Anda di halaman 1dari 8

ANTICANCER RESEARCH 26: 1909-1916 (2006)

DNA Damage Responses in Cell Cycle G2


Phase and Mitosis – Tracking and Targeting
ANNE HANSEN REE1,2, TROND STOKKE3, ÅSE BRATLAND1,2, SEBASTIAN PATZKE4,
RAGNHILD V. NOME1, SIGURD FOLKVORD1, LEONARDO A. MEZA-ZEPEDA1,
KJERSTI FLATMARK1,5, ØYSTEIN FODSTAD1 and YVONNE ANDERSSON1

Departments of 1Tumor Biology, 2Oncology, 3Radiation Biology, 4Immunology and


5Surgical Oncology, The Norwegian Radium Hospital, 0310 Oslo, Norway

Abstract. Background: In order to determine temporal if DNA is properly repaired or, if not, to cell death (2, 3).
responses of cell cycle populations to DNA damage, a rational Much effort has been directed to estimating the duration of
combination of cell cycle analyses is critical. Moreover, the individual cell cycle phases and changes in their kinetics to
targeting of cell cycle checkpoint responses may modify the define strategies that might override checkpoint responses
cytotoxic effect of DNA damage. Materials and Methods: The and, thereby, enhance therapeutic tumor cell killing. In
characteristics of cell cycle populations (DNA content, cell order to determine such temporal responses, a rational
cycle transitioning of S phase cells and size of mitotic cell combination of cell cycle analyses is critical.
fraction within the total G2/M phase population) in HeLa cells This study was aimed at presenting DNA damage-
exposed to ionizing radiation were analyzed using three induced cell cycle profiles deduced from the combination of
individual flow cytometry-based assays. The potential three individual cytometry assays. The relative distribution
radiosensitization from inhibiting DNA damage responses was of individual cell cycle populations was determined by
assessed by the colony formation assay. Results: Irradiation staining cell suspensions for DNA content. Cells in the
resulted in an initial accumulation of S phase cells in G2 S phase were labeled with 5-bromo-2’-deoxyuridine (BrdU)
phase, from which the arrested cells were subsequently released to allow tracking of their cell cycle transitioning, and the
to enter mitosis. Upon drug inhibition of G2 checkpoint size of the mitotic cell fraction within the total G2/M phase
signaling or mitotic progression, the cytotoxic effect of ionizing population was specifically measured by simultaneous
radiation on the HeLa cells was amplified. Conclusion: DNA analysis of DNA content and light scattering.
damage-induced cell cycle responses, analyzed by selected While the signaling pathway via the tumor-suppressor
cytometry assays and modified by specific targeting, might protein p53, the primary regulator of the G1 checkpoint, is
contribute to an understanding of how to improve radiotherapy often defective in human solid tumors, the DNA damage-
outcome. activated G2 checkpoint signaling, initiated by ATM,
communicates through mediators like Chk1 and Chk2 (1),
In proliferating cells, DNA damage delays normal cell cycle which are essential for the checkpoint response and might,
progression. A complex network of responses is activated as therefore, be candidates for therapeutic targeting (4). A
soon as the damage is registered in the genome, and these variety of pharmacological compounds, designed to target
are rapidly manifested at cell cycle checkpoints. Massive cell cycle regulatory mechanisms, has been shown to
insults to DNA, such as double-strand DNA breaks override the DNA damage defense response that prevents
following cellular exposure to ionizing radiation, may induce mitotic entry (4). Such G2 checkpoint targeting agents may
checkpoint responses in essentially any phase of the cell have therapeutic potential as radio- or chemosensitizers by
cycle (1), ultimately leading to the outcome of cell survival facilitating cell death through mitotic catastrophe.
Moreover, taxanes, which disrupt chromosome segregation
in mitotis, are currently utilized clinically as radiosensitizers
in the treatment of non-small cell lung cancer and head and
Correspondence to: Anne Hansen Ree, Department of Tumor
neck cancer (5).
Biology, The Norwegian Radium Hospital, 0310 Oslo, Norway. Tel:
+47 22-935421, Fax: +47 22-522421, email: a.h.ree@medisin.uio.no
Cervical cancer, which is rated among the most common
malignancies globally, is etiologically linked to infection with
Key Words: Cell cycle, G2 checkpoint, mitosis, molecular targeting, human papillomavirus types 16 and 18 (6). It has been
cervical carcinoma. known, for more than a decade, that the cervical carcinoma

0250-7005/2006 $2.00+.40 1909


ANTICANCER RESEARCH 26: 1909-1916 (2006)

HeLa cell line harbors the human papillomavirus type 18 NaCl, 0.5 mM EDTA, 0.1 mM phenylmethylsulphonyl fluoride and
strain and therefore expresses the viral oncoprotein E6, 10 nM phosphate buffer (pH7.4), before a 1/4 volume of 4%
which stably complexes with the cellular p53 protein, paraformaldehyde was added. After fixation on ice for at least 1 h,
the cells were stained with 12.5 Ìg/ml propidium iodide in 0.1%
encoded by the wild-type TP53 gene, to facilitate ubiquitin-
Nonidet P40, 150 mM NaCl, 0.5 mM EDTA and 10 mM phosphate
mediated p53 degradation (7, 8). By this mechanism, E6 buffer (pH7.4) containing 100 Ìg/ml RNase A, and were analyzed
also disrupts cell cycle G1 arrest in response to DNA by flow cytometry. The simultaneous measurement of propidium
damage (9). Given its lack of functional G1 checkpoint iodide fluorescence and light scattering allows for the
along with its rapid proliferation rate (estimated cell cycling determination of mitotic cells since this population binds more dye
time of ~18 h (data not shown)) and highly reproducible than G1- and S-phase cells and scatters less light than G2 phase
colony formation capacity (for evaluation of the in vitro end- cells, respectively (11). Cells treated with 500 ng/ml nocodazole
(Sigma-Aldrich Company Ltd.), a mitotic spindle poison, and
point effect of DNA damage), the HeLa cell line appeared
sampled after 12 and 24 h, were analyzed as positive controls for
to be a valid experimental model for tracking and targeting cell cycle arrest in mitosis.
cell cycle responses to DNA damage. All experiments for the flow cytometry analyses were repeated
at least three times independently.
Materials and Methods
Western blot analysis. Protein expression was measured by means of
Cell cultures and experimental treatments. The HeLa cells were the standard Western blot technique, essentially as previously
routinely grown in RPMI 1640 medium supplemented with 10% described (10). The primary antibodies were anti-p53 (SC-6243;
fetal bovine serum (FBS) and 2.0 mM glutamine. The breast Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-MDM2
carcinoma MCF-7 cells were kept in Dulbecco’s Minimum (MAB4134; Chemicon International, Temecula, CA, USA), anti-
Essential Medium supplemented with 5% FBS in addition to Phospho-Chk1 (#2341; Cell Signaling Technology Inc., Beverly,
glutamine. High-energy radiation from a 60Co source was delivered MA, USA) and anti-·-tubulin (CP06; Calbiochem/Merck
at a rate of approximately 0.75 Gy/min. The unirradiated control Biosciences Ltd.). The MAB4134 recognizes an epitope, possibly
cells were simultaneously placed at room temperature to obtain hypophosphorylated (12), within amino acid residues 154–167 of the
comparable conditions. In experiments using the selective Chk1 human MDM2 oncoprotein, and the anti-Phospho-Chk1 antibody
kinase inhibitor UCN-01 (National Cancer Institute, Bethesda, detects Chk1 only when activated by serine 345 phosphorylation.
MD, USA), this compound was added at a final concentration of Immune complexes were detected with appropriate horseradish
100 nM to the cell medium, as recommended by the supplier, 15 peroxidase-coupled secondary antibodies, and enzyme activity was
min before irradiation. visualized with enzyme-linked chemiluminescence (Amersham
Biosciences Ltd.). All experiments for Western blot analyses were
Flow cytometry analyses. repeated at least twice independently.
a) Hoechst 33258 staining and analysis: Cells were pelleted in PBS
and fixed in 100% methanol. The cells were subsequently stained Assessment of clonogenic regrowth. The clonogenic regrowth
with 1.5 Ìg/ml Hoechst 33258 in phosphate-buffered saline (PBS) efficiency of the HeLa cells was determined by plating single cells
before the DNA content was analyzed by FACS flow cytometry suspended in medium for 6 h prior to irradiation. The appropriate
(Becton Dickinson, Franklin Lakes, NJ, USA), essentially as plating density was aimed to produce 20 to 40 surviving colonies in
previously described (10). The Modfit cell cycle program (Verity each well of 6-well culture plates. Medium containing paclitaxel
Software House Inc., Topsham, ME, USA) was used to determine (Sigma-Aldrich Company Ltd.) was applied to the designated cell
the fractions of cells in the G1, S and G2/M phases from the cell cultures 12 h after irradiation and replaced by fresh medium after
cycle distribution. a drug exposure period of 24 h. For all experimental conditions,
b) BrdU labeling, staining and analysis: Cells were pulse-labeled with the cell colonies were fixed and stained with 0.1% crystal violet
30 ÌM BrdU (Sigma-Aldrich Company Ltd., Dorset, UK) in the after a total incubation time of 7 days following radiation exposure.
culture medium for 30 min and the experimental treatment was then Colonies of ≥50 cells were counted for computation of the
applied. Harvested cells were fixed in 100% methanol, as described surviving fraction, and at least four parallel samples were scored in
above, and extracted in 2 M HCl and 0.5% Triton X-100. The cells the thee independent repetitions performed for each treatment
were subsequently stained for BrdU incorporation for 30 min at room condition.
temperature with 20 Ìl anti-BrdU (Becton Dickinson) per one million
cells in 1 ml Tween 20 and 1% bovine serum albumin in 0.5% PBS, Results
followed by incubation for 30 min at room temperature with the
secondary FITC-labeled anti-mouse antibody (DakoCytomation A/S, Cell cycle redistribution following irradiation. Exponentially-
Glostrup, Denmark) diluted 1:25 in 0.5% Tween 20 and 1% bovine proliferating HeLa cells were exposed to a radiation dose of
serum albumin in PBS. Finally, the cells were stained for 15 min at 8.0 Gy, and the redistribution of the cells within the cell cycle
37ÆC with 5 Ìg/ml propidium iodide (Calbiochem/Merck Biosciences
phases was followed for up to 48 h after the DNA-damaging
Ltd., Nottingham, UK) and 100 Ìg/ml RNase A (Amersham
Biosciences Ltd., Buckinghamshire, UK) in PBS, and were analyzed
treatment. As seen from Figure 1, the radiation exposure
by flow cytometry. initially resulted in a progression of G1 and S phase cells into
c) Propidium iodide staining and analysis: Cells pelleted in PBS the G2/M phase, which was ~50% of the total cell count
were extracted in a 3/4 volume of 0.1% Nonidet P40, 150 mM after 12 h. This G2/M phase accumulation seemed to persist

1910
Ree et al: Cell Cycle Responses to DNA Damage

Figure 1. Time-dependent distribution of HeLa cell cycle phases following exposure to ionizing radiation. The cells were irradiated with a dose of 8.0 Gy
and were further incubated for the indicated time-periods before cellular DNA content was determined by flow cytometry analysis gated for Hoechst
33258 fluorescence. Cells with DNA contents characteristic for G1 and G2/M phase cells were found in channel numbers ~200 and ~400 along the
x-axes, respectively. Scales indicating cell counts (y-axes) are provided, as are the percentages of G1, S and G2/M phase cells of each total cell count
(below the DNA histograms).

for a period of less than 24 h after the DNA damage, as an (recovered to baseline; compare the right histogram of Figure
almost equally large fraction of the cells (~45%) was also 3 with the left and middle ones), consisting of cells that had
detected as a G1 phase population at 24 h. been released from the initial radiation-induced G2 arrest.
A more detailed profiling of the G2/M phase detention
showed that the DNA-damaged HeLa cells apparently Lack of functional cell cycle G1 checkpoint in HeLa cells. The
responded by initially delaying cell cycle progression throughout HeLa cells displayed a lack of characteristics consistent with
the G2 phase, before the cells subsequently escaped G2 arrest a functional G1 checkpoint; e.g., absence of both radiation-
and were released into mitosis (Figures 2 and 3). induced cell cycle G1 arrest (Figure 1) and mRNA
Indeed, those cells that had been BrdU-labeled shortly expression of CDKN1A, encoding the G1 cell cycle inhibitor
before irradiation (S phase cells) were detected within the p21 (data not shown). All exons of the TP53 gene were
G2/M phase population already 6 h later (Figure 2, lower left confirmed as wild-type upon analysis of DNA by constant
histogram) and seemed to be consolidated therein 12 h after denaturant gel electrophoresis (data not shown), a method
DNA damage (Figure 2, upper right histogram). At this that detects one-base substitutions, if any, in the DNA
time-point, the mitotic fraction of the G2/M phase fragments analyzed with high sensitivity (13).
population was significantly smaller than in the unirradiated Essentially, after DNA damage the protein encoded by
HeLa cells (compare the left and middle histograms of wild-type TP53 is rapidly stabilized by post-translational
Figure 3), clearly indicating that cells that had arrested in G2 modifications, leading to enhanced expression levels of p53
phase had failed to progress further into mitosis. But yet, (14). However, as seen in Figure 4A, irradiation (8.0 Gy)
12 h after DNA damage, the HeLa cells that had been in G1 of the HeLa cells did not up-regulate the level of p53
phase when irradiated seemed to retain the ability to make protein expression. The transient radiation-induced p53
cell cycle transition (observe the shift of cells negative for level in the wild-type TP53 MCF-7 cells was used as a
BrdU labeling in the upper right histogram of Figure 2). positive control.
Furthermore, 24 h after DNA damage, when the majority p53 stabilization results primarily from disruption of the
of the HeLa cells were detected in either the G1 or G2/M interaction between p53 and the MDM2 oncoprotein, which
phases of the cell cycle (as consistently demonstrated by all thereby protects p53 from ubiquitin-mediated degradation
sets of flow cytometry data), cells that had escaped G2 arrest (15). In contrast to the MCF-7 cells, in which the p53
(BrdU-negative cells) initially after irradiation now seemed induction 1 to 3 h after irradiation (8.0 Gy) was followed by
to recur in the next G1 phase, as did some cells that had been an up-regulation of MDM2 expression after 3 to 6 h, as
in the S phase (BrdU-positive cells) when DNA damage expected (16), the HeLa cells instead showed a rapidly
occured (Figure 2, lower right histogram). This is in induced overexpression of an upper major MDM2 band
accordance with the observed size of the mitotic fraction 15 min after the DNA-damaging treatment had been

1911
ANTICANCER RESEARCH 26: 1909-1916 (2006)

Figure 2. Time-dependent responses of HeLa S phase cells following exposure to ionizing radiation. The cells were pulse-labeled with BrdU before being
irradiated with a dose of 8.0 Gy and were further incubated for the indicated time-periods. The distribution of cells positive and negative for BrdU (x-axes)
was determined by flow cytometry analysis gated for anti-BrdU-FITC (R1), and emission values exceeding ~103 were considered positive for BrdU labeling.
The propidium iodide content was also analyzed, and G1 and G2/M phase cells were found in channel numbers ~50 and ~100 along the y-axes, respectively.

Figure 3. Time-dependent M phase effects in HeLa cells exposed to ionizing radiation. The cells were irradiated with a dose of 8.0 Gy and were further
incubated for the indicated time-periods before propidium iodide fluorescence (x-axes) and light scattering (y-axes) were determined by flow cytometry
analysis. The cells with DNA contents characteristic for G1 and G2/M phase cells were found in channel numbers ~200 and ~400 along the x-axes,
respectively. Along the y-axes, however, G1 and M phase cells were detected in channel numbers 200-400, whereas S and G2 phase cells were registered
in channel numbers 400-500 and 500-700, respectively. Mitotic cell populations are indicated along with the percentages of M phase cells of each total
cell count. The percentages of M phase cells upon treatment with 500 ng/ml nocodazole for 12 and 24 h were 8.6 and 11.4, respectively (data not shown).

1912
Ree et al: Cell Cycle Responses to DNA Damage

Figure 4. Checkpoint regulatory proteins in HeLa cells after exposure to ionizing radiation (IR). The cells were treated (+) with IR (8.0 Gy) or were left
untreated (–), and the expression of p53, MDM2 and phosphorylated Chk1 (Chk1-P) was analyzed by Western blot hybridization after the indicated
time- periods. In both sets of analyses, the expression of ·-tubulin was measured as the loading control. (A) The levels of p53 and MDM2 in the HeLa
cells were compared with those found in the breast carcinoma MCF-7 cell line, harboring intact p53 function. (B) The IR-dependent HeLa cell expression
of Chk1 phosphorylated on serine residue 345 was analyzed in the absence (–) or presence (+) of UCN-01 (100 nM).

initiated (Figure 4A). Human tumors with MDM2 gene


amplification, with or without concomitant oncogenic
MDM2 activity, may often show phenotypic inactivation of
their wild-type TP53 gene (17). In the HeLa cells, however,
MDM2 displayed as a normal copy number gene by Southern
blot analysis (18) (data not shown).

Clonogenic regrowth upon ionizing radiation – the impact of


cell cycle G2 and M phase responses. Based on the
observations that the HeLa cells were arrested in the G2
phase 12 h after irradiation but had entered mitosis 24 h
after DNA damage, the possibility of radiosensitization upon
inhibitory targeting of G2 checkpoint signaling or mitotic
progression, essentially by amplifying the cytotoxic effect of
ionizing radiation on clonogenic regrowth, was evaluated.
The HeLa cells were exposed to increasing doses of
ionizing radiation (2.0 to 8.0 Gy) to determine clonogenic
survival (Figure 5). This treatment caused a dose-dependent
loss of colony formation, with a surviving fraction of ~0.12
with the highest radiation dose applied. Figure 5. Inhibition of Chk1 or mitosis modulates the outcome of HeLa
clonogenic regrowth after exposure to ionizing radiation (IR). The cells
The regulatory mechanism of ionizing radiation on G2 phase
were treated with increasing IR doses in the absence (●) or presence of
responses signals through Chk1, as demonstrated previously either UCN-01 (100 nM ● ) or paclitaxel in increasing concentrations (1
(10, 19, 20). Consistently, as seen in Figure 4B, Chk1 nM ■ , 5 nM ■ and 10 nM ▼), to determine relative colony formation
phosphorylation was strongly induced in HeLa cells 1 h after compared to the unirradiated control (mean±SEM, n=3).

1913
ANTICANCER RESEARCH 26: 1909-1916 (2006)

radiation exposure (8.0 Gy). This induction was attenuated after of cell cycle phase transitioning. Twenty-four hours after
12 h. Furthermore, treatment with the Chk1 inhibitor UCN-01 irradiation, the cell cycle G1 population did not exclusively
(100 nM) counteracted the Chk1 phosphorylation response consist of cells that had escaped G2 arrest, but also of cells
almost completely after 1 h, but apparently less efficiently positive for BrdU labeling (irradiated S phase cells). Thus,
throughout the observation period of 12 h. the G2 checkpoint detention of DNA-damaged S phase cells
As a consequence of this inhibitory effect on DNA did not seem to be absolute, and a portion of the irradiated
damage-induced G2 phase responses mediated by Chk1, the S phase population seemed capable of escaping cell death,
possible radiosensitizing effect of UCN-01 (100 nM) on the which might be directly associated with the relative
HeLa cell clonogenicity was measured (Figure 5). Whereas resistance of HeLa cells to irradiation (observed clonogenic
the ability of clonogenic regrowth after irradiation with the regrowth fraction of ~0.12 at 8.0 Gy).
lower doses (2.0 and 5.0 Gy) was modestly reduced when The HeLa cells displayed a typical phenotype of tumor
UCN-01 was present compared to when absent, the cells lacking a functional G1 checkpoint as the G1 phase cells
cytotoxic effect of 8.0 Gy of ionizing radiation was ~3-fold rapidly disappeared while the G2/M phase population
amplified by UCN-01. expanded following DNA damage. The HeLa TP53 genotype
Since G2 phase-arrested HeLa cells had entered mitosis was confirmed to be wild-type but was inconsistent with any
24 h after radiation exposure, we chose to evaluate the radiation-induced p53 protein expression. It has been known,
possible radiosensitizing effect of a mitotic inhibitor during for more than a decade, that the viral oncoprotein E6
the period of cell cycle redistribution when the cells were expressed by HeLa cells facilitates ubiquitin-mediated p53
regenerating the ability of G2/M transitioning. Hence, degradation (7) by a mechanism that overrides endogenous
paclitaxel was added to the cells 12 h after irradiation and MDM2 activity (8, 21). Yet, the HeLa p53 protein has been
was subsequently removed after 24 h of drug exposure, and found to be functionally reactivated by therapeutics that
HeLa cell clonogenicity was compared with that of cells interfere with the p53-MDM2 interaction (22, 23).
treated with ionizing radiation alone. In the cells treated In the MCF-7 cells, irradiation led to a rapid decrease in
with paclitaxel (1-10 nM), the ability for clonogenic the expression level of the two major MDM2 polypeptides
regrowth after irradiation was reduced by a factor of 2-10, (possibly hypophosphorylated (12)) detected with an
depending on which paclitaxel concentrations and radiation antibody against amino acid residues 154-167, clearly
doses applied (Figure 5). preceding the accumulation of the p53 protein (12, 24). In
the HeLa cells, however, an apparent overexpression of the
Discussion upper MDM2 band, rather than its down-regulation, was
seen 15 min after the DNA-damaging treatment was
In this report, we aimed to distinguish temporal cell cycle initiated. This might suggest that the mechanism involving
responses to DNA damage by combining flow cytometry- MDM2 phosphorylation (12, 24) in the signaling pathway
based assays that characterize the distribution of individual from DNA damage to p53 activation is defective and may,
cell cycle phase populations and the kinetics of their in addition to the oncogenic E6 activity, partially account
transitioning. We chose to treat the relatively radioresistant for the lack of p53 stabilization.
HeLa cell line with rather high radiation doses in the hope The DNA damage response communicated through the
of obtaining clearly defined effects on cell cycle phenotypes G2 checkpoint signaling pathway was evaluated as a
and colony formation capacity. Irradiation resulted in an therapeutic target to possibly enhance HeLa cell
initial accumulation of S phase cells in the G2 phase, from radiosensitivity. Even though UCN-01 counteracted
which the arrested cells were subsequently released to enter radiation-induced Chk1 phosphorylation on serine residue
mitosis. Upon drug inhibition of the G2 checkpoint signaling 345, consistent with data demonstrated by others (25), the
or mitotic progression, the cytotoxic effect of ionizing radiosensitizing effect of this targeting drug seemed to be
radiation on HeLa cells was amplified, consistent with the modest compared to the enhanced cytotoxic effect
concept of an increased probability of tumor cell death previously reported in irradiated HeLa cells with a defective
when cellular DNA damage-responses are targeted. Chk1 phenotype (26). This difference might be explained by
Double-strand DNA breaks may induce checkpoint a transient nature of the pharmacological inhibition
responses in essentially any phase of the cell cycle (1). In indicated by the partial recovery of radiation-induced Chk1
the present study, we could follow the responses of BrdU- phosphorylation after 3 to 12 h of UCN-01 incubation.
labeled cells (S phase cells) as well as the size of the mitotic Moreover, the signal transduction cascade downstream of
fraction within the total G2/M phase population. Whereas the damaged DNA is very complex and involves a variety of
the DNA-damaged S phase cells initially arrested in G2 G2 checkpoint effector mechanisms (1), inviting alternative
phase before they progressed into mitosis, their irradiated effector pathways to compensate when one particular
counterparts from G1 phase retained the immediate ability effector molecule is inhibited.

1914
Ree et al: Cell Cycle Responses to DNA Damage

The possible radiosensitizing effect of the mitotic 5 Choy H: Taxanes in combined modality therapy for solid
inhibitor paclitaxel was evaluated using a concentration tumors. Crit Rev Oncol Hematol 37: 237-247, 2001.
range of the compound within which the fraction of mitotic 6 zur Hausen H: Papillomavirus infections – a major cause of
human cancers. Biochim Biophys Acta 1288: F55-F78, 1996.
HeLa cells has been found to increase sharply from lack of
7 Scheffner M, Werness BA, Huibregtse JM, Levine AJ and
mitotic arrest (at 1 nM) to 50-70% arrested cells (at 10 nM) Howley PM: The E6 oncoprotein encoded by human
(27, 28). Yet, the clonogenic survival data indicated that papillomavirus types 16 and 18 promotes the degradation of
paclitaxel sensitized the HeLa cells to the cytotoxic effect of p53. Cell 63: 1129-1136, 1990.
ionizing radiation within the entire concentration range. 8 Hoppe-Seyler F and Butz K: Repression of endogenous p53
The temporal aspect of using a mitotic inhibitor to obtain transactivation function in HeLa cervical carcinoma cells by
tumor cell radiosensitization, i.e., timing of paclitaxel human papillomavirus type 16 E6, human mdm-2, and mutant
p53. J Virol 67: 3111-3117, 1993.
treatment to the period of cell cycle redistribution when the
9 Kessis TD, Slebos RJ, Nelson WG, Kastan MB, Plunkett BS,
cells regenerate the ability of G2/M transitioning, is
Han SM, Lorincz AT, Hedrick L and Cho KR: Human
probably critical (29). papillomavirus 16 E6 expression disrupts the p53-mediated
There is strong scientific evidence that concomitant cellular response to DNA damage. Proc Natl Acad Sci USA 90:
cisplatin-based chemotherapy improves disease-free and 3988-3992, 1993.
overall survival compared to radiotherapy alone for high- 10 Ree AH, Bratland Å, Nome RV, Stokke T and Fodstad Ø:
risk early stage cervical cancer. Concurrent use of Repression of mRNA for the PLK cell cycle gene after DNA
cisplatin is also recommended in radiation treatment of damage requires BRCA1. Oncogene 22: 8952-8955, 2003.
11 Larsen JK, Munch-Petersen B, Christiansen J and Jørgensen K:
locally advanced disease stages, even though randomized
Flow cytometric discrimination of mitotic cells: resolution of M,
studies have failed to prove that the addition of cisplatin as well as G1, S, and G2 phase nuclei with mithramycin,
to irradiation is clearly advantageous for outcome. propidium iodide, and ethidium bromide after fixation with
Antimetabolites (hydroxyurea and 5-fluorouracil) have formaldehyde. Cytometry 7: 54-63, 1986.
also been evaluated as radiosensitizing compounds in 12 de Toledo SM, Azzam EI, Dahlberg WK, Gooding TB and
cervical cancer, but have not been found to be superior Little JB: ATM complexes with HDM2 and promotes its rapid
to cisplatin (30, 31). Whether cell cycle response data phosphorylation in a p53-independent manner in normal and
tumor human cells exposed to ionizing radiation. Oncogene 19:
obtained from cell line models like ours might translate
6185-6193, 2000.
into strategies to improve the radiotherapy outcome in 13 Hovig E, Smith-Sørensen B, Brøgger A and Børresen AL:
cervical carcinoma requires further experimental Constant denaturant gel electrophoresis, a modification of
approaches, but hints, if anything, at an appealing denaturing gradient gel electrophoresis in mutation detection.
concept. Mutat Res 262: 63-67, 1991.
14 Fei P and El-Deiry WS: p53 and radiation responses. Oncogene
Acknowledgements 22: 5774-5783, 2003.
15 Meek DW and Knippschild U: Posttranslational modification
of MDM2. Mol Cancer Res 1: 1017-1026, 2003.
This work was supported by the Norwegian Cancer Society grant
16 Xia L, Paik A and Li JJ: p53 activation in chronic radiation-
C-04083 (to Anne Hansen Ree). We thank Kyowa Hakko Kogyo
treated breast cancer cells: regulation of MDM2/p14ARF.
Co., Ltd. (Tokyo, Japan) and the Division of Cancer Treatment
Cancer Res 64: 221-228, 2004.
and Diagnosis, National Cancer Institute (Bethesda, MD, USA) for
17 Zhang R and Wang H: MDM2 oncogene as a novel target for
the gift of UCN-01. We greatly appreciate the technical support
human cancer therapy. Curr Pharm Des 6: 393-416, 2000.
from Mali Strand Ellefsen and Kirsti Solberg Landsverk
(Department of Radiation Biology, The Norwegian Radium 18 Forus A, Flørenes VA, Maelandsmo GM, Meltzer PS, Fodstad
Hospital, Oslo, Norway). Ø and Myklebost O: Mapping of amplification units in the
q13-14 region of chromosome 12 in human sarcomas: some
amplica do not include MDM2. Cell Growth Diff 4: 1065-
References 1070, 1993.
19 Ree AH, Bratland Å, Nome RV, Stokke T, Fodstad Ø and
1 Kastan MB and Bartek J: Cell-cycle checkpoints and cancer. Andersson Y: Inhibitory targeting of checkpoint kinase
Nature 432: 316-323, 2004. signaling impairs radiation-induced cell cycle gene regulation:
2 Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R a therapeutic strategy in tumor cell radiosensitization?
and Kroemer G: Cell death by mitotic catastrophe: a molecular Radiother Oncol 72: 305-310, 2004.
definition. Oncogene 23: 2825-2837, 2004. 20 Nome RV, Bratland Å, Harman G, Fodstad Ø, Andersson Y
3 Brown JM and Attardi LD: The role of apoptosis in cancer and Ree AH: Cell cycle checkpoint signaling involved in histone
development and treatment response. Nat Rev Cancer 5: 231- deacetylase inhibition and radiation-induced cell death. Mol
237, 2005. Cancer Ther 4: 1231-1238, 2005.
4 Zhou BBS and Bartek J: Targeting the checkpoint kinases: 21 Chen J, Marechal V and Levine AJ: Mapping of the p53 and
chemosensitization versus chemoprotection. Nat Rev Cancer 4: mdm-2 interaction domains. Mol Cell Biol 13: 4107-4114,
1-10, 2004. 1993.

1915
ANTICANCER RESEARCH 26: 1909-1916 (2006)

22 Hietanen S, Lain S, Krausz E, Blattner C and Lane DP: 28 Jordan MA, Ojima I, Rosas F, Distefano M, Wilson L, Scambia
Activation of p53 in cervical carcinoma cells by small molecules. G and Ferlini C: Effects of novel taxanes SB-T-1213 and
Proc Natl Acad Sci USA 97: 8501-8506, 2000. IDN5109 on tubulin polymerization and mitosis. Chem Biol 9:
23 Wesierska-Gadek J, Schloffer D, Kotala V and Horky M: 93-101, 2002.
Escape of p53 protein from E6-mediated degradation in HeLa 29 Yildiz F, Perez R and Redpath JL: Paclitaxel exposure time
cells after cisplatin therapy. Int J Cancer 101: 128-136, 2002. determines the nature of the interaction with radiation in HeLa
24 Khosravi R, Maya R, Gottlieb T, Oren M, Shiloh Y and Shkedy cells: the role of apoptosis. Eur J Cancer 36: 1426-1432, 2000.
D: Rapid ATM-dependent phosphorylation of MDM2 precedes 30 Einhorn N, Trope C, Ridderheim M, Boman K, Sorbe B and
p53 accumulation in response to DNA damage. Proc Natl Acad Cavallin-Stahl E: A systematic overview of radiation therapy
Sci USA 96: 14973-14977, 1999. effects in cervical cancer (cervix uteri). Acta Oncol 42: 546-
25 Tian H, Faje AT, Lee SL and Jorgensen TJ: Radiation-induced 556, 2003.
phosphorylation of Chk1 at S345 is associated with p53- 31 Loizzi V, Cormio G, Loverro G, Selvaggi L, Disaia PJ and
dependent cell cycle arrest pathways. Neoplasia 4: 171-180, 2002. Cappuccini F: Chemoradiation: a new approach for the
26 Koniaras K, Cuddihy AR, Christopoulos H, Hogg A and treatment of cervical cancer. Int J Gynecol Cancer 13: 580-
O’Connell MJ: Inhibition of Chk1-dependent G2 DNA damage 586, 2003.
checkpoint radiosensitizes p53 mutant human cells. Oncogene
20: 7453-7463, 2001.
27 Jordan MA, Toso RJ, Thrower D and Wilson L: Mechanism of
mitotic block and inhibition of cell proliferation by taxol at low Received January 11, 2006
concentrations. Proc Natl Acad Sci USA 90: 9552-9556, 1993. Accepted March 16, 2006

1916

Anda mungkin juga menyukai