ArticlePDF Available

5-Aza-2′-deoxycytidine Activates the p53/p21Waf1/Cip1 Pathway to Inhibit Cell Proliferation

Authors:

Abstract and Figures

In addition to its demethylating function, 5-aza-2′-deoxycytidine (5-aza-CdR) also plays an important role in inducing cell cycle arrest, differentiation, and cell death. However, the mechanism by which 5-aza-CdR induces antineoplastic activity is not clear. In this study, we found that 5-aza-CdR at limited concentrations (0.01–5 μm) induces inhibition of cell proliferation as well as increased p53/p21Waf1/Cip1 expression in A549 cells (wild-type p53) but not in H1299 (p53-null) and H719 cells (p53 mutant). The p53-dependent p21Waf1/Cip1 expression induced by 5-aza-CdR was not seen in A549 cells transfected with the wild-type human papilloma virus type-16 E6 gene that induces p53 degradation. Furthermore, deletion analysis and site-directed mutagenesis of the p21 promoter reveals that 5-aza-CdR induces p21Waf1/Cip1 expression through two p53 binding sites in the p21 promoter. Finally, 5-aza-CdR-induced p21Waf1/Cip1 expression was dependent on DNA damage but not on DNA demethylation as demonstrated by comet assay and bisulfite sequencing, respectively. Our data provide useful clues for judging the therapeutic efficacy of 5-aza-CdR in the treatment of human cancer cells.
Content may be subject to copyright.
5-Aza-2-deoxycytidine Activates the p53/p21
Waf1/Cip1
Pathway to
Inhibit Cell Proliferation*
Received for publication, October 24, 2003, and in revised form, January 12, 2004
Published, JBC Papers in Press, January 13, 2004, DOI 10.1074/jbc.M311703200
Wei-Guo Zhu‡§, Theresa Hileman, Yang Ke§, Peichang Wang‡, Shaoli Lu‡, Wenrui Duan,
Zunyan Dai**, Tanjun Tong‡, Miguel A. Villalona-Calero, Christoph Plass**,
and Gregory A. Otterson‡‡
From the Department of Biochemistry and Molecular Biology and the §Cancer Research Center, Peking University
Health Science Center, 38 Xueyuan Road, Beijing 100083, China, the Division of Hematology/Oncology, Department of
Internal Medicine, and the **Division of Human Cancer Genetics, Comprehensive Cancer Center, The Ohio State
University, Columbus, Ohio 43210
In addition to its demethylating function, 5-aza-2-de-
oxycytidine (5-aza-CdR) also plays an important role in
inducing cell cycle arrest, differentiation, and cell
death. However, the mechanism by which 5-aza-CdR in-
duces antineoplastic activity is not clear. In this study,
we found that 5-aza-CdR at limited concentrations
(0.01–5
M) induces inhibition of cell proliferation as
well as increased p53/p21
Waf1/Cip1
expression in A549
cells (wild-type p53) but not in H1299 (p53-null) and
H719 cells (p53 mutant). The p53-dependent p21
Waf1/Cip1
expression induced by 5-aza-CdR was not seen in A549
cells transfected with the wild-type human papilloma
virus type-16 E6 gene that induces p53 degradation. Fur-
thermore, deletion analysis and site-directed mutagen-
esis of the p21 promoter reveals that 5-aza-CdR induces
p21
Waf1/Cip1
expression through two p53 binding sites in
the p21 promoter. Finally, 5-aza-CdR-induced p21
Waf1/
Cip1
expression was dependent on DNA damage but not
on DNA demethylation as demonstrated by comet assay
and bisulfite sequencing, respectively. Our data provide
useful clues for judging the therapeutic efficacy of 5-aza-
CdR in the treatment of human cancer cells.
As demethylating agents, 5-aza-cytidine and 5-aza-2-deoxy-
cytidine (5-aza-CdR)
1
have been extensively used for epigenetic
research (1– 4). Both demethylating agents are incorporated
into DNA where they bind DNA methyltransferase (DNMT) in
an irreversible, covalent manner, thus sequestering the en-
zyme and preventing maintenance of the methylation state
(5–7). Consequently, silenced genes induced by hypermethyla-
tion are re-expressed after treatment with these demethylating
agents.
Originally, 5-aza-cytidine and 5-aza-CdR were developed as
anticancer agents (5, 8) and have been shown to have signifi-
cant cytotoxic and antineoplastic activities in many experimen-
tal tumors (9 –12). 5-Aza-CdR is reported to be noncarcinogenic
and incorporates into DNA but not RNA or protein (13, 14).
5-Aza-CdR has been found empirically to have more potent
therapeutic effects than 5-aza-cytidine in cell culture and ani-
mal models of human cancers.
However, 5-aza-CdR-induced cytotoxicity may not be linked
to its demethylating function (3, 15–17). In addition, the ther-
apeutic effects of 5-aza-CdR in the treatment of different hu-
man cancer cells are conflicting. 5-Aza-CdR appears to be ben-
eficial in the treatment of human leukemias (9, 18, 19),
myelodysplastic syndromes (20, 21), and hemoglobinopathies
(22, 23). On the other hand, there has been less positive expe-
rience in the effectiveness of 5-aza-CdR for the treatment of
human solid tumors (10, 24). Therefore, it is possible that one
or more critical factors may be involved in regulating the cel-
lular response to 5-aza-CdR treatment that vary in different
cell types.
p53 is a very important tumor suppressor gene and is re-
ported to be abnormal in more than 50% of human cancers (25).
Chemotherapeutic agents frequently act through the mecha-
nism of DNA damage, and p53 plays an important role in the
induction of cell cycle arrest and apoptosis in response to DNA
damage (26). 5-Aza-CdR has also shown anticancer activity
that may be related to its ability to induce DNA damage (15,
27). Based on the scenario mentioned above, it is hypothesized
that 5-aza-CdR may induce DNA damage, thereby activating
p53, which in turn increases p21
Waf1/Cip1
expression, leading to
the inhibition of cell proliferation.
To confirm the role of p53 in the 5-aza-CdR-induced inhibi-
tion of cell proliferation, human lung cancer cells with different
p53 status were selected as the targets for this study. As an
important downstream target of p53 activation, p21
Waf1/Cip1
plays a critical role in inhibiting cell proliferation; therefore,
p21
Waf1/Cip1
expression upon treatment of 5-aza-CdR in cells
with different p53 status was given special attention in this
study. Present data indicated that 5-aza-CdR is a strong DNA-
damaging agent, and 5-aza-CdR induces inhibition of cell pro-
liferation by activating the p53/p21 pathway.
EXPERIMENTAL PROCEDURES
Cells and Treatments—Human lung cancer cell lines A549, H1299,
and H719 were grown in RPMI 1640 supplemented with 10% fetal
bovine serum (heat-inactivated at 56 °C for 45 min) and penicillin/
streptomycin, in a humidified, 5% CO
2
atmosphere and 37 °C incubator.
These cells were treated with 5-aza-CdR (0.01–20
M; Sigma) for up to
72 h. Fresh medium containing 5-aza-CdR was added every 24 h. The
treated cells then were washed with phosphate-buffered saline, placed
* This work was supported by 973 Plan Grant 2002CB713701 and
National Natural and Scientific Foundation Grant 30371613, by a Pe-
king University 985 Plan grant (to W.-G. Z.), by a Translational Re-
search Grant from the Valvano Foundation (to G. A. O.), and by Grant
P30CA16058 from the NCI, National Institutes of Health (Bethesda,
MD). 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.
To whom correspondence may be addressed. E-mail: zhuweiguo@
bjmu.edu.cn.
‡‡ To whom correspondence may be addressed. Tel.: 614-293-6786;
Fax: 614-293-7529; E-mail: otterson-1@medctr.osu.edu.
1
The abbreviations used are: 5-aza-CdR, 5-aza-2-deoxycytidine;
DNMT, DNA methyltransferase; RLU, relative luciferase activity;
HPV, human papilloma virus; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-
diphenyltetrazolium bromide; CG, cytosine-guanine dinucleotide.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 15, Issue of April 9, pp. 15161–15166, 2004
© 2004 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org 15161
by guest, on December 14, 2010www.jbc.orgDownloaded from
in drug-free medium, and harvested at 24 h after incubation at 37 °C.
Western BlottingProtein expression was detected by Western blot-
ting as previously described with minor modifications (12, 16). Briefly,
the cells were harvested with a scraper and then washed with cold
phosphate-buffered saline once. The cells were then lysed in lysis buffer
(50 mMTris-HCl, 250 mMNaCl, 5 mMEDTA, 50 mMNaF, 0.15% Igepal
CA-630, and 1.5 mMphenylmethylsulfonyl fluoride). Equal amounts of
proteins (100 150
g) were size-fractionated on 9 15% SDS-PAGE.
The antibodies used are anti-p21
Waf1/Cip1
(F-5; Santa Cruz; 1
g/ml),
anti-p53 (DO-1; Oncogene Research Products; 0.5
g/ml), and
-tubulin
(Oncogene Research Products; 0.3
g/ml).
Generation of Stable Clone and TransfectionWild-type human pap-
illoma virus type-16 E6 gene (HPV E6) was a gift from Dr. H. Ding
(Department of Radiology, Ohio State University) (28). The HPV E6
gene was inserted into pCMV-neo, and the pCMV-neo-E6 was trans-
fected into A549 cells by using a transfection kit (Qiagen) according to
the manufacturers instructions. The stable clone of A549-pCMV-
neo-E6 (A549-E6) was maintained in medium containing G418 at
500
g/ml.
Methylation-specific PCR and Methylation DetectionDNA was ex-
tracted and then treated with bisulfite as previously described with
minor modifications (29). Briefly, genomic DNA (1
g) in a volume of 50
l was denatured by NaOH (final concentration, 0.275 M) for 10 min at
42 °C. The denatured DNA was then treated with 10
lof10mM
hydroquinone and 520
lof3Msodium bisulfite at 50 °C overnight. The
primers for p21 were as follows: forward primer, 5-GGG AGG AGG
GAA GTG TTT TT-3, and reverse primer, 5-ACA ACT ACT CAC ACC
TCA ACT-3. The PCR conditions were initiated with a denaturing step
of 95 °C for 10 min, followed by 36 cycles of 96 °C for 30 s, 53 °C for 20 s,
and 72 °C for 20 s and were concluded with 72 °C for 7 min. The PCR
products were purified with a purification kit (Qiaquick) and then
incubated with HhaI (50 °C) for 2 h and TaqI at 65 °C for 2 h, respec-
tively. Digested DNA was then size-fractionated via polyacrylamide gel
electrophoresis to detect the methylation status.
Bisulfite SequencingDNA was treated with bisulfite and purified
for PCR as described above. The PCR products were gel-extracted
(Qiagen) and ligated into a plasmid vector, pCR2.1-TOPO, using the TA
cloning system (Invitrogen). Plasmid-transformed bacteria TOP10 F
were cultured overnight, and the plasmid DNA was isolated (Qiagen).
At least 10 separate clones were chosen for sequence analysis.
Transient Transfection and Measurement of Relative Luciferase Ac-
tivityVectors used for transfection include pWWP-Luc and pWP101-
Luc (30, 31). The human wild-type p21 promoter luciferase fusion
plasmid, pWWP-Luc, was made from a 2.4-kb genomic fragment of p21
promoter containing the transcriptional start site and two p53 binding
sites and then subcloned into the luciferase reporter vector, pGL-3Ba-
sic. pWP101-Luc does not contain any p53 binding site. A549 cells
transfected with the pWWP-Luc or the pWP101-Luc were treated with
5-aza-CdR (0.55
M, 24 h) and then harvested to analyze the relative
luciferase activity.
Site-directed MutagenesisMutant p21 promoter constructs were
generated using a site-directed mutagenesis kit (QuikChange; Strat-
agene, La Jolla, CA). pWWP-Luc (containing two p53 binding sites) was
used as the mutagenesis template. The p53 recognition elements con-
sist of four tandem PuPuPuC (A/T) pentamers (32). The first p53 bind-
ing site, GAACA (33) (2234 to 2230 relative to the translational start
site) was replaced with GAAAC, and the second binding site, AGACT
(33) (1344 to 1340 relative to the translational start site) was re-
placed with AGAAT following the manufacturers directions.
Comet Assay for Detecting DNA Strand BreaksThe comet assay,
also called the single-cell gel electrophoresis, was performed as de-
scribed previously (34). In brief, fully frosted microscopic slides were
covered with 110
l of 0.5% normal melting agarose at 60 °C. The slides
were immediately covered with a coverslip and then kept at 4 °C for 15
min to allow the agarose to solidify. About 10
5
cells of 5-aza-CdR treated
or untreated cells in 40
l of phosphate-buffered saline were mixed with
equal amount (40
l) of 1% lower melting agarose to form a cell sus-
pension. After gently removing the coverslip, the cell suspension was
pipetted onto the first agarose layer, spread using a coverslip, and
maintained at 4 °C for 15 min to allow it to solidify. After removal of the
coverslips, the slides were immersed in fresh prepared cold lysing
solution (2.5 MNaCl, 100 mMNa
2
EDTA, 10 mm Tris, pH 10.0, 1%
sodium sarcosinate) with 1% Triton X-100 for 40 min at 4 °C. The slides
were then placed in a horizontal gel electrophoresis tank filled with
fresh electrophoresis solution (1 mMNa
2
EDTA, 300 mMNaOH, pH
13.0) for 10 min. The slides were then placed in Tris buffer (0.4 MTris,
pH 7.5) for 15 min twice (neutralizing the excess alkali) after electro-
phoresis at 4 °C. The slides were then stained with 75
l of propidium
iodide (5
g/ml) for 30 min.
The slides were examined at 600magnification, and the pictures
were taken under a fluorescence microscope (TCS; Leica, Mannheim,
Germany). To score the percentage of DNA in the tail, the image
FIG.1.5-Aza-CdR-induced cell proliferation inhibition is asso-
ciated with p21
Waf1/Cip1
expression. Cell viability was measured
with MTT when A549, H1299, and H719 cells were treated with 5-aza-
CdR at 0 20
Mfor 72 h (A) and at 1
Mfor different intervals (B). C,
representative Western blots show changes in p21
Waf1/Cip1
expression in
A549 or H1299 cells after treatment with 5-aza-CdR at different con-
centrations.
-Tubulin is presented as a loading control.
Increased Expression of p21
Waf1/Cip1
by 5-Aza-CdR
15162
by guest, on December 14, 2010www.jbc.orgDownloaded from
analysis system was used (Q550CW; Leica). The percentage of comet
tail area (DNA tail area/total DNA area) and the comet tail length (from
the center of the DNA head to the end of the DNA tail) were analyzed
in 50 cells for one slide.
RESULTS
Dose-dependent Inhibition of Cell Proliferation by 5-Aza-
CdRIn this study, human lung cancer cell lines A549 (wild-
type p53), H1299 (p53-null), and H719 (mutant p53) were
treated with 5-aza-CdR at different concentrations for 72 h
(Fig. 1A) and with different intervals of 5-aza-CdR treatment at
1
M, respectively (Fig. 1B). The cell viability was determined
by MTT assay. A dose- and duration-dependent inhibition of
cell proliferation was observed only in A549 cells but not in
H1299 and H719 cells (Fig. 1, Aand B). As shown in Fig. 1A, for
example, cell viability was decreased to 77% of the untreated
control when A549 cells were treated with 5-aza-CdR even at
very low dose (0.078
M) and 51% or 39% of the untreated
control when cells were treated with 5-aza-CdR at 1.25 or 5
M,
respectively. However, 5-aza-CdR was unable to induce an obvi-
ous decrease in cell viability in H1299 cells or H719 cells until the
concentrations were well above 1
M(Fig. 1, Aand B). Cell
viability was decreased in H1299 and H719 cells only when they
were treated at very high concentrations (1020
M).
Inhibition of cell proliferation is a reflection of cell cycle
arrest that is mainly controlled by proteins from the INK4
family and the CIP/KIP family of cyclin-dependent kinase in-
hibitors (35). A549 cells have been reported to have a homozy-
gous deletion of CDKN2a (36). Therefore, we hypothesized that
p21
Waf1/Cip1
expression may be a critical factor for the 5-aza-
CdR-induced inhibition of cell proliferation in A549 cells. Fig.
1Cindicates that increased p21
Waf1/Cip1
expression is a dose-
dependent effect of 5-aza-CdR treatment in A549 cells but not
in H1299 cells. The increased expression of p21
Waf1/Cip1
was
observed in the A549 cells even when treated at a very low
concentration (0.01
M) of 5-aza-CdR. However, 5-aza-CdR
even at a very high concentration (10
M) was unable to induce
increased expression of p21
Waf1/Cip1
in H1299 cells (Fig. 1C).
Because p53 is a mutant in H719 cells (30), increased
p21
Waf1/Cip1
expression upon 5-aza-CdR treatment was simi-
larly not observed in H719 cells in this study (data not shown).
5-Aza-CdR-induced Inhibition of Cell Proliferation Is De-
pendent on p53/p21
Waf1/Cip1
PathwayTo investigate whether
5-aza-CdR-induced p21
Waf1/Cip1
expression is through activa-
tion of p53, a wild-type HPV E6 gene was inserted into pCMV-
neo. After transfection and selection with G418, the pCMV-
neo-E6 stable clone (A549-E6) was established. For testing the
functionality of the stable clone, the A549-E6 cells were irra-
diated with
-rays, and then p53 and p21
Waf1/Cip1
levels were
determined by Western immunoblot analysis. As shown in Fig.
2A, the p53 level in A549-E6 cells was much lower than that in
A549 cells after exposure to 2 or 8 grays of
-rays. Similarly,
p21
Waf1/Cip1
levels in the A549 and the A549-E6 cells after
irradiation showed the same trend as the p53 expression (Fig.
2B). These data suggested that the expression of HPV E6 in the
A549-E6 clone is sufficient to degrade the p53 level in the
transfected A549 cells. Next, A549 and A549-E6 cells were
treated with 5-aza-CdR at different concentrations for 72 h,
and then MTT assay was performed to test cell viability in a
fashion analogous to the experiments demonstrated in Fig. 1A.
As shown in Fig. 2C, cell viability in the A549-E6 was much
higher than that in the A549 cells after treatment with 5-aza-
CdR at different concentrations. These results support the
hypothesis that cell viability after treatment with 5-aza-CdR is
related to wild-type p53 in the A549 cells. To investigate the
reasons for the difference between A549 and A549-E6 cells, p53
and p21
Waf1/Cip1
expression was evaluated by Western im-
munoblot (Fig. 2, DE). Clearly, both p53 levels as well as
p21
Waf1/Cip1
levels were significantly increased in A549 cells
after treatment with 5-aza-CdR (Fig. 2, DE). This was not
seen in the A549-E6 cells, even when they were treated with
5-aza-CdR at higher concentrations (up to 10
M; Fig. 2E).
Although p21
Waf1/Cip1
expression is slightly increased in the
A549-E6 cells upon treatment with 5-aza-CdR, the effect is
very attenuated when compared with the parental A549 cells
(Fig. 2E).
For further confirmation of the critical role of p53 in the
5-aza-CdR-induced p21
Waf1/Cip1
expression, a full-length p21
promoter construct (pWWP-Luc) that included two p53 binding
elements and a truncated p21 promoter (pWP101-Luc) that
lacked any p53 binding site (Fig. 3A) were transiently trans-
fected into A549 cells, and relative luciferase activity (RLU)
was evaluated after treatment with 5-aza-CdR in both trans-
fectants. As shown in Fig. 3B, the RLU in the pWWP-Luc-
transfected A549 cells was much higher than that in the
pWP101-Luc-transfected A549 cells after treatment with
5-aza-CdR. The 5-aza-CdR-induced increase in the RLU was
dose-dependent. The RLU in pWWP-transfected cells, for ex-
ample, increased 3-fold when treated with 5-aza-CdR at 5
M
compared with the same transfectants treated at 0.5
M(Fig.
FIG.2. Changes in cell viability and p21
Waf1/Cip1
and p53 pro-
tein expression in A549 cells or A549-E6cells treated with 5-aza-
CdR, respectively. A549 cells or A549-E6 cells were irradiated with
-rays at 2 or 8 grays, and then Western blot was performed to detect
the changes of p53 (A) and p21
Waf1/Cip
(B). A549 and A549-E6 cells were
also treated with 5-aza-CdR at different concentrations, and then MTT
assay was performed to test cell viability (C), and Western blot was
performed to detect the changes of p53 (D) and p21
Waf1/Cip1
(E).
Increased Expression of p21
Waf1/Cip1
by 5-Aza-CdR 15163
by guest, on December 14, 2010www.jbc.orgDownloaded from
3B). The RLU, however, was not significantly increased in the
pWP101-transfected A549 cells after 5-aza-CdR treatments
(Fig. 3B). Because there is no endogenous p53 in H1299 cells,
the RLU was not increased in the pWWP-transfected H1299
cells after treatment with 5-aza-CdR (Fig. 3C).
To determine which p53 binding site of the p21 promoter was
important for the 5-aza-CdR treatment, pWWP-Luc containing
the mutated first p53 binding site (Mut-1) and second p53
binding site (Mut-2) were constructed (Fig. 3A) and then trans-
fected into A549 cells that were subsequently treated with
5-aza-CdR. As shown in Fig. 3D, the RLUs decreased from 3.4
to 1.42 and 1.51, respectively, after 5-aza-CdR treatment when
the Mut-1 or Mut-2 transfectants were compared with wild-
type pWWP-transfected cells (Fig. 3D). No difference in the
RLU between the Mut-1 and Mut-2 transfected cells was ob-
served after 5-aza-CdR treatment (Fig. 3D). The data suggest
that both p53 binding sites in the p21 promoter are important
in inducing p21
Waf1/Cip1
expression in response to 5-aza-CdR
treatment.
5-Aza-CdR Activates the p53/p21 Pathway through DNA
DamageBecause 5-aza-CdR is a DNA methyltransferase in-
hibitor, it was necessary to rule out the possibility that the p21
promoter is fully or partially methylated in A549 cells. To
detect the methylation status of the p21 promoter, we per-
formed combined bisulfite restriction analysis and bisulfite
sequencing of the p21 promoter from A549 and H1299 cells.
The combined bisulfite restriction analysis confirmed that the
CpG island of the p21 promoter (233 to 2; Fig. 3A) was
unmethylated (Fig. 4). Bisulfite sequencing analysis confirmed
these findings (Table I). Ten separate clones were selected for
the sequencing. The promoter regions of the p21 gene in A549
and H1299 cells were shown to be almost totally unmethylated
in the 24 CGs analyzed by bisulfite sequencing (1 of 240 CGs in
A549 cells and 3 of 240 CGs in H1299 cells are methylated,
respectively; Table I).
To investigate whether 5-aza-CdR plays a direct role in dam-
aging DNA, comet assay was performed as well. A549 cells
were treated with 5-aza-CdR at 0.1, 1, and 5
Mfor 72 h and
then harvested for this assay. As shown in Fig. 5, dose-depend-
ent DNA damage was observed after 5-aza-CdR treatment.
Compared with the untreated control (Fig. 5A), 5-aza-CdR even
at a low concentration (0.1
M) induced DNA damage, as indi-
cated by the presence of a DNA tail (Fig. 5B). Greater DNA tail
area and longer DNA tail length (a distance from DNA head to
the end of DNA tail) show more extensive DNA damage. These
5-aza-CdR-induced DNA-damaging features are observed more
frequently in the cells treated with a higher concentration of
5-aza-CdR than that with a lower concentration of 5-aza-CdR
(Fig. 5, compare Cand Dwith A). The quantitative data of
5-aza-CdR-induced DNA damage as determined by comet as-
say are shown in Table II. For example, the percentage of DNA
tail area relative to the total area and the DNA tail length are
increased 2.6- and 1.4-fold, respectively, when cells were
treated with 5-aza-CdR at 0.1
Mcompared with that in the
untreated cells (Table II). Similarly, in the cells treated with
5-aza-CdR at 5
M, the percentage and the length are increased
FIG.3.Changes in relative luciferase activity in A549 or H1299 cells after 5-aza-CdR treatment. A, a schematic diagram indicating p53
binding sites and CpG island of the p21 promoter. There are two p53 binding sites in the p21 promoter, GAACA (2234 to 2230 relative to
transcriptional start site, first binding site) and AGACT (1344 to 1340 relative to transcriptional start site, second binding site). The p53
binding sites were altered by site-directed mutagenesis, to GAAAC (first binding site) and AGAAT (2
nd
binding site). The region (313 to 552
relative to transcriptional start site) shows a range of CpG island in the p21 promoter (NCBI U24170). The dashed line (233 to 2 relative to
transcriptional start site) shows the area that underwent bisulfite sequencing. Band C, pWWP or truncated p21 promoter, pWP101, was
transfected into A549 (B) or H1299 cells (C). The transfected cells were then treated with 5-aza-CdR at different concentrations for 48 h. D, mutated
pWWP containing the first p53 mutated binding site (Mut-1) or the second p53 mutated binding site (Mut-2) was transfected into A549 cells and
then treated with 5-aza-CdR at 1
Mfor 48 h, respectively. The luciferase activity was normalized for the amount of protein in the cell lysate. All
of the luciferase experiments were carried out at least three times in triplicate.
Increased Expression of p21
Waf1/Cip1
by 5-Aza-CdR
15164
by guest, on December 14, 2010www.jbc.orgDownloaded from
4.2- and 3.8-fold, respectively (Table II). These results suggest
that 5-aza-CdR inhibits cell proliferation by damaging DNA,
which causes activation of the p53/p21
Waf1/Cip1
pathway.
DISCUSSION
Increased expression of p21
Waf1/Cip1
after inhibition of DNA
methyltransferase has been reported by several investigators
(3739). To date, at least two separate mechanisms explain this
effect. The first mechanism involves a demethylating function.
5-Aza-CdR, for example, was reported to bind to DNMT and
inactivate the enzyme (40), inducing a re-expression of
p21
Waf1/Cip1
in cells that are hypermethylated in the promoter
of the p21 gene (30, 41, 42). A second mechanism for enhanced
p21
Waf1/Cip1
expression is independent of DNA methylation.
For instance, a significant increase in p21
Waf1/Cip1
expression
was observed in human cancer T24 cells following treatment
with DNMT antisense oligonucleotides (43) or DNMT antago-
nist (37) and in normal human fibroblasts with 5-aza-CdR (44),
but the promoter region of p21 in these cells is totally unmethy-
lated. These data indicate that inhibition of DNMT itself, un-
related to methylation status, may activate p21
Waf1/Cip1
expres-
sion. Consistent with these reports, in the present study, the
5-aza-CdR-induced p21
Waf1/Cip1
expression in A549 cells is not
associated with DNA methylation because the promoter region
of p21 is almost completely unmethylated (Fig. 4 and Table I).
As one of the principle downstream effectors of p53,
p21
Waf1/Cip1
is a cyclin-dependent kinase inhibitory protein and
plays a role in preventing cyclin E/Cdk2 and cyclin A/Cdk2
kinase from promoting cell cycle progression (45). The ability of
p53 to induce cell cycle arrest in the G
1
phase in response to
DNA damage is largely dependent on p21
Waf1/Cip1
expression
(26). Therefore, the p53/p21
Waf1/Cip1
pathway confers the dam-
aged cells enough time to repair DNA and ensure that the cell
cycle progresses correctly. There is conflicting data as to
whether expression of p21
Waf1/Cip1
in cells after inhibition of
DNMT is dependent on p53 changes. 5-Aza-CdR induced an
elevated p53 level in human colon cancer cells, but p21
Waf1/Cip1
expression is not completely dependent on p53 status. In their
experimental system, Karpf et al. (27) showed that p21
Waf1/Cip1
expression induced by 5-aza-CdR was not only observed in
human colon tumor cells HCT (p53
/
) but also seen in HCT
(p53
/
) cells, although the degree of elevated p21
Waf1/Cip1
is
different in both cell lines. Milutinovic also reported that
DNMT antagonists or antisense oligonucleotides can induce
rapid expression of p21
Waf1/Cip1
in A549 cells, but p53 levels are
not changed (37). In this case, the DNMT antagonist or anti-
sense oligonucleotides are not incorporated into DNA; therefore
direct DNA damage with subsequent enhanced p53 expression
does not occur. However, in the present study, p21
Waf1/Cip1
expression in A549 cells after 5-aza-CdR treatment in limited
doses is totally dependent on p53 expression (Fig. 2, DE). This
p53-dependent p21
Waf1/Cip1
expression is demonstrated by the
following evidence: 1) Elevated p21
Waf1/Cip1
expression after
5-aza-CdR treatment occurred only in A549 cells (with wild-
type p53) but not in H1299 cells (p53-null) (Fig. 1C); 2) 5-aza-
CdR-induced p21
Waf1/Cip1
expression in A549 cells is much de-
creased when p53 levels were diminished by transfection with
an HPV E6 gene that promotes p53 degradation (Fig. 2, DE);
and 3) deletion and mutation analysis showed increased rela-
tive luciferase activities after 5-aza-CdR treatment in cells
with transfected full-length p21 promoter (with intact p53
binding sites) but not in those with a truncated p21 promoter
(without p53 binding sites) or full-length p21 promoter (with
mutant p53 sites) (Fig. 3, Band D). Therefore, it is reasonable
to hypothesize that the antineoplastic effect of 5-aza-CdR
TABLE I
Methylation status in A549 or H1299 cells measured
by bisulfite sequencing
Cell Methylated CG Unmethylated CG Methylation
%
A549 1 239 0.42
H1299 3 237 1.25
TABLE II
Comparison of degree of DNA damage after 5-aza-CdR treatment
using the comet assay in A549 cells
Treatment Tail area/total area DNA tail length
%
0
M17 9 18.33 2.1
0.1
M44 21 25.38 3.5
1
M58.3 1 60.07 10.2
5
M70.6 3 70.2 15
FIG.4. Methylation status analysis of the p21promoter as-
sayed by combined bisulfite restriction analysis. DNA extracted
from A549 or H1299 cells was treated with bisulfite, and then methy-
lation specific PCR was performed. Within the sequence of the PCR
product there is one TaqI site (T) and two HhaI sites (H). The PCR
product was incubated with HhaI and TaqI, respectively, for appropri-
ate times and then loaded into a polyacrylamide gel to detect methyl-
ation pattern. DNA was treated with CpG methylase and then treated
with bisulfite as a positive control. Non-bisulfite-treated DNA was used
as a negative control.
FIG.5.Detection of 5-aza-CdR DNA damage by COMET assay.
DNA damage is characterized by the percentage of DNA tail area/whole
DNA area (%) and the comet tail length (from the center of DNA head
to the end of the DNA tail) in A549 cells. The bigger the DNA tail area
(%) or the longer the DNA tail length, the more significant the damage.
A, control, the DNA is intact without DNA tail. B, 5-aza-CdR, 0.1
M.C,
5-aza-CdR, 1
M.D, 5-aza-CdR, 5
M.
Increased Expression of p21
Waf1/Cip1
by 5-Aza-CdR 15165
by guest, on December 14, 2010www.jbc.orgDownloaded from
may activate p53 and subsequently induce an increase in
p21
Waf1/Cip1
expression through which cell proliferation is
inhibited.
Increases in p53/p21
Waf1/Cip1
levels in mammalian cells are
often observed when cells are exposed to DNA-damaging
agents including irradiation (46, 47), UV light (48, 49), and
chemicals (50). Upon exposure to these DNA-damaging ele-
ments, p53 is activated, taking part in post-translational mod-
ifications including phosphorylation and acetylation (26, 51,
52). Previously, 5-aza-CdR was reported to be a potential DNA-
damaging agent in embryonic cells, and its cytotoxicity was
related to DNMT itself and not the secondary demethylation of
genomic DNA (15). 5-Aza-CdR was incorporated into DNA, and
then the spontaneous degradation of the incorporated analog
may result in DNA damage (11, 53). It has also been shown
that 5-aza-CdR, when incorporated into DNA, covalently links
with DNMT, an event that could cause DNA damage because of
the structural instability at its incorporation sites (54) or by
obstructing DNA synthesis (15, 55). Consistent with these re-
ports, our data for the first time demonstrates directly that
5-aza-CdR is a DNA-damaging agent when assayed by comet
assay (Fig. 5 and Table II).
Interestingly, growth inhibition and cytotoxicity induced by
5-aza-CdR at higher doses (20
M) are not dependent on p53
status because the cells tested are mostly dead after 5-aza-CdR
treatment (data not shown). This p53-dependent inhibition of
cell proliferation by a limited dose of 5-aza-CdR treatment has
a great potential advantage in the treatment of different hu-
man cancers. The major reported side effect of 5-aza-CdR when
pushed to the maximum tolerated dose is hematopoietic toxic-
ity (10). A more limited dose of 5-aza-CdR treatment may
decrease these toxic effects on normal tissue and cells. In ad-
dition, the limited dose of 5-aza-CdR required to induce effects
in wild-type p53 cells may explain why different human cancer
cells appear to have different sensitivities to 5-aza-CdR treat-
ment. p53 status may be a critical factor for judging the antin-
eoplastic effects of 5-aza-CdR in cancer cells. Although there is
insufficient data to show a relationship between p53 status and
efficacy of 5-aza-CdR in patients, several studies suggest that
p53 status may be a key predictive factor for the efficacy of
5-aza-CdR treatments (10, 24, 56). For example, 5-aza-CdR
appears to be very effective in the treatment of adult human
chronic myelogenous leukemias, which generally have wild-
type p53 (56, 57) but is less demonstrably effective in the
treatment of solid tumors, such as human lung cancers, which
typically have mutant p53 (24, 58). Our data suggest that the
future clinical development of 5-aza-CdR may depend on ge-
netic factors such as the p53 status of the treated tumor.
AcknowledgmentsWe thank Drs. T. Sakai and H. Ding for provid-
ing us with the vectors used in this study. We also thank Dr. Y. Shang
and Quanhui Zhen, Shan Chen, and Zhe Li for technical help. We
appreciate Dr. X. B. Yao (University of Science and Technology of
China) for support and encouragement during the course of this study.
REFERENCES
1. Fruhwald, M. C., and Plass, C. (2002) Mol. Genet Metab. 75, 116
2. Christman, J. K. (2002) Oncogene 21, 54835495
3. Karpf, A. R., and Jones, D. A. (2002) Oncogene 21, 54965503
4. Baylin, S. B., Herman, J. G., Graff, J. R., Vertino, P. M., and Issa, J. P. (1998)
Adv. Cancer Res. 72, 141196
5. Haaf, T. (1995) Pharmacol. Ther. 65, 1946
6. Jones, P. A., and Taylor, S. M. (1980) Cell 20, 8593
7. Santi, D. V., Norment, A., and Garrett, C. E. (1984) Proc. Natl. Acad. Sci.
U. S. A. 81, 69936997
8. Li, L. H., Olin, E. J., Buskirk, H. H., and Reineke, L. M. (1970) Cancer Res. 30,
2760 2769
9. Momparler, R. L., Cote, S., and Eliopoulos, N. (1997) Leukemia 11, 175180
10. Momparler, R. L., and Bovenzi, V. (2000) J. Cell. Physiol. 183, 145154
11. Covey, J. M., DIncalci, M., Tilchen, E. J., Zaharko, D. S., and Kohn, K. W.
(1986) Cancer Res. 46, 55115517
12. Zhu, W. G., Lakshmanan, R. R., Beal, M. D., and Otterson, G. A. (2001) Cancer
Res. 61, 13271333
13. Glazer, R. I., and Knode, M. C. (1984) Mol. Pharmacol. 26, 381387
14. Vesely, J., and Cihak, A. (1977) Cancer Res. 37, 36843689
15. Juttermann, R., Li, E., and Jaenisch, R. (1994) Proc. Natl. Acad. Sci. U. S. A.
91, 1179711801
16. Zhu, W. G., Dai, Z., Ding, H., Srinivasan, K., Hall, J., Duan, W., Villalona-
Calero, M. A., Plass, C., and Otterson, G. A. (2001) Oncogene 20, 77877796
17. Soengas, M. S., Capodieci, P., Polsky, D., Mora, J., Esteller, M., Opitz-Araya,
X., McCombie, R., Herman, J. G., Gerald, W. L., Lazebnik, Y. A., Cordon-
Cardo, C., and Lowe, S. W. (2001) Nature 409, 207211
18. Lubbert, M. (2000) Curr. Top. Microbiol. Immunol. 249, 135164
19. Wilson, V. L., Jones, P. A., and Momparler, R. L. (1983) Cancer Res. 43,
34933496
20. Wijermans, P. (2000) J. Clin. Oncol. 18, 956962
21. Daskalakis, M., Nguyen, T. T., Nguyen, C., Guldberg, P., Kohler, G., Wijer-
mans, P., Jones, P. A., and Lubbert, M. (2002) Blood 100, 29572964
22. Koshy, M., Dorn, L., Bressler, L., Molokie, R., Lavelle, D., Talischy, N., Hoff-
man, R., van Overveld, W., and DeSimone, J. (2000) Blood 96, 2379 2384
23. DeSimone, J., Koshy, M., Dorn, L., Lavelle, D., Bressler, L., Molokie, R., and
Talischy, N. (2002) Blood 99, 39053908
24. Goffin, J., and Eisenhauer, E. (2002) Ann. Oncol. 13, 16991716
25. van Oijen, M. G., and Slootweg, P. J. (2000) Clin. Cancer Res. 6, 21382145
26. Lakin, N. D., and Jackson, S. P. (1999) Oncogene 18, 76447655
27. Karpf, A. R., Moore, B. C., Ririe, T. O., and Jones, D. A. (2001) Mol. Pharmacol.
59, 751757
28. Ding, H., Duan, W., Zhu, W. G., Ju, R., Srinivasan, K., Otterson, G. A., and
Villalona-Calero, M. A. (2003) Biochem. Biophys. Res. Commun. 305,
950 956
29. Herman, J. G., Graff, J. R., Myohanen, S., Nelkin, B. D., and Baylin, S. B.
(1996) Proc. Natl. Acad. Sci. U. S. A. 93, 98219826
30. Zhu, W. G., Srinivasan, K., Dai, Z., Duan, W., Druhan, L. J., Ding, H., Yee, L.,
Villalona-Calero, M. A., Plass, C., and Otterson, G. A. (2003) Mol. Cell. Biol.
23, 4056 4065
31. Nakano, K., Mizuno, T., Sowa, Y., Orita, T., Yoshino, T., Okuyama, Y., Fujita,
T., Ohtani-Fujita, N., Matsukawa, Y., Tokino, T., Yamagishi, H., Oka, T.,
Nomura, H., and Sakai, T. (1997) J. Biol. Chem. 272, 22199 22206
32. el-Deiry, W. S., Kern, S. E., Pietenpol, J. A., Kinzler, K. W., and Vogelstein, B.
(1992) Nat. Genet. 1, 4549
33. el-Deiry, W. S., Tokino, T., Waldman, T., Oliner, J. D., Velculescu, V. E.,
Burrell, M., Hill, D. E., Healy, E., Rees, J. L., and Hamilton, S. R. (1995)
Cancer Res. 55, 2910 2919
34. Anderson, D., Yu, T. W., Phillips, B. J., and Schmezer, P. (1994) Mutat. Res.
307, 261271
35. Sherr, C. J., and McCormick, F. (2002) Cancer Cell 2, 103112
36. Fukuoka, K., Adachi, J., Nishio, K., Arioka, H., Kurokawa, H., Fukumoto, H.,
Ishida, T., Nomoto, T., Yokote, H., Tomonari, A., Narita, N., Yokota, J., and
Saijo, N. (1997) Jpn. J. Cancer Res. 88, 1009 1016
37. Milutinovic, S., Knox, J. D., and Szyf, M. (2000) J. Biol. Chem. 275, 63536359
38. MacLeod, A. R., and Szyf, M. (1995) J. Biol. Chem. 270, 80378043
39. Ramchandani, S., MacLeod, A. R., Pinard, M., von Hofe, E., and Szyf, M. (1997)
Proc. Natl. Acad. Sci. U. S. A. 94, 684689
40. Ferguson, A. T., Vertino, P. M., Spitzner, J. R., Baylin, S. B., Muller, M. T., and
Davidson, N. E. (1997) J. Biol. Chem. 272, 32260 32266
41. Zhu, W. G., and Otterson, G. A. (2003) Curr. Med. Chem. Anti-Canc. Agents 3,
187199
42. Allan, L. A., Duhig, T., Read, M., and Fried, M. (2000) Mol. Cell. Biol. 20,
12911298
43. Fournel, M., Sapieha, P., Beaulieu, N., Besterman, J. M., and MacLeod, A. R.
(1999) J. Biol. Chem. 274, 24250 24256
44. Young, J. I., and Smith, J. R. (2001) J. Biol. Chem. 276, 1961019616
45. Ko, L. J., and Prives, C. (1996) Genes Dev. 10, 10541072
46. Siliciano, J. D., Canman, C. E., Taya, Y., Sakaguchi, K., Appella, E., and
Kastan, M. B. (1997) Genes Dev. 11, 34713481
47. Shieh, S. Y., Taya, Y., and Prives, C. (1999) EMBO J. 18, 18151823
48. Blagosklonny, M. V., Demidenko, Z. N., and Fojo, T. (2002) Cell Cycle 1, 6774
49. Shieh, S. Y., Ikeda, M., Taya, Y., and Prives, C. (1997) Cell 91, 325334
50. Bunz, F., Hwang, P. M., Torrance, C., Waldman, T., Zhang, Y., Dillehay, L.,
Williams, J., Lengauer, C., Kinzler, K. W., and Vogelstein, B. (1999) J. Clin.
Invest. 104, 263269
51. Wang, Y., and Prives, C. (1995) Nature 376, 8891
52. Wang, Y., and Eckhart, W. (1992) Proc. Natl. Acad. Sci. U. S. A. 89, 42314235
53. DIncalci, M., Covey, J. M., Zaharko, D. S., and Kohn, K. W. (1985) Cancer Res.
45, 31973202
54. Lin, K. T., Momparler, R. L., and Rivard, G. E. (1981) J. Pharm. Sci. 70,
1228 1232
55. Santi, D. V., Garrett, C. E., and Barr, P. J. (1983) Cell 33, 910
56. Sacchi, S., Kantarjian, H. M., OBrien, S., Cortes, J., Rios, M. B., Giles, F. J.,
Beran, M., Koller, C. A., Keating, M. J., and Talpaz, M. (1999) Cancer 86,
26322641
57. Peller, S., Yona, R., Kopilova, Y., Prokocimer, M., Goldfinger, N., Uysal, A.,
Karabulut, H. G., Tukun, A., Bokesoy, I., Tuncman, G., and Rotter, V.
(1998) Genes Chromosomes Cancer 21, 27
58. Ahrendt, S. A., Hu, Y., Buta, M., McDermott, M. P., Benoit, N., Yang, S. C.,
Wu, L., and Sidransky, D. (2003) J. Natl. Cancer Inst. 95, 961970
Increased Expression of p21
Waf1/Cip1
by 5-Aza-CdR
15166
by guest, on December 14, 2010www.jbc.orgDownloaded from
... Abnormal DNA methylation (hypermethylation and hypomethylation) is the most characteristic epigenetic change of this disease, and has been associated with the initiation and development of prostate cancer [21][22][23]. Numerous studies reported that 5-Aza exerted antitumor activity by activating p53/p21, and a few related mechanisms have been investigated in prostate cancer [24][25][26][27]. However, some conflicting results were also reported. ...
... 5-aza-CdR-induced p53-mediated p21 Waf1/Cip1 expression was dependent on either DNA damage or DNA demethylation [26,27]. However, down regulation of both p53 and p21 were usually observed in malignant tissue, including prostate cancer tissue/cells (supplementary data Figure 1) [53]. ...
Article
Full-text available
DNA methylation is a DNA methyltransferase-mediated epigenetic modification affecting gene expression. This process is involved in the initiation and development of malignant disease. 5-Aza-2'-deoxycytidine (5-Aza), a classic DNA methyltransferase inhibitor, possesses antitumor proliferation activity. However, whether 5-Aza induces cytotoxicity in solid tumors warrants further investigated. In this study, human prostate cancer (CaP) cells were treated with 5-Aza and subjected to cell viability and cytotoxicity analysis. Reverse transcription-polymerase chain reaction and methylation-specific polymerase chain reaction assay were utilized to test the gene expression and methylation status of the p53 and p21 gene promoters. The results showed that 5-Aza differentially inhibited spontaneous proliferation, arrested the cell cycle at S phase in DU145, at G1 phase in 22RV1 and LNCaP cells, and G2 phase in normal RWPE-1 cells, as well as induced the expression of phospho-H2A.X and tumor suppressive mammary serine protease inhibitor (maspin) in all three types of CaP cells. 5-Aza also increased p53 and p21 transcription through promoter demethylation, and decreased the expression of oncogene c-Myc in 22RV1 and LNCaP cells. Western blotting analysis showed that the poly (ADP-ribose) polymerase cleavage was detected in DU145 and 22RV1 cells. Moreover, there were no significant changes in p53, p21 and c-Myc expression in DU145 cells following treatment with 5-Aza. Thus, in responsible for its apoptotic induction and DNA damage, the mechanism of the antitumor activities of 5-Aza may involve in an increase of tumor suppressive maspin, upregulation of wild type p53-mediated p21 expression and a decrease of oncogene c-Myc level in 22RV1 and LNCaP cells, and enhancing the tumor suppressive maspin expression in DU145 cells. These results enriched our understanding of the multifaceted antitumor activity of 5-Aza, and provided the expression basis of biomarkers for its possible clinical application in prostate cancer.
... Treatment with DNA demethylating agent 5-aza-2ˈ-de-oxycytidine (5-Aza-CdR) has been indicated that affect genomic methylation and resulting in silenced genes reactivation in colon cancer [7,8]. In addition to colon cancer, it has been reported that 5-aza-CdR increases p53/ p21Waf1/Cip1 expression in lung cancer and prostate cancer [9,10], p27kip1 in esophageal squamous cell carcinoma [11], and p57KIP2 in lung and breast cancers [12]. Previously, we evaluated the effect of 5-Aza-CdR on DNMT1 gene expression in hepatocellular carcinoma (HCC) which encouraged us to design the current study [13]. ...
Article
Full-text available
Background: Dysregulation of the cell cycle has been reported in various cancers. Inactivation of the cyclin-dependent kinases inhibitors (CDKIs), CIP/KIP family, such as p21Cip1/Waf1/Sdi1, p27Kip1, and p57Kip2 genes because of hypermethylation has been shown in several cancers. Treatment with DNA demethylating agent 5-aza-2ˈ-deoxycytidine (5-Aza-CdR) has been indicated that affect genomic methylation and resulting in silenced genes reactivation in colon cancer. Previously, we evaluated the effect of 5-Aza-CdR on DNA methyltransferase 1 (DNMT1) gene expression in hepatocellular carcinoma (HCC) which encouraged us to design the current study. The present study aimed to evaluate the effect of 5-Aza-CdR on p21Cip1/Waf1/Sdi1, p27Kip1, p57Kip2, and DNAT1 genes expression, cell growth inhibition and apoptosis induction in colon cancer SW 480 and SW 948 cell lines. Materials and Methods: The effect of 5-aza-CdR on the SW 480 and SW 948 cells growth, apoptosis induction and genes expression were assessed by MTT assay, flow cytometry, and real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis respectively. Results: 5-aza-CdR inhibited cell growth as time- and dose-dependent manner significantly (P<0.001). The agent reactivated p15INK4, p16INK4, p18INK4, and p19INK4 genes expression and induced apoptosis at a concentration of 5 μM significantly. Besides, 5-aza-CdR had a more significant effect on the SW 480 cell line in comparison to SW 948 cell line. Conclusion: 5-Aza-CdR plays a key role in the up-regulation of p21Cip1/Waf1/Sdi1, p27Kip1, and p57Kip2 and down-regulation of DNMT1 genes resulting in cell growth inhibition and apoptosis induction. [GMJ.2020;9:e1899]
... 5-aza-2'-deoxycytidine (5-aza) is a typical agent to activate methylated and silenced genes by promoter demethylation. The anti-tumor effect of 5-aza in different types of human cancer cell lines, including HCC, has extensively been confirmed [15][16][17]. In the present study, we found that ID1 expression is controlled by promoter methylation based on the results from methylation specific PCR experiment. ...
Article
Full-text available
Sorafenib resistance greatly restricts its clinical application in patients with hepatocellular carcinoma (HCC). Numerous studies have reported that ID1 exerts a crucial effect in cancer initiation and development. Our previous research revealed an inhibitory role of ID1 in sorafenib resistance. However, the upstream regulatory mechanism of ID1 expression is unclear. Here, we discovered that ID1 expression is negatively correlated with promoter methylation, which is regulated by DNMT3B. Knock down of DNMT3B significantly inhibited ID1 methylation status, and resulted in an increase of ID1 expression. The de‐methylating agent 5‐aza‐2'‐deoxycytidine (5‐aza) remarkably up‐regulated ID1 expression. The combination of 5‐aza with sorafenib showed a synergistic effect on the inhibition of cell viability.
... Total proteins were extracted, quantified, and then analyzed by performing the SDS-PAGE and immunoblotting as we previously described (Zhu et al., 2004). Equal amounts of proteins (20-60 µg) from each sample were subjected to SDS-PAGE, and then transferred to nitrocellulose membranes. ...
Article
Full-text available
Disordered choline metabolism is associated with tumor progression. Glycerophosphocholine phosphodiesterase 1 (GPCPD1) is critical for cleaving glycerophosphocholine (GPC) to produce choline. However, whether and how GPCPD1 is epigenetically regulated remains largely unknown. In the current study, we report that histone H3 lysine 9 (H3K9) methyltransferase GLP (G9a-like Protein) is essential for transcriptional activation of GPCPD1 through H3K9me1 to promote tumor cell migration and invasion. Knocking down GLP or inhibiting its methyltransferase activity impaired GPCPD1 expression and decreased the choline levels. Importantly, we confirmed that both GPCPD1 and choline levels are positively correlated with cancer cell migration. The reduced migration and invasion of GPCPD1-knockdown cells were rescued by choline treatment. Interestingly, GPCPD1 gene expression was found regulated by transcription factor Krüppel-like Factor 5 (KLF5). KLF5 recruitment was GLP-dependent and was indispensable for GPC-induced GPCPD1 expression. These data suggest that GLP promotes tumor cell migration and invasion by transcriptionally activating GPCPD1 . GLP and KLF5 are potential therapeutic targets in future cancer treatment.
... We also analyzed the gene and protein expression levels of the cell cycle regulating protein p21 Cip1/Waf1 (p21) as well as 14-3-3 σ (stratifin, SFN), to evaluate the effects of the DNMT inhibition. The promoter of the SFN gene encoding the 14-3-3 σ protein is hypermethylated in melanocytes and melanoma cells, and p21 expression can be upregulated by 5-Aza treatment in melanoma cells, as shown previously [25][26][27]. We observed a strong induction of SFN gene expression and a moderate induction of the CDKN1A gene encoding p21 upon 5-Aza treatment in almost all tested cells (Supplementary Figure S1B,C). ...
Article
Full-text available
The neural crest transcription factor BRN3A is essential for the proliferation and survival of melanoma cells. It is frequently expressed in melanoma but not in normal melanocytes or benign nevi. The mechanisms underlying the aberrant expression of BRN3A are unknown. Here, we investigated the epigenetic regulation of BRN3A in melanocytes and melanoma cell lines treated with DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC) inhibitors. DNMT and HAT inhibition did not significantly alter BRN3A expression levels, whereas panHDAC inhibition by trichostatin A led to increased expression. Treatment with the isoform-specific HDAC inhibitor mocetinostat, but not with PCI-34051, also increased BRN3A expression levels, suggesting that class I HDACs HDAC1, HDAC2, and HDAC3, and class IV HDAC11, were involved in the regulation of BRN3A expression. Transient silencing of HDACs 1, 2, 3, and 11 by siRNAs revealed that, specifically, HDAC2 inhibition was able to increase BRN3A expression. ChIP-Seq analysis uncovered that HDAC2 inhibition specifically increased H3K27ac levels at a distal enhancer region of the BRN3A gene. Altogether, our data suggest that HDAC2 is a key epigenetic regulator of BRN3A in melanocytes and melanoma cells. These results highlight the importance of epigenetic mechanisms in regulating melanoma oncogenes.
... 273 5-aza compounds at high concentrations are cytotoxic and cause cell cycle arrest (Zhu et al. 2004). We asked 274 whether shifts in AI in the target genes might be due to nonspecific cytotoxicity. ...
Article
Full-text available
In mammalian cells, maternal and paternal alleles usually have similar transcriptional activity. Epigenetic mechanisms such as X-chromosome inactivation (XCI) and imprinting were historically viewed as rare exceptions to this rule. Discovery of autosomal monoallelic expression (MAE) a decade ago revealed an additional allele-specific mode regulating thousands of mammalian genes. Despite MAE prevalence, its mechanistic basis remains unknown. Using an RNA sequencing-based screen for reactivation of silenced alleles, we identified DNA methylation as key mechanism of MAE mitotic maintenance. In contrast with the all-or-nothing allelic choice in XCI, allele-specific expression in MAE loci is tunable, with exact allelic imbalance dependent on the extent of DNA methylation. In a subset of MAE genes, allelic imbalance was insensitive to DNA demethylation, implicating additional mechanisms in MAE maintenance in these loci. Our findings identify a key mechanism of MAE maintenance and provide basis for understanding the biological role of MAE.
... p53 is an important tumor suppressor gene, and its upregulation may increase TKIs sensitivity in CML [34]. DAC and Ara-C are predominantly incorporated into DNA, thereby inducing DNA damage and p53 activation, ultimately leading to apoptosis [35,36]. Because OR21 is a prodrug of DAC, it also induced apoptosis in p53-wild-type cells, as did DAC. ...
Article
ABL1 tyrosine kinase inhibitors (TKIs) dramatically improve the prognosis of chronic myeloid leukemia (CML), but 10–20% of patients achieve suboptimal responses with low TKIs sensitivity. Furthermore, residual leukemic stem cells (LSCs) are involved in the molecular relapse after TKIs discontinuation. Aberrant DNA hypermethylation contributes to low TKIs sensitivity and the persistence of LSCs in CML. DNMT1 is a key regulator of hematopoietic stem cells, suggesting that aberrant DNA hypermethylation targeting DNMT1 represents a potential therapeutic target for CML. We investigated the efficacy of OR-2100 (OR21), the first orally available single-compound prodrug of decitabine. OR21 exhibited anti-tumor effects as a monotherapy, and in combination therapy it increased TKI-induced apoptosis and induction of tumor suppressor genes including PTPN6 encoding SHP-1 in CML cells. OR21 in combination with imatinib significantly suppressed tumor growth in a xenotransplant model. OR21 and combination therapy decreased the abundance of LSCs and inhibited engraftment in a BCR-ABL1–transduced mouse model. These results demonstrate that targeting DNMT1 using OR21 exerts anti-tumor effects and impairs LSCs in CML. Therefore, combination treatment of TKIs and OR21 represents a promising treatment strategy in CML.
Article
Background 5-Aza-2′-deoxycytidine (5-Aza-CdR) is a demethylating agent that has various biological effects related to DNA methylation. DNA methylation plays important roles in learning and memory. We have reported that 5-Aza-CdR improved the performance of mice in the water maze and step-down tests. Some behaviours have been well recognized to be mediated by neurogenesis in the hippocampus. The Notch signalling pathway plays a key role in adult hippocampal neurogenesis. In this study, we examined whether 5-Aza-CdR (DNA methyltransferase inhibitor) affects neurogenesis and Notch1 expression. Methods The learning and memory behaviour of mice was evaluated by a conditioned avoidance learning 24 h after 5-Aza-CdR treatment. The mRNA and protein expression levels of Notch1 and HES1 were measured by real-time PCR and Western blotting. The 5-bromo-2′-deoxyuridine (BrdU)-positive cells and the expression of Notch1 in the hippocampal DG were observed through laser confocal microscopy. To further clarify whether 5-Aza-CdR affects behaviour through neurogenesis, the expression level of Notch1, cell viability and cell cycle were analysed using the HT22 cell line. Results The behaviour in conditioned avoidance learning was improved, while neurogenesis and the Notch1 pathway were increased in the hippocampus of mice that were injected with 5-Aza-CdR. In vitro experiments showed that 5-Aza-CdR increased the expression of the Notch1 pathway and upregulated S-phase in the cell cycle and cell viability. Conclusions Our results suggest that the effect of 5-Aza-CdR on behaviour may be related to an increase in neurogenesis with upregulation of the Notch1 pathway in the hippocampus.
Article
Failure of hypomethylation agent (HMA) treatments is an important issue in myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). Recent studies indicated that function of wildtype TP53 positively impacts outcome of HMA treatments. We investigated the combination of the HMA azacitidine (AZA) with DS-3032b and DS-5272, novel antagonists of the TP53 negative regulator MDM2, in cellular and animal models of MDS and CMML. In TP53 wildtype myeloid cell line, combinational effects of DS-3032b or DS-5272 with AZA were observed. In Tet2-knockout mouse model of MDS and CMML, DS-5272 and AZA combination ameliorated disease-like phenotype. RNA-Seq analysis in mouse bone marrow hematopoietic stem and progenitors indicated that DS-5272 and AZA combination caused down-regulation of leukemia stem cell marker genes and activation of pathways of TP53 function and stability. These findings demonstrate that combining an MDM2 antagonist with AZA has potential to improve AZA treatment in TP53 wildtype MDS and CMML.
Article
The DNA hypomethylating agents (HMA) azacitidine (AZA) and decitabine (DAC) improve survival and transfusion independence in myelodysplastic syndrome (MDS) and enable a low intensity cytotoxic treatment for aged AML patients unsuitable for intensive chemotherapy, particularly in combination with novel agents. The proposed mechanism of AZA and DAC relies on active DNA replication and therefore patient responses are only observed after multiple cycles of treatment. Although extended dosing may provide the optimal scheduling, the reliance of injectable formulation of the drug limits it to intermittent treatment. Recently, an oral formulation of AZA demonstrated significantly improved patient relapse free survival (RFS) and overall survival (OS) when used as maintenance after chemotherapy for AML. In addition, both DAC and AZA were found to be highly effective to improve survival in elderly patients with AML through combination with other drugs. These recent exciting results have changed the therapeutic paradigm for elderly patients with AML. In light of this, we review current knowledge on HMA mechanism of action, clinical trials exploring dosing and scheduling, and recent HMA combination therapies to enhance efficacy.
Article
Full-text available
Previous lines of evidence have shown that inhibition of DNA methyltransferase (MeTase) can arrest tumor cell growth; however, the mechanisms involved were not clear. In this manuscript we show that out of 16 known tumor suppressors and cell cycle regulators, the cyclin-dependent kinase inhibitor p21 is the only tumor suppressor induced in the human lung cancer cell line, A549, following inhibition of DNA MeTase by a novel DNA MeTase antagonist or antisense oligonucleotides. The rapid induction of p21 expression points to a mechanism that does not involve demethylation of p21 promoter. Consistent with this hypothesis, we show that part of the CpG island upstream of the endogenous p21 gene is unmethylated and that the expression of unmethylated p21 promoter luciferase reporter constructs is induced following inhibition of DNA MeTase. These results are consistent with the hypothesis that the level of DNA MeTase in a cell can control the expression of a nodal tumor suppressor by a mechanism that does not involve DNA methylation.
Article
The methylation of DNA is an epigenetic modification that can play an important role in the control of gene expression in mammalian cells. The enzyme involved in this process is DNA methyltransferase, which catalyzes the transfer of a methyl group from S‐adenosyl‐methionine to cytosine residues to form 5‐methylcytosine, a modified base that is found mostly at CpG sites in the genome. The presence of methylated CpG islands in the promoter region of genes can suppress their expression. This process may be due to the presence of 5‐methylcytosine that apparently interferes with the binding of transcription factors or other DNA‐binding proteins to block transcription. In different types of tumors, aberrant or accidental methylation of CpG islands in the promoter region has been observed for many cancer‐related genes resulting in the silencing of their expression. How this aberrant hypermethylation takes place is not known. The genes involved include tumor suppressor genes, genes that suppress metastasis and angiogenesis, and genes that repair DNA suggesting that epigenetics plays an important role in tumorigenesis. The potent and specific inhibitor of DNA methylation, 5‐aza‐2′‐deoxycytidine (5‐AZA‐CdR) has been demonstrated to reactivate the expression most of these “malignancy” suppressor genes in human tumor cell lines. These genes may be interesting targets for chemotherapy with inhibitors of DNA methylation in patients with cancer and this may help clarify the importance of this epigenetic mechanism in tumorigenesis. J. Cell. Physiol. 183:145–154, 2000. © 2000 Wiley‐Liss, Inc.
Article
Augmentation of the fetal hemoglobin (HbF) levels is of therapeutic benefit in patients with sickle cell anemia. Hydroxyurea (HU), by increasing HbF, lowers rates of pain crisis, episodes of acute chest syndrome, and requirements for blood transfusions. For patients with no HbF elevation after HU treatment, augmentation of HbF levels by 5-aza-2′-deoxycytidine (5-aza-CdR, decitabine) could serve as an alternate mode of treatment. Eight adult patients participated in a dose-escalating phase I/II study with 5-aza-CdR at doses ranging from 0.15 to 0.30 mg/kg given 5 days a week for 2 weeks. HbF, F cell, F/F cell, γ-globin synthesis ratio, complete blood count, and chemistry were measured. The average γ-globin synthesis relative to non-α-globin synthesis prior to therapy was 3.19% ± 1.43% and increased to 13.66% ± 4.35% after treatment. HbF increased from 3.55% ± 2.47% to 13.45% ± 3.69%. F cells increased from 21% ± 14.8% to 55% ± 13.5% and HbF/F cell increased from 17% to 24%. In the HU nonresponders HbF levels increased from 2.28% ± 1.61% to 2.6% ± 2.15% on HU, whereas on 5-aza-CdR HbF increased to 12.70% ± 1.81%. Total hemoglobin increased by 1 g/dL in 6 of 8 patients with only minor reversible toxicities, and all patients tolerated the drug. Maximum HbF was attained within 4 weeks of treatment and persisted for 2 weeks before falling below 90% of the maximum. Therefore 5-aza-CdR could be effective in increasing HbF in patients with sickle cell anemia who failed to increase HbF with HU. Demonstration of sustained F levels with additional treatment cycles without toxicity is currently being performed.
Article
Activation of p53 can occur in response to a number of cellular stresses, including DNA damage, hypoxia and nucleotide deprivation. Several forms of DNA damage have been shown to activate p53, including those generated by ionising radiation (IR), radio-mimetic drugs, ultraviolet light (UV) and chemicals such as methyl methane sulfonate (MMS). Under normal conditions, p53 levels are maintained at a low state by virtue of the extremely short-half life of the polypeptide. In addition to this, p53 normally exists in an largely inactive state that is relatively inefficient at binding to DNA and activating transcription. Activation of p53 in response to DNA damage is associated with a rapid increase in its levels and with an increased ability of p53 to bind DNA and mediate transcriptional activation. This then leads to the activation of a number of genes whose products trigger cell-cycle arrest, apoptosis, or DNA repair. Recent work has suggested that this regulation is brought about largely through DNA damage triggering a series of phosphorylation, de-phosphorylation and acetylation events on the p53 polypeptide. Here, we discuss the nature of these modifications, the enzymes that bring them about, and how changes in p53 modification lead to p53 activation.
Article
BACKGROUND The prognoses of patients with chronic myelogenous leukemia in blastic phase (CML-BP) are extremely poor. Treatment of patients with nonlymphoid CML-BP is associated with very low response rates, a median survival of 2–3 months, and significant toxicities. The aim of this study was to evaluate the results of therapy in CML-BP with different treatments in relation to response rate, survival, and toxicity.METHODSA total of 162 adults patients with a diagnosis of nonlymphoid CML-BP referred from 1986 to 1997 were included in this analysis. Only first salvage therapy was considered for the purpose of this analysis. The blastic phase of CML was defined by the presence of 30% or more blasts in the blood or bone marrow, or extramedullary disease. Ninety patients were treated with intensive chemotherapy, 31 with decitabine, and 41 with other single agents.RESULTSThirty-six patients (22%) had an objective response. Response rates were similar among patients treated with intensive chemotherapy (28%) or with decitabine (26%). In aggregate, other single agents showed objective response rates of 7%. The median duration of remission for all patients was 29 weeks and the median overall survival 22 weeks. Patients treated with decitabine showed a trend toward better survival, despite a higher percentage of older patients (P < 0.004). The median survival times were 29 weeks with decitabine, 21 weeks with intensive chemotherapy, and 22 weeks with other agents. When only older patients were considered, survival was significantly better with decitabine versus other treatments (P < 0.01). A multivariate analysis of prognostic factors for survival confirmed the independent, significant favorable effect of decitabine therapy (P = 0.047). In all groups complications of myelosuppression were the most significant side effects. Severe nonhematologic toxicities were not observed in patients treated with decitabine; they occurred in 20% and 17% of patients treated with intensive chemotherapy or other single agents, respectively.CONCLUSIONS Compared with intensive chemotherapy, decitabine showed favorable results, with similar objective response rates, a better nonhematologic toxicity profile, and a trend for better survival, particularly among older patients. Studies will now attempt to combine decitabine with other promising approaches, such as homoharringtonine, low dose cytarabine, and interferon-α, in all CML phases. Cancer 1999;86:2632–41. © 1999 American Cancer Society.
Article
The methylation of DNA is an epigenetic modification that can play an important role in the control of gene expression in mammalian cells. The enzyme involved in this process is DNA methyltransferase, which catalyzes the transfer of a methyl group from S-adenosyl-methionine to cytosine residues to form 5-methylcytosine, a modified base that is found mostly at CpG sites in the genome. The presence of methylated CpG islands in the promoter region of genes can suppress their expression. This process may be due to the presence of 5-methylcytosine that apparently interferes with the binding of transcription factors or other DNA-binding proteins to block transcription. In different types of tumors, aberrant or accidental methylation of CpG islands in the promoter region has been observed for many cancer-related genes resulting in the silencing of their expression. How this aberrant hypermethylation takes place is not known. The genes involved include tumor suppressor genes, genes that suppress metastasis and angiogenesis, and genes that repair DNA suggesting that epigenetics plays an important role in tumorigenesis. The potent and specific inhibitor of DNA methylation, 5-aza-2′-deoxycytidine (5-AZA-CdR) has been demonstrated to reactivate the expression most of these “malignancy” suppressor genes in human tumor cell lines. These genes may be interesting targets for chemotherapy with inhibitors of DNA methylation in patients with cancer and this may help clarify the importance of this epigenetic mechanism in tumorigenesis. J. Cell. Physiol. 183:145–154, 2000. © 2000 Wiley-Liss, Inc.
Article
The chemical stability of 5-aza-2′-deoxycytidine (I) in acidic, neutral, and alkaline solutions was analyzed by high-performance liquid chromatography. In alkaline solution, I underwent rapid reversible decomposition to N-(formylamidino)-N'β-D-2-deoxyribofuranosylurea (II), which decomposed irreversibly to form 1-β-D-2′-deoxyribofurano-syl-3-guanylurea (III). The pseudo-first-order rate constants for this reaction were determined. The decomposition of I in alkaline solution was identical to that reported previously for the related analog, 5-aza-cytidine. However, in neutral solution (or water), there was a marked difference in the decomposition of I and 5-azacytidine. The same decomposition products were formed from 5-azacytidine in neutral solution as in alkaline solution. However, in neutral solution, I decomposed to II and three unknown compounds that were chromophoric at 254 nm. Compound I was most stable when stored in neutral solution at low temperature.
Article
5-Azacytidine (5-aza-C) analogs demonstrate a remarkable ability to induce heritable changes in gene and phenotypic expression. These cellular processes are associated with the demethylation of specific DNA sequences. On the other hand, 5-aza-C analogs have dramatic effects on chromosomes, leading to decondensation of chromatin structure, chromosomal instability and an advance in replication timing. Condensation inhibition of genetically inactive chromatin occurs when the DNA is still hemimethylated or fully methylated. In cell cultures prolonged for several replication cycles, chromosomal rearrangements and instability affect the 5-aza-C-sensitive regions. Moreover, the normally late-replicating inactive chromatin undergoes a transient temporal shift to an earlier DNA replication, characteristic of activatable chromatin. zThe induced alterations of chromosome structure and behavior may trigger the 5-aza-C-dependent process of cellular reprogramming. Apart from their differentiating and gene-modifying effects, 5-aza-C analogs can tumorigenically transform cells and modulate their metastatic potential. High doses of 5-aza-C analogs have cytotoxic and antineoplastic activities.
Article
5-Aza-2'-deoxycytidine administered at a daily dose of 1.5 mg/kg increased the life-span of P388 leukemia-bearing BALB/c X DBA/2 F1 mice by 5 times and that of second generation lymphoma-bearing AKR mice by 2.5 times. Higher doses (total dose, 20 mg/kg) led to favorable results when administered in two portions on Days 4 and 5 after the s.c. inoculation of leukemic cells. The same total dose given on 5 consecutive days was toxic. The lethal dose that killed 50% of the animals was 190 mg/kg. The drug was also effective in L1210 leukemia. 5-Aza-2'-deoxycytidine inhibited the phosphorylation of 2'-deoxycytidine in the acid-soluble pool of cells from leukemic AKR mice as well as its incorporation into DNA. In vitro the inhibition of the uptake of 2'-deoxycytidine into cells from leukemic mice by 5-aza-2-deoxycytidine had a competitive character (Ki, 8 X 10(-5) M). Although 5-aza-2'-deoxy[4-14C]cytidine of low-specific activity was not detected in DNA isolated from the lives of leukemic mice, the same tritium-labeled drug of high-specific radioactivity was selectively localized in the nuclei of leukemic cells as revealed by autoradiography. The incorporation of [3H]-5-aza-2'-deoxycytidine into DNA of cells from leukemic mice was confirmed by the chromatographic separation of DNA on a column of kieselguhr coated with methylated albumin.
Article
Recent experiments have suggested that p53 action may be mediated through its interaction with DNA. We have now identified 18 human genomic clones that bind to p53 in vitro. Precise mapping of the binding sequences within these clones revealed a consensus binding site with a striking internal symmetry, consisting of two copies of the 10 base pair motif 5'-PuPuPuC(A/T)(T/A)GPyPyPy-3' separated by 0-13 base pairs. One copy of the motif was insufficient for binding, and subtle alterations of the motif, even when present in multiple copies, resulted in loss of affinity for p53. Mutants of p53, representing each of the four "hot spots" frequently altered in human cancers, failed to bind to the consensus dimer. These results define the DNA sequence elements with which p53 interacts in vitro and which may be important for p53 action in vivo.