2′-C-Cyano-2′-deoxy-1-β-d-arabino-pentofuranosylcytosine (CNDAC), the prodrug (sapacitabine) of which is in clinical trials, has the novel mechanism of action of causing single-strand breaks after incorporating into DNA. Cells respond to this unique lesion by activating the G2 checkpoint, affected by the Chk1-Cdc25C-cyclin-dependent kinase 1/cyclin B pathway. This study aims at defining DNA damage checkpoint sensors that activate this response to CNDAC, particularly focusing on the major phosphatidylinositol 3-kinase–like protein kinase family proteins. First, fibroblasts, deficient in ataxia-telangiectasia mutated (ATM), transfected with empty vector or repleted with ATM, were arrested in G2 by CNDAC to similar extents, suggesting ATM is not required to activate the G2 checkpoint. Second, chromatin associations of RPA70 and RPA32, subunits of the ssDNA-binding protein, and the ataxia-telangiectasia and Rad3-related (ATR) substrate Rad17 and its phosphorylated form were increased on CNDAC exposure, suggesting activation of ATR kinase. The G2 checkpoint was abrogated due to depletion of ATR by small interfering RNA, and impaired in ATR-Seckel cells, indicating participation of ATR in this G2 checkpoint pathway. Third, the G2 checkpoint was more stringent in glioma cells with wild-type DNA-dependent protein kinase catalytic subunit (DNA-PKcs) than those with mutant DNA-PKcs, as shown by mitotic index counting. CNDAC-induced G2 arrest was abrogated by specific DNA-PKcs inhibitors or small interfering RNA knockdown in ML-1 and/or HeLa cells. Finally, two phosphatidylinositol 3-kinase–like protein kinase inhibitors, caffeine and wortmannin, abolished the CNDAC-induced G2 checkpoint in a spectrum of cell lines. Together, our data showed that ATR and DNA-PK cooperate in CNDAC-induced activation of the G2 checkpoint pathway. [Mol Cancer Ther 2008;7(1):133–42]

Because cells are threatened by endogenous or exogenous genotoxic stresses, they have evolved delicate protective machineries to maintain the genomic integrity. In response to environmental perturbations/toxicants such as ionizing radiation (IR), UV light, and DNA-damaging agents, cells delay cell cycle progression by initiating cell cycle checkpoints to allow time for repair of damaged DNA. According to current models, the DNA damage checkpoints conceptually have four functional components: sensors, mediators, transducers, and effectors (1). Sensors of DNA damage, typically ataxia-telangiectasia mutated (ATM), ataxia-telangiectasia and Rad3-related (ATR), and DNA-dependent protein kinase (DNA-PK), which belong to the phosphatidylinositol 3-kinase–like protein kinase (PIKK) family and share homology of the phosphatidylinositol 3-kinase domain harboring the catalytic site of the active protein kinases (2), detect various insults to the genome and initiate signal transduction cascades to affect the S-phase and G2 checkpoint pathways (3).

Although some functional redundancy exists among the PIKK sensors, the detection of different types of DNA damage resulting from specific forms of genotoxic stress is generally addressed by unique sensors. For example, ATM is the major sensor responding to the double-strand breaks (DSB) caused by IR (2, 4). Mediated by the Mre11-Rad50-Nbs1 complex (57), ATM is rapidly activated through intermolecular autophosphorylation and subsequent dimer dissociation (8). The activated ATM phosphorylates the DNA damage checkpoint kinase Chk2, which transduces signals downstream. In contrast, ATR plays a more crucial role when cells are exposed to UV radiation, stalled replication forks, or chemicals that make bulky base adducts (1, 2). ssDNA generated by these events becomes coated with replication protein A (RPA), which attracts the ATR-interacting protein (ATRIP). ATR associates with this complex and initiates cell cycle checkpoints via phosphorylation of another DNA damage checkpoint kinase, Chk1. Recent studies from different laboratories provided evidence for ATM activity upstream of ATR recruitment to IR-damaged chromatin (911), revealing cross-talk between these two important sensors.

DNA-PK is another PIKK family member known to be a nuclear serine/threonine kinase and a molecular sensor in the DNA damage response. Composed of a catalytic subunit (DNA-PKcs) and two regulatory subunits (Ku70 and Ku80; ref. 12), DNA-PK is one of the core components of mammalian repair of DSBs through the nonhomologous end-joining pathway. It has been proposed to sense DNA damage and enhance signal transduction via phosphorylation of downstream targets, including H2AX and p53 (13). However, a role for DNA-PK in the activation of transducers of the DNA damage signaling pathway in cellular responses to genotoxic stress has not been established.

Designed on a basis of a DNA self-strand–breaking mechanism, 2′-C-cyano-2′-deoxy-1-β-d-arabino-pentofuranosylcytosine (CNDAC) is incorporated within the DNA of proliferating cells and induces a single-strand break (SSB) by a β-elimination process (14, 15). The DNA nick results from the rearrangement of the CNDAC molecule to form 2′-C-cyano-2′,3′-didehydro-2′,3′-dideoxycytidine (CNddC), which is a de facto DNA chain terminator (16). Unlike the nucleoside analogues 1-β-d-arabinofuranosylcytosine, gemcitabine, and fludarabine that cause S-phase arrest, cells respond to this damage by activating the G2 checkpoint (17). Although CNDAC-induced cell cycle arrest is instituted through activation of the Chk1-Cdc25C-cyclin-dependent kinase 1 (Cdk1)/cyclin B checkpoint pathway, participants in the signaling events upstream of Chk1 kinase remain obscure.

The aim of this study was to elucidate the sensor proteins that detect CNDAC-induced DNA damage and activate the G2 checkpoint but not to focus on cell survival mechanisms. Strategies taking advantage of genetically defined cell lines and inhibition of PIKK family proteins by small-molecule inhibitors or RNA interfering technology were used to determine the apical molecules in the cellular responses to DNA damage generated after CNDAC incorporation. Our results show that both ATR and DNA-PK participate in activation of the G2 checkpoint but that ATM is not essential for this response.

Human Cell Culture

Acute myelogenous leukemia cell line ML-1 (gifted by Dr. Michael B. Kastan, St. Jude Children's Research Hospital, Memphis, TN) was maintained in exponential growth phase in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum at 37°C in a humidified atmosphere with 5% CO2. AT22IJE-T (AT-C), a fibroblast cell line derived from an ataxia-telangiectasia patient, and lines stably transfected with either an episomal expression vector (AT22IJE-TpEBS7, AT-V) or full-length ATM cDNA (AT22IJE-TpEBS7-YZ5, AT-AT) were gifts from Dr. Yosef Shiloh (Tel Aviv University, Tel Aviv, Israel; ref. 18) and were cultured in DMEM with high glucose and 20% fetal bovine serum. Glioma-derived cell lines M059-K (wild-type) and M059-J (DNA-PKcs deficient) were obtained from Dr. M.J. Allalunis-Turner (Brookhaven National Laboratory, Upton, NY; refs. 19, 20) and grown in α-MEM supplemented with 20% fetal bovine serum. Colon carcinoma cell line HCT116 (gifted by Dr. Minoru Koi, National Institute of Environmental Health Sciences, Research Triangle Park, NC) was maintained in DMEM with high glucose and 10% fetal bovine serum. Cervical cancer cell line HeLa CCL2 was purchased from the American Type Culture Collection and cultured in MEM with nonessential amino acids, sodium pyruvate, and 10% fetal bovine serum. EBV-transformed lymphoblastoid cell lines, wild-type control line GM02188, and Seckel syndrome lines GM09703 and GM18367A were from Coriell Cell Repositories and were grown in RPMI 1640 supplemented with 20% heat-inactivated fetal bovine serum. All cells were free of Mycoplasma, as determined by an ELISA kit (Life Technologies MycoTest kit).

Chemicals and Antibodies

CNDAC was synthesized as described (21). A stock solution (15–25 mmol/L) was prepared in PBS (pH 6.5), sterilized by filtration, stored at −20°C, and diluted in sterile PBS just before use. Camptothecin and PIKK inhibitor wortmannin were obtained from Sigma-Aldrich, Inc., and DNA-PK inhibitors NU7026 and IC86621 were obtained from Calbiochem/EMD Biosciences and stored as 10 mmol/L in DMSO aliquots at −70°C. Caffeine was also from Sigma-Aldrich and prepared freshly to 100 mmol/L in warm water and filtered before use. All other chemicals were reagent grade.

Sources of antibodies are as follows: rabbit antibodies against phosphorylated Ser317 (polyclonal) and Ser345 (monoclonal) of Chk1 and lamin A/C and rabbit polyclonal antibodies against phosphorylated Thr68 of Chk2 and phosphorylated Ser645 of Rad17 (Cell Signaling Technology); mouse monoclonal antibodies to Chk1 and RhoA and rabbit polyclonal antibodies to Chk1, H2A, and Rad17 (Santa Cruz Biotechnology); rabbit polyclonal antibody to ATM (Novus Biologicals); mouse anti-DNA-PKcs (Lab Vision Corp.); mouse monoclonal antibody to phosphorylated Ser139 of H2AX (γ-H2AX) and rabbit polyclonal antibodies to ATRIP (Upstate Biotechnology); mouse anti-RPA70 (Oncogene Research Products/EMD Biosciences); rabbit anti-RPA32 and mouse anti-ATR (GeneTex, Inc.); mouse anti-Orc2 (BD PharMingen International); mouse monoclonal antibody to β-actin and phosphorylated Ser10 of histone H3 (Sigma-Aldrich); rabbit anti-XRCC4 (AbD Serotec); anti-mouse or anti-rabbit IgG horseradish peroxidase–conjugated antibody (Amersham Biosciences); and FITC-conjugated goat anti-mouse IgG (Jackson ImmunoResearch Laboratories, Inc.).

Cell Cycle Arrest and Analysis

The CNDAC and camptothecin concentrations were chosen based on dose-finding studies for the maximal G2 arrest after drug exposure for one normal population doubling time. Cells were washed with ice-cold PBS (pH 7.4) and fixed in 70% ethanol. Fixed cells were washed with PBS before incubation with 25 μg/mL of propidium iodide (Sigma-Aldrich) and 2.5 μg/mL of DNase-free RNase A (Roche). Fluorescence was measured on a Becton Dickinson FACSCalibur flow cytometer. At least 20,000 cells were measured for each sample.

Immunofluorescent Detection of Phosphorylated Histone H3

Cells were immunostained for phosphorylated Ser10 of histone H3, nuclear stained with propidium iodide, and subjected to detection by flow cytometry, as described previously (17).

Quantification of Mitotic Index

After centrifugation of cells to slides by Cytospin (Thermo Electron Corp.), cells were fixed with 4% paraformaldehyde in PBS (pH 7.4), stained with 4′,6-diamidino-2-phenylindole (2 μg/mL in PBS), and mounted in Vectashield mounting medium (Vector Laboratories, Inc.). Mitotic morphology was identified by the appearance of nuclear DNA condensation under an epifluorescence microscope (Nikon). At least 400 randomly selected cells on each slide were counted.

Immunoblotting of Whole Cell Lysates and Subcellular Fractions

Cell lysates were subjected to isolation by SDS-PAGE and immunoblotting, as described previously (17). Subcellular fractionation was done according to the method of Zou et al. (22) and Wang et al. (23). Fractions (S1, cytoplasmic; S2, nuclear soluble; P2, chromatin bound) were loaded on 4% to 12% Bis-Tris gradient gels (Bio-Rad Laboratories) or 10% SDS-polyacrylamide gels. Immunoblots were quantitated by Li-Cor Odyssey software in indicated experiments.

Depletion of ATR or DNA-PKcs by Small Interfering RNA

Small interfering RNA (siRNA) transfections were done with Opti-MEM I (Invitrogen Corp.) containing Lipofectamine 2000 (Invitrogen) or DharmaFECT I (Dharmacon Research, Inc.) and 100 nmol/L final concentrations of oligos, following experimental optimization of the manufacturers' instructions. siRNA duplexes of 21-nucleotide RNAs with a 2-nucleotide 3′-overhang were purchased from Dharmacon Research. The siRNA target sequences of ATR (24) are 5′-AACCTCCGTGATGTTGCTTGA-3′ (ATR-2 duplex). Lamin A/C (25), whose target sequences are 5′-AACTGGACTTCCAGAAGAACA-3′, and siCONTROL RISC-free siRNA were used as positive and negative control siRNAs, respectively. The siRNAs directed against DNA-PKcs were a SmartPool of four duplexes (PRKDC). Transfection mixtures were removed after 6 h and cells were incubated with complete medium for additional 18 or 42 h before introduction of CNDAC. Cells were harvested 24 h after drug treatment and subjected to flow cytometric analysis.

Confocal Microscopy of Immunofluorescent γ-H2AX

Drug-treated adherent cells were immunostained for γ-H2AX as described previously (17). γ-H2AX and nuclear staining were viewed with a FV500 laser scanning confocal microscope (Olympus) using a 63× objective. The projections of multiple slices were saved as TIF files.

Statistical Methods

General linear model was used to compare the mean difference of mitotic indices between or among three cell lines and different interventions. All statistical tests are two sided, and P < 0.05 was considered statistically significant. Statistical analysis was done using Statistical Analysis System Release 9.1 (SAS Institute). Comparison of mitotic indices of different treatments in individual cell lines was conducted by Student's t test.

Activation of CNDAC-Induced G2 Checkpoint Is Independent of ATM

The first approach to define the role of PIKKs in G2 checkpoint activation by CNDAC was to test if cells that lack ATM function would arrest in G2 in response to CNDAC, as do wild-type cells. Treatment of human fibroblast lines that were either lacking ATM function (AT-C), transfected with an empty vector (AT-V), or repleted for ATM (AT-AT) with 4 to 8 μmol/L CNDAC for 24 h increased the G2 population equally in all lines (Fig. 1A). Second, the stringency of the G2 checkpoint activated by CNDAC was compared between ATM-deficient and ATM-repleted cells by scoring cells that had escaped from G2 and were trapped by nocodazole in M phase. Mitotic indices of both lines in response to a series of concentrations of CNDAC were comparable, suggesting similar stringency of the G2 checkpoint (Fig. 1B). Third, the short-term effect of CNDAC on the G2-M checkpoint was compared with that of IR. Consistent with previous findings (26), cells deficient in ATM function failed to arrest progression into mitosis within 1 to 4 h after irradiation to the same extent as wild-type cells (Fig. 1C). When AT-AT fibroblasts were exposed to 10 Gy γ-irradiation, the mitotic ratio (percentage of total cells in mitosis, determined by positive staining for histone H3 phosphorylation, relative to that of untreated cells) dropped from 100% to 20% at 1 h and decreased further to 6% at 2 h and maintained this low level by 4 h (Supplementary Fig. S1).3

3

Supplementary materials for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

In contrast, when incubated with CNDAC for up to 4 h, the mitotic ratios of both lines reduced at similar rates to ∼60% by 4 h (Supplementary Fig. S1);3 therefore, the relative mitotic ratio of AT-AT to AT-C remained constant (0.98–1.08; Fig. 1C). The difference in short-term response of cells to these two agents reflects their distinct mechanisms of action in that IR has an immediate effect on cells in all phases of the cell cycle, whereas incorporation of CNDAC triphosphate during S phase is required for subsequent effects of the drug. Finally, changes in biochemical markers of checkpoint activation and DNA damage were also compared among these isogenic paired lines. Chk1 kinase became phosphorylated on Ser345 in all lines but to a greater level in ATM-complemented cells (Fig. 1D). The CNDAC-induced increases in the ratios of phosphorylated Chk2/total Chk2 were similar in the three lines, suggesting that this response is independent of ATM. Likely, this response was secondary to CNDAC-induced DNA damage. Another ATM substrate, p53, was phosphorylated on Ser15 in response to CNDAC to similar extents in all three lines (Fig. 1D). Phosphorylation of p53 on Ser20 and Ser37 was also similarly positive in all lines (data not shown). In addition to immunoblotting, immunostaining revealed augmentation of H2AX phosphorylation on Ser139 in all lines, as manifested by nuclear foci, which was more remarkable in ATM-repleted cells (Supplementary Fig. S2A).3 Together, the data suggest that ATM is not required to activate the G2 checkpoint pathway in response to CNDAC.

Figure 1.

ATM is dispensable for activation of the G2 checkpoint by CNDAC. A, exponentially growing AT cell lines (AT-C, ATM mutant; AT-V, AT-C stably transfected with an empty vector; AT-AT, AT-C repleted with full-length ATM) were treated with various concentrations of CNDAC for 24 h. Cell cycle distribution was determined by flow cytometry. B, stringency of the G2 checkpoint activated by CNDAC is similar in ATM-complemented (black columns) and ATM-mutant (white columns) cells. Both lines were exposed to CNDAC (0.5, 2, and 8 μmol/L, respectively) for 24 h. Nocodazole (400 ng/mL) was introduced and coincubated for additional 24 h. A minimum of 500 nuclei was counted for each mitotic index measurement. C, short exposure (1–4 h) of AT-C and AT-AT cells to CNDAC (8 μmol/L) or IR (10 Gy) displayed differential mitotic ratios. Percentage of total cells of treatment in mitosis, staining positive for histone H3 phosphorylation, relative to that of control cells was expressed as mitotic ratio (MR). Mitotic ratio of AT-AT cells relative to mitotic ratio of AT-C cells was plotted as a function of time after treatment. D, AT-C and AT-AT cells were exposed to 8 μmol/L CNDAC for 24 h. Whole-cell lysates were fractionated by SDS-PAGE and subjected to immunoblot analysis with indicated antibodies.

Figure 1.

ATM is dispensable for activation of the G2 checkpoint by CNDAC. A, exponentially growing AT cell lines (AT-C, ATM mutant; AT-V, AT-C stably transfected with an empty vector; AT-AT, AT-C repleted with full-length ATM) were treated with various concentrations of CNDAC for 24 h. Cell cycle distribution was determined by flow cytometry. B, stringency of the G2 checkpoint activated by CNDAC is similar in ATM-complemented (black columns) and ATM-mutant (white columns) cells. Both lines were exposed to CNDAC (0.5, 2, and 8 μmol/L, respectively) for 24 h. Nocodazole (400 ng/mL) was introduced and coincubated for additional 24 h. A minimum of 500 nuclei was counted for each mitotic index measurement. C, short exposure (1–4 h) of AT-C and AT-AT cells to CNDAC (8 μmol/L) or IR (10 Gy) displayed differential mitotic ratios. Percentage of total cells of treatment in mitosis, staining positive for histone H3 phosphorylation, relative to that of control cells was expressed as mitotic ratio (MR). Mitotic ratio of AT-AT cells relative to mitotic ratio of AT-C cells was plotted as a function of time after treatment. D, AT-C and AT-AT cells were exposed to 8 μmol/L CNDAC for 24 h. Whole-cell lysates were fractionated by SDS-PAGE and subjected to immunoblot analysis with indicated antibodies.

Close modal

ATR Is Required for Optimal G2 Checkpoint Activation by CNDAC

To determine the role of ATR in activation of the CNDAC-induced G2 checkpoint, factors that are required for ATR recruitment and activation were investigated. One such protein, RPA, a ubiquitous ssDNA-binding protein, consisting of three subunits of 70, 32, and 14 kDa, associates with DNA at sites of damage (2729). ATRIP, the partner of ATR, is attracted to RPA-ssDNA complexes, thereby initiating ATR-dependent DNA damage sensing processes (30, 31). These factors were evaluated in HeLa cells, which were arrested in G2 after a 24-h incubation with 4 μmol/L CNDAC. Although no changes in cellular RPA protein level were detectable in whole-cell lysates after CNDAC treatment, densitometric analysis indicated that chromatin-associated RPA70 and RPA32 increased about 2- and 5-fold, respectively, in response to CNDAC (Fig. 2A). In similar experiments with HCT116 cells, analysis of RPA70 and RPA32 during 3 days of exposure to 3 μmol/L CNDAC showed that, as with HeLa cells, both proteins increased their association with chromatin during checkpoint activation. Chromatin-bound Rad17, an ATR substrate, also accumulated during CNDAC treatment, as did its phosphorylated form (Fig. 2B), suggesting activation of ATR kinase in response to CNDAC.

Figure 2.

ATR is also responsible for activation of the G2 checkpoint by CNDAC. A and B, chromatin association of RPA increases in response to CNDAC. Exponentially growing HeLa cells (AC) were incubated with 4 μmol/L CNDAC for 24 h. HCT116 cells were exposed to 3 μmol/L CNDAC continuously for 3 d. Cells were harvested at indicated times and subjected to subcellular fractionation. Fractions: cytoplasmic (S1), nuclear soluble (S2), and chromatin binding (P2). RPA70, RPA32, Orc2, and RhoA in the indicated fractions were detected by immunoblotting. Results are representative of several experiments. B, HeLa cells were treated and fractionated as in A. ATR, ATRIP, and phosphorylated and total Rad17 in the indicated fractions were detected by immunoblotting. Asterisk, nonspecific binding to the antibody. C, ATR level was knocked down by siRNA in HeLa cells and 8 μmol/L CNDAC was added 48 h after transfection. Cells were incubated for an additional 24 h and analyzed by immunoblotting with indicated antibodies and by flow cytometry. In the cell cycle histograms, percentages indicate portions with a G2-M DNA content. D, ATR-Seckel cells show defects in the G2 checkpoint activated by CNDAC. Cells with wild-type (GM02188) and deficient in ATR (GM09703 and GM18367A) were exposed to CNDAC (40 and 60 μmol/L) for 38 h. Colcemid (100 ng/mL) was added and coincubated for additional 15 h. A minimum of 400 nuclei was counted for each mitotic index measurement. Columns, mean of three separate determinations in two similar experiments; bars, SD. L.E., longer exposure of the immunoblot.

Figure 2.

ATR is also responsible for activation of the G2 checkpoint by CNDAC. A and B, chromatin association of RPA increases in response to CNDAC. Exponentially growing HeLa cells (AC) were incubated with 4 μmol/L CNDAC for 24 h. HCT116 cells were exposed to 3 μmol/L CNDAC continuously for 3 d. Cells were harvested at indicated times and subjected to subcellular fractionation. Fractions: cytoplasmic (S1), nuclear soluble (S2), and chromatin binding (P2). RPA70, RPA32, Orc2, and RhoA in the indicated fractions were detected by immunoblotting. Results are representative of several experiments. B, HeLa cells were treated and fractionated as in A. ATR, ATRIP, and phosphorylated and total Rad17 in the indicated fractions were detected by immunoblotting. Asterisk, nonspecific binding to the antibody. C, ATR level was knocked down by siRNA in HeLa cells and 8 μmol/L CNDAC was added 48 h after transfection. Cells were incubated for an additional 24 h and analyzed by immunoblotting with indicated antibodies and by flow cytometry. In the cell cycle histograms, percentages indicate portions with a G2-M DNA content. D, ATR-Seckel cells show defects in the G2 checkpoint activated by CNDAC. Cells with wild-type (GM02188) and deficient in ATR (GM09703 and GM18367A) were exposed to CNDAC (40 and 60 μmol/L) for 38 h. Colcemid (100 ng/mL) was added and coincubated for additional 15 h. A minimum of 400 nuclei was counted for each mitotic index measurement. Columns, mean of three separate determinations in two similar experiments; bars, SD. L.E., longer exposure of the immunoblot.

Close modal

To further evaluate the role of ATR in G2 checkpoint activation by CNDAC, the ATR protein level in HeLa cells was knocked down to <30% of control levels by siRNA at 72 h after transfection. Phosphorylation of Chk1 kinase on Ser345 and Cdk1 kinase on Tyr15 was markedly enhanced on CNDAC treatment (Fig. 2C), as indicated by increases in the phosphorylated protein levels normalized to total proteins. This is consistent with their functional roles in signaling CNDAC-induced G2 checkpoint activation (17). This response was diminished for both proteins following siRNA knockdown of ATR. Flow cytometric analysis of portions of these same cultures showed a reduction of the CNDAC-induced G2 arrest in siRNA-treated cells.

Finally, G2 population accumulations in response to 20 to 60 μmol/L CNDAC treatment were observed in wild-type human lymphoblasts and two lines from Seckel syndrome patients with varying expression of ATR (Supplementary Fig. S3).3 Comparison of the stringency in the G2 checkpoint showed that ∼17% of the control B lymphocytes derived from a healthy donor (GM02188) were in mitosis when incubated with colcemid; this percentage dropped to 2% or 1% when cells were preincubated with 40 or 60 μmol/L of CNDAC (11% or 6%, respectively, relative to colcemid alone; P < 0.01; Fig. 2D). Similarly, when the Seckel cell line GM09703 (32), which retains ∼50% of ATR protein level compared with GM02188, was treated with 40 or 60 μmol/L of CNDAC followed by colcemid, cells exhibited mitotic scorings of 9% or 14%, respectively, relative to that of colcemid alone (P < 0.01). In contrast, another cell line, GM018367, which is derived from a Seckel syndrome patient with a splicing mutation (2101A→G) in the ATR gene (33) and has little residual expression of ATR, presented a mitotic index of 26% in response to colcemid alone. There were 9% or 11% cells in mitosis on combination treatment with CNDAC and colcemid (35% or 41%, respectively, relative to colcemid alone; P < 0.01; Fig. 2D), indicating the G2 checkpoint of Seckel cells with extremely low levels of ATR is impaired compared with ATR intact cells. Comparison of differences of mitotic indices of colcemid alone and CNDAC treatment between lines indicated that the wild-type and most severely affected Seckel cell line (GM18367A) were highly significant (P < 0.0001) in their ability to arrest in G2, whereas the less severely affected Seckel line (GM09703) was marginally different (P = 0.042) from the wild-type. Thus, optimal activation and maintenance of the G2 checkpoint by CNDAC is compromised in the absence of ATR.

DNA-PK Is Also Required for G2 Checkpoint Activation by CNDAC

To define the role of DNA-PK in activating the G2 checkpoint by CNDAC, we used human glioma cell lines proficient (M059-K) and deficient (M059-J) in DNA-PKcs. Both M059-K and M059-J cells arrested in G2 on CNDAC treatment (data not shown). Twenty-four–hour incubation of 16 μmol/L CNDAC, which caused maximal G2 arrest, was associated with Chk1 phosphorylation on Ser317 in both lines (Fig. 3A), as was H2AX phosphorylation. Each of these activities was relatively stronger in the wild-type cells. Confocal microscopy revealed more intense nuclear foci of γ-H2AX on CNDAC treatment in wild-type M059-K cells (Supplementary Fig. S2B).3 Significantly more M059-J cells progressed through G2 following CNDAC treatment and were trapped in mitosis by nocodazole compared with the wild-type (Fig. 3B). M059-K cells incubated with CNDAC had a mitotic index of 4.2%, which was only one fifth of that in cells treated with nocodazole alone (21%), whereas the mitotic index of DNA-PKcs–deficient M059-J cells (16.1%) was about half of that in cells treated with nocodazole alone (30.3%). M059-K cells pretreated with IC86621, a specific inhibitor of DNA-PKcs, before CNDAC presented mitotic indexes of 7.5% (P < 0.05) and 8.4% (P < 0.01) at 20 and 40 μmol/L, respectively, significantly greater than that of cells not exposed to the inhibitor (4.2%). These results suggest that inhibition of DNA-PKcs could mimic the response of the DNA-PK mutant to CNDAC. Thus, the G2 checkpoint activated by CNDAC becomes less stringent when DNA-PK is not functional. Subsequently, IC86621 and a second inhibitor of DNA-PKcs, NU7026, were used to further characterize the functional role of DNA-PK in the CNDAC-induced G2 checkpoint. Although these compounds alone had little effect on cell cycle distribution in ML-1 and HeLa cells, they efficiently prevented CNDAC-induced G2 arrest (Fig. 3C).

Figure 3.

DNA-PK is partially responsible for activation of the G2 checkpoint by CNDAC. A, exponentially growing DNA-PK cell lines (M059-K, wild-type; M059-J, deficient) were treated with 16 μmol/L CNDAC for 24 h. Whole-cell lysates were isolated by SDS-PAGE and subjected to immunoblot analysis with indicated antibodies. Arrows, target proteins; asterisks, nonspecific binding to antibodies. B, M059-K (black columns) and M059-J (white columns) cells were preincubated with the DNA-PK inhibitor IC86621 (20 and 40 μmol/L) for 1 h before exposure to CNDAC. Nocodazole (700 ng/mL) was introduced 13 h after addition of CNDAC and coincubated for an additional 24 h. A minimum of 400 nuclei was counted for each mitotic index measurement. Triplicate samples were analyzed by Student's t test. *, P < 0.05; **, P < 0.01. Inset, immunoblot analysis of XRCC4 in M059-K cells exposed to IC86621 (40 μmol/L) for indicated times. Cells sampled 1.5 h after γ-irradiation (100 Gy) were used as a positive control for XRCC4 phosphorylation. C, ML-1 and HeLa cells, grown in exponential phases, were exposed to indicated concentrations of CNDAC for 24 h following 1-h preincubation with two DNA-PK inhibitors, NU7026 or IC86621. Cell cycle distribution was determined by flow cytometry. D, DNA-PKcs level was knocked down by siRNA in HeLa cells and 8 μmol/L CNDAC was added 48 h after transfection. Cells were incubated for an additional 24 h and monitored by immunoblotting with indicated antibodies and by flow cytometric analysis. N.D., extremely weak band not quantitated by densitometry. Percentages in the flow cytometry profiles indicate cells with a G2-M DNA content among whole populations.

Figure 3.

DNA-PK is partially responsible for activation of the G2 checkpoint by CNDAC. A, exponentially growing DNA-PK cell lines (M059-K, wild-type; M059-J, deficient) were treated with 16 μmol/L CNDAC for 24 h. Whole-cell lysates were isolated by SDS-PAGE and subjected to immunoblot analysis with indicated antibodies. Arrows, target proteins; asterisks, nonspecific binding to antibodies. B, M059-K (black columns) and M059-J (white columns) cells were preincubated with the DNA-PK inhibitor IC86621 (20 and 40 μmol/L) for 1 h before exposure to CNDAC. Nocodazole (700 ng/mL) was introduced 13 h after addition of CNDAC and coincubated for an additional 24 h. A minimum of 400 nuclei was counted for each mitotic index measurement. Triplicate samples were analyzed by Student's t test. *, P < 0.05; **, P < 0.01. Inset, immunoblot analysis of XRCC4 in M059-K cells exposed to IC86621 (40 μmol/L) for indicated times. Cells sampled 1.5 h after γ-irradiation (100 Gy) were used as a positive control for XRCC4 phosphorylation. C, ML-1 and HeLa cells, grown in exponential phases, were exposed to indicated concentrations of CNDAC for 24 h following 1-h preincubation with two DNA-PK inhibitors, NU7026 or IC86621. Cell cycle distribution was determined by flow cytometry. D, DNA-PKcs level was knocked down by siRNA in HeLa cells and 8 μmol/L CNDAC was added 48 h after transfection. Cells were incubated for an additional 24 h and monitored by immunoblotting with indicated antibodies and by flow cytometric analysis. N.D., extremely weak band not quantitated by densitometry. Percentages in the flow cytometry profiles indicate cells with a G2-M DNA content among whole populations.

Close modal

Finally, DNA-PKcs was knocked down by siRNA in HeLa cells to a significantly lower level (10–20% of control cells) by 72 h (Fig. 3D). Cells treated with control siRNA exhibited increased phosphorylation on Chk1 kinase on Ser345 and Cdk1 kinase on Tyr15, reflecting the G2 checkpoint response to CNDAC (17). In contrast, this G2 checkpoint response was markedly reduced in DNA-PKcs–depleted cells. These molecular changes were associated with the loss of ability of DNA-PKcs knockdown cells to arrest in response to CNDAC treatment. Taken together, these results support the conclusion that DNA-PK is required for effective G2 checkpoint activation by CNDAC.

Inhibition of PIKKs Abolishes CNDAC-Induced G2 Arrest

Cellular responses to two PIKK inhibitors, caffeine and wortmannin, were used to investigate the actions of these kinases on initiation of the G2 checkpoint by CNDAC (Fig. 4A). Camptothecin, which is a topoisomerase I poison and has known responses to these PIKK inhibitors (34), was incorporated in the experimental design as a positive control (Fig. 4B). Three tumor cell lines of different origins (ML-1, HCT116, and HeLa) and an ATM-deficient fibroblast line (AT-C) were evaluated. Caffeine alone had little effect on cell cycle distribution in the above four cell lines but abolished CNDAC-induced G2 arrest in these lines. Relative to its activity against DNA-PKcs, caffeine is a 50- and 10-fold more potent inhibitor of ATM and ATR, respectively (35). Considering that ATM is likely not involved, these results support a role for ATR in G2 checkpoint activation. Wortmannin (10 μmol/L) completely abolished CNDAC-induced G2 arrest in ML-1 and ATM-mutant cells. At this relatively low concentration, DNA-PK and ATM are inhibited, but not ATR (36). As ATM protein was not detectable by immunoblot in ML-1 cells, even under the condition of IR (data not shown), these results indicate that DNA-PK is essential for activation of the G2 checkpoint by CNDAC in these cells. CNDAC-induced G2 arrest was reduced in HCT116 and HeLa cells by 10 μmol/L wortmannin from 54% and 42% to 33% and 27%, respectively, and further abolished to a level less than untreated cells (17% and 16%, respectively) by 100 μmol/L wortmannin, indicating participation of ATR in the G2 checkpoint pathway in these lines. Overall, caffeine and wortmannin inhibited camptothecin-induced G2 arrest in very similar patterns as for CNDAC.

Figure 4.

PIKK inhibitors abolish CNDAC-induced G2 arrest in comparison with camptothecin. Four cell lines, in exponential growth, were exposed to 3 mmol/L caffeine for 2 h or to either 10 or 100 μmol/L of wortmannin for 1 h. CNDAC (A) or camptothecin (CPT; B) was then added at the indicated concentrations for 24 h when cell cycle distribution was measured by flow cytometry. Percentages indicate G2-M proportions among whole-cell populations.

Figure 4.

PIKK inhibitors abolish CNDAC-induced G2 arrest in comparison with camptothecin. Four cell lines, in exponential growth, were exposed to 3 mmol/L caffeine for 2 h or to either 10 or 100 μmol/L of wortmannin for 1 h. CNDAC (A) or camptothecin (CPT; B) was then added at the indicated concentrations for 24 h when cell cycle distribution was measured by flow cytometry. Percentages indicate G2-M proportions among whole-cell populations.

Close modal

Our results support a model in which ATR and DNA-PK likely cooperate to activate the G2 checkpoint in response to CNDAC (Fig. 5). Incorporation of CNDAC into DNA leads through β-elimination to formation of CNddC, a de facto DNA chain terminator. As preliminary studies suggest that CNddC is a poor substrate for excision (37), it seems unlikely that the 3′-strand terminated by CNddC is a substrate for exonucleolytic processing. However, the resulting single-strand break or nick could be processed by nuclease(s) to forms recessed on the 5′-terminus and expanded to form a gap. Subsequently, the exposed ssDNA is coated by RPA proteins (Fig. 2A). This RPA-ssDNA complex recruits ATRIP and its binding protein, ATR, to the site of DNA damage. Rad17, which is independently recruited onto the damaged chromatin (Fig. 2B), is then phosphorylated by ATR, which may promote the activation of ATR (31). Recruitment of ATR onto damaged chromatin facilitates its phosphorylation of other substrates, such as Chk1. Activated Chk1 then phosphorylates Cdc25C, which inhibits its phosphatase activity (17). As a consequence, the equilibrium between the unphosphorylated and phosphorylated forms of Cdk1 is shifted to the inactive phosphorylated form and cell cycle progression is arrested in G2 (Fig. 3D). However, approaches to test the central role of ATR in such a response pathway, including siRNA knockdown of ATR (Fig. 2C) and ATR-deficient cells (Fig. 2D), failed to completely abolish the G2 checkpoint response to CNDAC, suggesting the participation of other sensor molecules. As cells deficient in ATM had a normal checkpoint activation to CNDAC (Fig. 1), we examined the possibility that DNA-PK may participate in this response.

Figure 5.

A model for activation of the G2 checkpoint by CNDAC. See explanation in the text.

Figure 5.

A model for activation of the G2 checkpoint by CNDAC. See explanation in the text.

Close modal

Although the nature of CNDAC-induced DNA damage has not been fully disclosed, it is clear that single-strand breaks induced by CNDAC can be transformed into DSBs (17). DNA ends, either DSBs or ssDNAs processed from nicks, are able to activate DNA-PK through Ku70 and Ku80 (3841). Although the roles of these DNA-binding subunits remain to be investigated, it is expected that the sensing capabilities of the complex would be dependent on their function. Cells that lack DNA-PKcs had a less stringent G2 arrest after CNDAC treatment than did wild-type (Fig. 3B). Similar results were obtained when DNA-PKcs was depleted with siRNA procedures (Fig. 3D) or after pharmacologic inhibition of the kinase (Figs. 3C and 4). The phosphorylation of Chk1 in response to CNDAC was reduced in cells deficient in DNA-PKcs (Fig. 3A) and after knockdown of the protein with siRNA (Fig. 3D). The phosphorylated form of Cdk1 was greatly diminished in DNA-PKcs–depleted cells, further suggesting the participation of DNA-PK in the Chk1-Cdk1–mediated checkpoint response to CNDAC. Although DNA-PK has a well-studied role in the error-prone DSB repair pathway nonhomologous end-joining, which is not cell cycle specific (42, 43), there are few reports of DNA-PK functioning in checkpoint activation (42).

Thus, both ATR and DNA-PK are clearly involved with activation of the G2 checkpoint in response to CNDAC. Prior studies have shown that each is activated by different forms of damaged DNA. Therefore, the codependency for full checkpoint activation may reflect processing of the original nick to more complex forms. Nick modification may not include double-strand DNA break formation, as ATM does not seem to have a role in the G2 checkpoint response. Alternatively, if DSBs do arise from CNddC-induced aberrant processing, DNA-PK and perhaps ATR may have a compensatory action in cells that lack ATM. DNA-PKcs possibly participates in activating Chk1, although phosphorylation of Chk1 by DNA-PKcs is not well documented. Our data presented above showed a substantial role of DNA-PKcs in this checkpoint activation. In addition, DNA-PK could contribute to CNDAC-induced G2 arrest through p53, which has been reported to coimmunoprecipitate with and be phosphorylated by DNA-PKcs (44), and help to stabilize or maintain G2 arrest after DNA damage (45, 46).

Currently, ATM and ATR are known to play distinct and cooperative roles in DNA damage responses (4). Although they share substrates, they show selectivity on substrates in response to different genotoxic stresses. Recent publications from different laboratories indicate that, during S and G2 phases, ATR activation in response to DSBs is downstream of ATM (911, 47). Our data excluded ATM, but not ATR, from activating the G2 checkpoint by CNDAC. It is likely that ATM is bypassed in the early events of damage sensing because the initial DNA damage caused by CNDAC is single-strand breaks. The exonucleases or endonucleases that process CNDAC-induced single-strand breaks or nicks and expand them into gaps recognized by RPA are unknown. Mre11 (48) and nucleases involved in excision repair pathways could be candidates for this action.

The current studies have provided more insights into the upstream signaling cascade of G2 checkpoint activation by CNDAC. Our investigation has shown that ATR and DNA-PK are responsible for sensing DNA damage induced by CNDAC and subsequent activation of the G2 checkpoint, whereas ATM is dispensable in this DNA damage response. Roles of these sensor proteins in cell survival after CNDAC treatment, which is also important and distinct from the goals of this article, are under investigation and will be addressed separately. As an orally bioavailable prodrug of CNDAC, sapacitabine is currently in clinical trials, these sensors and components of the pathways they regulate may be considered as potential targets for combination strategies. Inhibitors of both DNA-PK (49) and Chk1 (50) are currently undergoing clinical evaluation.

Grant support: CA28596, CA32839, CA100632, and Cancer Center Support Grant CA16672 from the National Cancer Institute, Department of Health and Human Services.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Dr. Xin Wang (Department of Experimental Radiation Oncology), Drs. Walter Hittelman and Tao Lu (Department of Experimental Therapeutics), and Dr. Xiao Zhou (Department of Cytokine and Supportive Oncology, M. D. Anderson Cancer Center) for their expert technical assistance and valuable discussion.

1
Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints.
Annu Rev Biochem
2004
;
73
:
39
–85.
2
Shiloh Y. ATM and related protein kinases: safeguarding genome integrity.
Nat Rev Cancer
2003
;
3
:
155
–68.
3
Hiom K. DNA repair: how to PIKK a partner.
Curr Biol
2005
;
15
:
R473
–5.
4
Shiloh Y. The ATM-mediated DNA-damage response: taking shape.
Trends Biochem Sci
2006
;
31
:
402
–10.
5
Lee JH, Paull TT. Direct activation of the ATM protein kinase by the Mre11/Rad50/Nbs1 complex.
Science
2004
;
304
:
93
–6.
6
Lee JH, Paull TT. ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex.
Science
2005
;
308
:
551
–4.
7
Paull TT, Lee JH. The Mre11/Rad50/Nbs1 complex and its role as a DNA double-strand break sensor for ATM.
Cell Cycle
2005
;
4
:
737
–40.
8
Bakkenist CJ, Kastan MB. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation.
Nature
2003
;
421
:
499
–506.
9
Jazayeri A, Falck J, Lukas C, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks.
Nat Cell Biol
2006
;
8
:
37
–45.
10
Myers JS, Cortez D. Rapid activation of ATR by ionizing radiation requires ATM and Mre11.
J Biol Chem
2006
;
281
:
9346
–50.
11
Garcia-Muse T, Boulton SJ. Distinct modes of ATR activation after replication stress and DNA double-strand breaks in Caenorhabditis elegans.
EMBO J
2005
;
24
:
4345
–55.
12
Smith GC, Divecha N, Lakin ND, Jackson SP. DNA-dependent protein kinase and related proteins.
Biochem Soc Symp
1999
;
64
:
91
–104.
13
Collis SJ, DeWeese TL, Jeggo PA, Parker AR. The life and death of DNA-PK.
Oncogene
2005
;
24
:
949
–61.
14
Hanaoka K, Suzuki M, Kobayashi T, et al. Antitumor activity and novel DNA-self-strand-breaking mechanism of CNDAC (1-(2-C-cyano-2-deoxy-β-d-arabino-pentofuranosyl) cytosine) and its N4-palmitoyl derivative (CS-682).
Int J Cancer
1999
;
82
:
226
–36.
15
Azuma A, Huang P, Matsuda A, Plunkett W. 2′-C-cyano-2′-deoxy-1-β-d-arabino-pentofuranosylcytosine: a novel anticancer nucleoside analog that causes both DNA strand breaks and G(2) arrest.
Mol Pharmacol
2001
;
59
:
725
–31.
16
Azuma A, Nakajima Y, Nishizono N, et al. Nucleosides and nucleotides. 122. 2′-C-cyano-2′-deoxy-1-β-d-arabinofuranosylcytosine and its derivatives. A new class of nucleoside with a broad antitumor spectrum.
J Med Chem
1993
;
36
:
4183
–9.
17
Liu X, Guo Y, Li Y, et al. Molecular basis for G2 arrest induced by 2′-C-cyano-2′-deoxy-1-β-d-arabino-pentofuranosylcytosine and consequences of checkpoint abrogation.
Cancer Res
2005
;
65
:
6874
–81.
18
Ziv Y, Bar-Shira A, Pecker I, et al. Recombinant ATM protein complements the cellular A-T phenotype.
Oncogene
1997
;
15
:
159
–67.
19
Anderson CW, Allalunis-Turner MJ. Human TP53 from the malignant glioma-derived cell lines M059J and M059K has a cancer-associated mutation in exon 8.
Radiat Res
2000
;
154
:
473
–6.
20
Anderson CW, Dunn JJ, Freimuth PI, Galloway AM, Allalunis-Turner MJ. Frameshift mutation in PRKDC, the gene for DNA-PKcs, in the DNA repair-defective, human, glioma-derived cell line M059J.
Radiat Res
2001
;
156
:
2
–9.
21
Matsuda A, Nakajima Y, Azuma A, Tanaka M, Sasaki T. Nucleosides and nucleotides. 100. 2′-C-cyano-2′-deoxy-1-β-d-arabinofuranosyl-cytosine (CNDAC): design of a potential mechanism-based DNA-strand-breaking antineoplastic nucleoside.
J Med Chem
1991
;
34
:
2917
–9.
22
Zou L, Cortez D, Elledge SJ. Regulation of ATR substrate selection by Rad17-dependent loading of Rad9 complexes onto chromatin.
Genes Dev
2002
;
16
:
198
–208.
23
Wang X, Zou L, Lu T, et al. Rad17 phosphorylation is required for claspin recruitment and Chk1 activation in response to replication stress.
Mol Cell
2006
;
23
:
331
–41.
24
Casper AM, Nghiem P, Arlt MF, Glover TW. ATR regulates fragile site stability.
Cell
2002
;
111
:
779
–89.
25
Shang Y, Brown M. Molecular determinants for the tissue specificity of SERMs.
Science
2002
;
295
:
2465
–8.
26
Xu B, Kim ST, Lim DS, Kastan MB. Two molecularly distinct G(2)/M checkpoints are induced by ionizing irradiation.
Mol Cell Biol
2002
;
22
:
1049
–59.
27
Binz SK, Sheehan AM, Wold MS. Replication protein A phosphorylation and the cellular response to DNA damage.
DNA Repair (Amst)
2004
;
3
:
1015
–24.
28
Bomgarden RD, Yean D, Yee MC, Cimprich KA. A novel protein activity mediates DNA binding of an ATR-ATRIP complex.
J Biol Chem
2004
;
279
:
13346
–53.
29
Barr SM, Leung CG, Chang EE, Cimprich KA. ATR kinase activity regulates the intranuclear translocation of ATR and RPA following ionizing radiation.
Curr Biol
2003
;
13
:
1047
–51.
30
Cortez D, Guntuku S, Qin J, Elledge SJ. ATR and ATRIP: partners in checkpoint signaling.
Science
2001
;
294
:
1713
–6.
31
Zou L, Elledge SJ. Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes.
Science
2003
;
300
:
1542
–8.
32
Alderton GK, Joenje H, Varon R, et al. Seckel syndrome exhibits cellular features demonstrating defects in the ATR-signalling pathway.
Hum Mol Genet
2004
;
13
:
3127
–38.
33
O'Driscoll M, Ruiz-Perez VL, Woods CG, Jeggo PA, Goodship JA. A splicing mutation affecting expression of ataxia-telangiectasia and Rad3-related protein (ATR) results in Seckel syndrome.
Nat Genet
2003
;
33
:
497
–501.
34
Pommier Y, Redon C, Rao VA, et al. Repair of and checkpoint response to topoisomerase I-mediated DNA damage.
Mutat Res
2003
;
532
:
173
–203.
35
Sarkaria JN, Busby EC, Tibbetts RS, et al. Inhibition of ATM and ATR kinase activities by the radiosensitizing agent, caffeine.
Cancer Res
1999
;
59
:
4375
–82.
36
Sarkaria JN, Tibbetts RS, Busby EC, et al. Inhibition of phosphoinositide 3-kinase related kinases by the radiosensitizing agent wortmannin.
Cancer Res
1998
;
58
:
4375
–82.
37
Wang Y, Matsuda A, Plunkett W. Excision of 2′-C-cyano-2′-deoxy-1-β-d-arabino-pentofuranosylcytosine (CNDAC) residues from DNA.
Proc Am Assoc Cancer Res
2006
;
47
:
122
.
38
Blier PR, Griffith AJ, Craft J, Hardin JA. Binding of Ku protein to DNA. Measurement of affinity for ends and demonstration of binding to nicks.
J Biol Chem
1993
;
268
:
7594
–601.
39
Morozov VE, Falzon M, Anderson CW, Kuff EL. DNA-dependent protein kinase is activated by nicks and larger single-stranded gaps.
J Biol Chem
1994
;
269
:
16684
–8.
40
Gottlieb TM, Jackson SP. The DNA-dependent protein kinase: requirement for DNA ends and association with Ku antigen.
Cell
1993
;
72
:
131
–42.
41
Suwa A, Hirakata M, Takeda Y, et al. DNA-dependent protein kinase (Ku protein-p350 complex) assembles on double-stranded DNA.
Proc Natl Acad Sci U S A
1994
;
91
:
6904
–8.
42
Burma S, Chen DJ. Role of DNA-PK in the cellular response to DNA double-strand breaks.
DNA Repair (Amst)
2004
;
3
:
909
–18.
43
Lieber MR, Ma Y, Pannicke U, Schwarz K. The mechanism of vertebrate nonhomologous DNA end joining and its role in V(D)J recombination.
DNA Repair (Amst)
2004
;
3
:
817
–26.
44
Achanta G, Pelicano H, Feng L, Plunkett W, Huang P. Interaction of p53 and DNA-PK in response to nucleoside analogues: potential role as a sensor complex for DNA damage.
Cancer Res
2001
;
61
:
8723
–9.
45
Bunz F, Dutriaux A, Lengauer C, et al. Requirement for p53 and p21 to sustain G2 arrest after DNA damage.
Science
1998
;
282
:
1497
–501.
46
Taylor WR, Stark GR. Regulation of the G2/M transition by p53.
Oncogene
2001
;
20
:
1803
–15.
47
Cuadrado M, Martinez-Pastor B, Murga M, et al. ATM regulates ATR chromatin loading in response to DNA double-strand breaks.
J Exp Med
2006
;
203
:
297
–303.
48
Paull TT, Gellert M. The 3′ to 5′ exonuclease activity of Mre 11 facilitates repair of DNA double-strand breaks.
Mol Cell
1998
;
1
:
969
–79.
49
Zhao Y, Thomas HD, Batey MA, et al. Preclinical evaluation of a potent novel DNA-dependent protein kinase inhibitor NU7441.
Cancer Res
2006
;
66
:
5354
–62.
50
Sampath D, Cortes J, Estrov Z, et al. Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial.
Blood
2006
;
107
:
2517
–24.