axiovert 10 microscope, Carl Zeiss) and DNA tail lengths were quantified in a blinded manner by counting a minimum of 50 cells per condition in independent experiments.

Treated cells were harvested at different times and were prepared for flow cytometer analysis. Approximately 2.5×106 cells/ml were resuspended in 250 μl of PBS and fixed by adding the same amount of 4% paraformaldehyde (PFA, Merck, Overijse, Belgium). After permeabilization and blocking with PBS containing 0.5% Triton X-100 (Acros Organics, Geel, Belgium) and 5% donkey serum (Jackson Immunoresearch Laboratories, Newmarket, UK) for 20 min, an anti-phosphorylated Ser139 γ-H2Ax mouse monoclonal antibody (1:500, Millipore) was incubated for 90 min at room temperature. Three PBS washes later, we incubated cells with an FITC-conjugated secondary antibody (1:500, Jackson Immunoresearch Laboratories). Indirect immunofluorescence staining was immediately analyzed after three more PBS washes (FACS Calibur, BD Biosciences, Erembodegem, Belgium).

Statistical analysis

Statistical analyses were performed using the Prism 5.0c for Mac software (Graphpad Inc., La Jolla, CA). One-way ANOVA and Mann-Whitney U tests were performed when appropriate and as described in the results section.

Results

Irradiation-induced CK2 kinase activity in malignant glioma cells

Exposure of LN18 and U87 cells to ionizing radiations (γ rays, 4 Gy) increased the catalytic activity of CK2 within 30 min, by respectively 25±5% and 45±2.5%. Both the basal and radiation-induced CK2 activities were significantly abolished by pretreatment of the cell cultures with 40 μM Apigenin for 1 h (mean ± SD, n=3, P<0.05 for both, ANOVA with Tukey’s post tests; Fig. 1).

Irradiation-induced NF-κB activation in malignant glioma cells

Ionizing radiation activates NF-κB in tumors and glioblastomas via an ATM-NEMO-IKK-kinase dependent pathway (30). UV-induced DNA damage, however, also activates CK2 (31), leading to an IKK-kinase-independent C-terminal phosphorylation and degradation of I-κBα, and NF-κB activation (32). In LN18 and in U87 cells, ionizing irradiation (10 Gy) increased within 1 h the activity of an NF-κB-driven luciferase reporter gene by 31±6.6% and 66±34%, respectively, (mean ± SD, n=3, P<0.05, one-way ANOVA with Tukey’s post tests). The baseline activity of the reporter gene was inhibited following apigenin treatment and remained significantly reduced in these cells despite irradiation (P<0.05, 40 μM, Fig. 2).

CK2 inhibition and DNA-repair in malignant glioma cells

CK2 has recently emerged as a regulator of the DNA damage response machinery (33). We thus performed COMET assays to measure the influence of CK2 inhibition on ds-DNA break formation in U87 and LN18 cells following γ irradiation (10 Gy). As shown in Fig. 3A, si-mediated CK2 depletion slightly decreased the peak amplitude of COMET tails in LN18 cells 3 h following a 10 Gy irradiation (P<0.05, Mann-Witney U test). It however had the opposite effect in U87 cells (P<0.05, Mann-Witney U test). The mean tail amplitude returned to baseline in mock-treated and siCK2-treated LN18 cells after 24 h. Tail size also returned to baseline in siCK2-treated U87 cells, in sharp contrast to mock-transfected cells where tails still remained significantly longer than at baseline at this time point (P<0.0001, Mann-Witney U test).

We also assessed the kinetics of γ-H2Ax foci formation in LN18 and U87 cells treated with apigenin (40 μM) using FACS cytometry. In both cell types, radiation treatment (10 Gy) increased the amount of γ-H2Ax immunoreactivity with respect to baseline conditions, with a peak within 1 to 3 h. γ-H2Ax signal returned towards baseline in control and apigenin-treated in both cell types within 24 h. Apigenin treatment did not alter these post-irradiation kinetics of γ-H2Ax immunoreactivity (data not shown).

CK2 is also known to inhibit the DNA-repair kinase DNA-PK (15), and the Chk2 checkpoint kinase is phosphorylated on tyrosine 68 by DNA-PK following irradiation (34). Tyr68 phosphorylation of Chk2 was induced in LN18 and U87 within 15 min after irradiation (4 Gy). This event was potentiated and more durable in both cell types by a pre-treatment with 40 μM apigenin (Fig. 3B).

CK2 inhibition and cell survival following γ irradiation

Both U87 and LN18 cells displayed a moderate but significant reduction in viability following 4 G-rays of γ irradiation (25.5±13.2% and 27±19.5%, respectively, P<0.05, one-way ANOVA) as assessed using an MTS test (Fig. 4A). This viability was not further reduced following co-treatment with 40 μM apigenin. At this concentration, apigenin treatment also failed to radiosensitize U87 and LN18 cells in clonogenic assays (Fig. 4B, upper pannel).

As CK2-independent effects of apigenin have been reported (35), we also assessed the effect of siRNA-mediated CK2 kinase depletion on the radiosensitization of malignant glioma cells. The clonogenic survival of U87 and LN18 cells treated with CK2-targetting siRNA prior to irradiation did not differ from scramble siRNA treated controls (Fig. 4B, lower pannel).

Discussion

CK2 has recently appeared as a regulator of ds-DNA break (DSB)-triggered signaling cascades in normal, carcinoma and even in some malignant glioma cells (36).

Accordingly, CK2-α, the active kinase subunit of CK2, was activated within minutes of radiation treatment in our malignant glioma cells. siRNA-mediated CK2 depletion significantly increased the maximal peak of DSB in LN18 cells, but not in U87 cells. However, CK2 knock-down did not inhibit the repair of ds-DNA breaks in our cell lines, but even slightly improved it, as evidenced by the normalization of COMET assays within 24 h after irradiation in both cell lines and the faster return of γ-H2Ax immunoreactivity towards the baseline in U87 cells following apigenin treatment.

In standard conditions, homologous recombination (HR) only plays an accessory role in ds-DNA repair following ionizing radiation in gliomas, and these tumors rather proceed via non-homologous end joining (NHEJ) (37). During NHEJ, CK2 phosphorylates XRCC4 and helps recruit repair enzymes like PNKP and APLF to this scaffold protein (13,15,36,38,39). According to this, and in contrast to our COMET and γ-H2Ax findings, CK2 inhibition should thus inhibit DNA repair. In better agreement with our results however, CK2 inhibition did not impair ds-DNA rejoining in fibroblasts or colon carcinoma cells (40). As a tentative explanation, CK2 also inhibits the DNA-dependent protein kinase subunit DNA-PKcs in glioblastomas (15). DNA-PK is itself a key inhibitor of HR (41), and gliomas might thus escape CK2 inhibition-induced NHEJ inhibition via an increase in homologous recombination. In agreement with this hypothesis, apigenin treatment of glioma cells increased the radiation-induced phosphorylation of the DNA-PK target Chk2 (34,42) in our experiments.

In colon carcinoma cells and fibroblasts, although CK2 inhibition does not alter the rejoining of DSB, it does slow down the dephosphorylation of γ-H2Ax and its dissociation from the DNA after repair (40). Such a lengthened γ-H2Ax decay is believed to amplify checkpoint signaling in the presence of minimal residual DNA damage and lead to cell death (43). We did not observe this phenomenon in our experiments.

In our experiments and in agreement with previous reports (44,45), CK2 inhibition also reduced the constitutive level of NF-κB reporter activity in both cell lines. The post-radiation transcriptional activity of this factor also remained significantly lower in apigenin-treated irradiated cells than in the control, non-irradiated cells. Apigenin-treated cells, however, still responded to irradiation with a minimal induction of NF-κB (data not shown), and our results thus do not contradict the paradigm that CK2 triggers NF-κB activation in response to UV-induced DNA damage but not following exposure to ionizing radiation (4649).

Pharmacological NF-κB inhibitors are nonetheless known to modulate the fate of tumor cells following irradiation. They were reported to radiosensitize glioblastomas (50,51), but NF-κB was also, on the contrary, recently reported to mediate apoptosis following the irradiation of primary cultures and progenitor cells of gliomas lines (52). In line with these contrasting reports and its favorable effect on DSB-repair in our experiments, CK2 inhibition did not radiosensitize our glioma cells. This neutrality seems to be independent of TP53 mutational status, as we confirmed by exon sequencing that LN18 and U87 cells express, respectively mutant and wild-type variants of this CK2 target (data not shown) (5355). Aspecific effects of apigenin were also ruled out by repeating clonogenic assays following siRNA mediated depletion of CK2-α.

Although we cannot rule out that CK2 inhibition could radio-sensitize glioblastoma cells with defective DNA-PK, the lack of radiosensitization of gliomas that we have observed contrasts with that of non-small cell lung carcinomas, fibroblasts and colon carcinomas cells following CK2 inhibition (6,40). Since DNA-PK mutations occur in only 3% of glioblastomas (TCGA data portal, accessed January 16th, 2011; the TCGA research network) (56) we believe that glioma patients should not be included in clinical trials that assess the radiosensitizing role of CK2 inhibitors. Further studies of DNA repair mechanisms in primary brain tumors and a preclinical evaluation of therapies combining CK2 inhibitor with other DNA-damaging agents with DNA-PK inhibitors are required to improve the therapeutic options for these tumors.

In spite of its modulation of DNA-damage signaling cascades, CK2 inhibition fails to inhibit DNA repair following ionizing radiation and to radiosensitize glioma cells, independently of their TP53 status. This contrasts with other tumor types, urging caution regarding the inclusion of malignant glioma patients in clinical studies that will assess the radiosensitizing role of CK2 inhibitors in solid cancers.

Acknowledgements

We wish to thank Dr Sandra Ormenese for her help with the flow cytometry, as well as Mr. Olivier Pierard and Ms. Catherine Waltener for their expert technical assistance. P.A.R. is a research associate at the Belgian Fund for Scientific Research. This study was supported by grant PNC 29-006 of the Belgian Ministry of Health to V.B. and P.A.R., grant 1.5.162.10 of the National Research Fund of Belgium to P.A.R., a grant from the Belgian Foundation against Cancer to V.B. and P.A.R., and a grant from the Centre Anticancéreux of the University of Liège to V.B.

References

1 

Stupp R, Mason WP, van den Bent MJ, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Robe PA, Bentires-Alj M, Bonif M, et al: In vitro and in vivo activity of the nuclear factor-kappaB inhibitor sulfasalazine in human glioblastomas. Clin Cancer Res. 10:5595–5603. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Bredel M, Scholtens DM, Yadav AK, et al: NFKBIA deletion in glioblastomas. N Engl J Med. 364:627–637. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Squatrito M and Holland EC: DNA damage response and growth factor signaling pathways in gliomagenesis and therapeutic resistance. Cancer Res. 71:5945–5949. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Becherel OJ, Jakob B, Cherry AL, et al: CK2 phosphorylation-dependent interaction between aprataxin and MDC1 in the DNA damage response. Nucleic Acids Res. 38:1489–1503. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Lin YC, Hung MS, Lin CK, et al: CK2 inhibitors enhance the radiosensitivity of human non-small cell lung cancer cells through inhibition of stat3 activation. Cancer Biother Radiopharm. 26:381–388. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Brown MS, Diallo OT, Hu M, et al: CK2 modulation of NF-κB, TP53, and the malignant phenotype in head and neck cancer by anti-CK2 oligonucleotides in vitro or in vivo via Sub-50-nm nanocapsules. Clin Cancer Res. 16:2295–2307. 2010.

8 

Eddy SF, Guo S, Demicco EG, et al: Inducible IkappaB kinase/IkappaB kinase epsilon expression is induced by CK2 and promotes aberrant nuclear factor-kappaB activation in breast cancer cells. Cancer Res. 65:11375–11383. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Yu M, Yeh J and Van Waes C: Protein kinase casein kinase 2 mediates inhibitor-kappaB kinase and aberrant nuclear factor-kappaB activation by serum factor(s) in head and neck squamous carcinoma cells. Cancer Res. 66:6722–6731. 2006. View Article : Google Scholar

10 

Siepmann M, Kumar S, Mayer G and Walter J: Casein kinase 2 dependent phosphorylation of neprilysin regulates receptor tyrosine kinase signaling to Akt. PLoS One. 5:E131342010. View Article : Google Scholar : PubMed/NCBI

11 

Horejsí Z, Takai H, Adelman CA, et al: CK2 phospho-dependent binding of R2TP complex to TEL2 is essential for mTOR and SMG1 stability. Mol Cell. 39:839–850. 2010.PubMed/NCBI

12 

Maccario H, Perera NM, Davidson L, Downes CP and Leslie NR: PTEN is destabilized by phosphorylation on Thr366. Biochem J. 405:439–444. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Kang H, Jung JW, Kim MK and Chung JH: CK2 is the regulator of SIRT1 substrate-binding affinity, deacetylase activity and cellular response to DNA-damage. PLoS One. 4:E66112009. View Article : Google Scholar : PubMed/NCBI

14 

Takeishi Y, Ohashi E, Ogawa K, Masai H, Obuse C and Tsurimoto T: Casein kinase 2-dependent phosphorylation of human Rad9 mediates the interaction between human Rad9-Hus1-Rad1 complex and TopBP1. Genes Cells. 15:761–771. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Olsen BB, Issinger OG and Guerra B: Regulation of DNA-dependent protein kinase by protein kinase CK2 in human glioblastoma cells. Oncogene. 29:6016–6026. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Ström CE, Mortusewicz O, Finch D, et al: CK2 phosphorylation of XRCC1 facilitates dissociation from DNA and single-strand break formation during base excision repair. DNA Repair. 10:961–969. 2011.PubMed/NCBI

17 

Kaminska B, Ellert-Miklaszewska A, Oberbek A, et al: Efficacy and mechanism of anti-tumor action of new potential CK2 inhibitors toward glioblastoma cells. Int J Oncol. 35:1091–1100. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Prudent R, Moucadel V, Nguyen CH, et al: Antitumor activity of pyridocarbazole and benzopyridoindole derivatives that inhibit protein kinase CK2. Cancer Res. 70:9865–9874. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Pierre F, Chua PC, O’Brien SE, et al: Pre-clinical characterization of CX-4945, a potent and selective small molecule inhibitor of CK2 for the treatment of cancer. Mol Cell Biochem. 356:37–43. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Battistutta R, Cozza G, Pierre F, et al: Unprecedented selectivity and structural determinants of a new class of protein kinase CK2 inhibitors in clinical trials for the treatment of cancer. Biochemistry. 50:8478–8488. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Schneider CC, Hessenauer A, Montenarh M and Götz C: p53 is dispensable for the induction of apoptosis after inhibition of protein kinase CK2. Prostate. 70:126–134. 2010.PubMed/NCBI

22 

Shehata M, Schnabl S, Demirtas D, et al: Reconstitution of PTEN activity by CK2 inhibitors and interference with the PI3-K/Akt cascade counteract the antiapoptotic effect of human stromal cells in chronic lymphocytic leukemia. Blood. 116:2513–2521. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Li C, Liu X, Lin X and Chen X: Structure-activity relationship of 7 flavonoids on recombinant human protein kinase CK2 holoenzyme. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 34:20–26. 2009.PubMed/NCBI

24 

Sarno S, de Moliner E, Ruzzene M, et al: Biochemical and three-dimensional-structural study of the specific inhibition of protein kinase CK2 by [5-oxo-5,6-dihydroindolo-(1,2-a)quinazolin-7-yl] acetic acid (IQA). Biochem J. 374:639–646. 2003.PubMed/NCBI

25 

Zhao M, Ma J, Zhu HY, et al: Apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through targeting the trinity of CK2, Cdc37 and Hsp90. Mol Cancer. 10:1042011. View Article : Google Scholar : PubMed/NCBI

26 

Zhong Y, Krisanapun C, Lee SH, et al: Molecular targets of apigenin in colorectal cancer cells: involvement of p21, NAG-1 and p53. Eur J Cancer. 46:3365–3374. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Slusarz A, Shenouda NS, Sakla MS, et al: Common botanical compounds inhibit the hedgehog signaling pathway in prostate cancer. Cancer Res. 70:3382–3390. 2010. View Article : Google Scholar

28 

Das A, Banik NL and Ray SK: Flavonoids activated caspases for apoptosis in human glioblastoma T98G and U87MG cells but not in human normal astrocytes. Cancer. 116:164–176. 2010.PubMed/NCBI

29 

Chakravarti A, Zhai GG, Zhang M, et al: Survivin enhances radiation resistance in primary human glioblastoma cells via caspase-independent mechanisms. Oncogene. 23:7494–7506. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Miyamoto S: Nuclear initiated NF-κB signaling: NEMO and ATM take center stage. Cell Res. 21:116–130. 2010.

31 

Keller DM, Zeng X, Wang Y, et al: A DNA damage-induced p53 serine 392 kinase complex contains CK2, hSpt16, and SSRP1. Mol Cell. 7:283–292. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Tsuchiya Y, Asano T, Nakayama K, Kato T Jr, Karin M and Kamata H: Nuclear IKKbeta is an adaptor protein for IkappaBalpha ubiquitination and degradation in UV-induced NF-kappaB activation. Mol Cell. 39:570–582. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Cheung WL, Turner FB, Krishnamoorthy T, et al: Phosphorylation of histone H4 serine 1 during DNA damage requires casein kinase II in S. cerevisiae. Curr Biol. 15:656–660. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Li J and Stern DF: DNA damage regulates Chk2 association with chromatin. J Biol Chem. 280:37948–37956. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Agullo G, Gamet-Payrastre L, Manenti S, et al: Relationship between flavonoid structure and inhibition of phosphatidylinositol 3-kinase: a comparison with tyrosine kinase and protein kinase C inhibition. Biochem Pharmacol. 53:1649–1657. 1997. View Article : Google Scholar : PubMed/NCBI

36 

Koch CA, Agyei R, Galicia S, et al: Xrcc4 physically links DNA end processing by polynucleotide kinase to DNA ligation by DNA ligase IV. EMBO J. 23:3874–3885. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Quiros S, Roos WP and Kaina B: Rad51 and BRCA2 - new molecular targets for sensitizing glioma cells to alkylating anti-cancer drugs. PLoS One. 6:E271832011. View Article : Google Scholar : PubMed/NCBI

38 

Macrae CJ, McCulloch RD, Ylanko J, Durocher D and Koch CA: APLF (C2orf13) facilitates nonhomologous end-joining and undergoes ATM-dependent hyperphosphorylation following ionizing radiation. DNA Repair. 7:292–302. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Iles N, Rulten S, El-Khamisy SF and Caldecott KW: APLF (C2orf13) is a novel human protein involved in the cellular response to chromosomal DNA strand breaks. Mol Cell Biol. 27:3793–3803. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Zwicker F, Ebert M, Huber PE, Debus J and Weber KJ: A specific inhibitor of protein kinase CK2 delays gamma-H2Ax foci removal and reduces clonogenic survival of irradiated mammalian cells. Radiat Oncol. 6:152011. View Article : Google Scholar

41 

Neal JA and Meek K: Choosing the right path: does DNA-PK help make the decision? Mutat Res. 711:73–86. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Hill R and Lee PW: The DNA-dependent protein kinase (DNA-PK): more than just a case of making ends meet? Cell Cycle. 9:3460–3469. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Kinner A, Wu W, Staudt C and Iliakis G: Gamma-H2AX in recognition and signaling of DNA double-strand breaks in the context of chromatin. Nucleic Acids Res. 36:5678–5694. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Shen J, Channavajhala P, Seldin DC and Sonenshein GE: Phosphorylation by the protein kinase CK2 promotes calpain-mediated degradation of IkappaBalpha. J Immunol. 167:4919–4925. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Viatour P, Merville MP, Bours V and Chariot A: Phosphorylation of NF-κB and IκB proteins: implications in cancer and inflammation. Trends Biochem Sci. 30:43–52. 2005.

46 

Janssens S and Tschopp J: Signals from within: the DNA-damage-induced NF-κB response. Cell Death Differ. 13:773–784. 2006.

47 

Kato T, Delhase M, Hoffmann A and Karin M: CK2 is a C-terminal IkappaB kinase responsible for NF-kappaB activation during the UV response. Mol Cell. 12:829–839. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Li N: ATM is required for Ikappa B kinase (IKK) activation in response to DNA double strand breaks. J Biol Chem. 276:8898–8903. 2000. View Article : Google Scholar

49 

Veuger SJ, Hunter JE and Durkacz BW: Ionizing radiation-induced NF-κB activation requires PARP-1 function to confer radioresistance. Oncogene. 28:832–842. 2008.

50 

Hunter JE, Willmore E, Irving JAE, Hostomsky Z, Veuger SJ and Durkacz BW: NF-κB mediates radio-sensitization by the PARP-1 inhibitor, AG-014699. Oncogene. 31:251–264. 2012.

51 

Tsuboi Y, Kurimoto M, Nagai S, et al: Induction of autophagic cell death and radiosensitization by the pharmacological inhibition of nuclear factor-kappa B activation in human glioma cell lines. J Neurosurg. 110:594–604. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Berger R, Jennewein C, Marschall V, et al: NF-κB is required for Smac mimetic-mediated sensitization of glioblastoma cells for γ-irradiation-induced apoptosis. Mol Cancer Ther. 10:1867–1875. 2011.

53 

Asai A, Miyagi Y, Sugiyama A, et al: Negative effects of wild-type p53 and s-Myc on cellular growth and tumorigenicity of glioma cells. Implication of the tumor suppressor genes for gene therapy. J Neurooncol. 19:259–268. 1994. View Article : Google Scholar : PubMed/NCBI

54 

Wischhusen J, Naumann U, Ohgaki H, Rastinejad F and Weller M: CP-31398, a novel p53-stabilizing agent, induces p53-dependent and p53-independent glioma cell death. Oncogene. 22:8233–8245. 2003. View Article : Google Scholar : PubMed/NCBI

55 

Meek DW and Cox M: Induction and activation of the p53 pathway: a role for the protein kinase CK2? Mol Cell Biochem. 356:133–138. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Cancer Genome Atlas Research Network: Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 455:1061–1068. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August 2012
Volume 41 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kroonen J, Artesi M, Capraro V, Nguyen-Khac M, Willems M, Chakravarti A, Bours V and Robe PA: Casein kinase 2 inhibition modulates the DNA damage response but fails to radiosensitize malignant glioma cells. Int J Oncol 41: 776-782, 2012
APA
Kroonen, J., Artesi, M., Capraro, V., Nguyen-Khac, M., Willems, M., Chakravarti, A. ... Robe, P.A. (2012). Casein kinase 2 inhibition modulates the DNA damage response but fails to radiosensitize malignant glioma cells. International Journal of Oncology, 41, 776-782. https://doi.org/10.3892/ijo.2012.1489
MLA
Kroonen, J., Artesi, M., Capraro, V., Nguyen-Khac, M., Willems, M., Chakravarti, A., Bours, V., Robe, P. A."Casein kinase 2 inhibition modulates the DNA damage response but fails to radiosensitize malignant glioma cells". International Journal of Oncology 41.2 (2012): 776-782.
Chicago
Kroonen, J., Artesi, M., Capraro, V., Nguyen-Khac, M., Willems, M., Chakravarti, A., Bours, V., Robe, P. A."Casein kinase 2 inhibition modulates the DNA damage response but fails to radiosensitize malignant glioma cells". International Journal of Oncology 41, no. 2 (2012): 776-782. https://doi.org/10.3892/ijo.2012.1489