Aspisol inhibits tumor growth and induces apoptosis in breast cancer
Nonsteroidal anti-inflammatory drugs inhibit cell proliferation and induce apoptosis in various cancer cell lines, which is considered to be an important mechanism for their anti-tumor activity and cancer prevention. However, the molecular mechanisms through which these compounds induce apoptosis ar...
Gespeichert in:
Datum: | 2008 |
---|---|
Hauptverfasser: | , , , , |
Format: | Artikel |
Sprache: | English |
Veröffentlicht: |
Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України
2008
|
Schriftenreihe: | Experimental Oncology |
Schlagworte: | |
Online Zugang: | http://dspace.nbuv.gov.ua/handle/123456789/139930 |
Tags: |
Tag hinzufügen
Keine Tags, Fügen Sie den ersten Tag hinzu!
|
Назва журналу: | Digital Library of Periodicals of National Academy of Sciences of Ukraine |
Zitieren: | Aspisol inhibits tumor growth and induces apoptosis in breast cancer / X.G. Zhu, L. Tao, Z.R. Mei, H.P. Wu, Z.W. Jiang // Experimental Oncology. — 2008. — Т. 30, № 4. — С. 289–294. — Бібліогр.: 25 назв. — англ. |
Institution
Digital Library of Periodicals of National Academy of Sciences of Ukraineid |
irk-123456789-139930 |
---|---|
record_format |
dspace |
spelling |
irk-123456789-1399302018-06-22T03:04:38Z Aspisol inhibits tumor growth and induces apoptosis in breast cancer Zhu, X.G. Tao, L. Mei, Z.R. Wu, H.P. Jiang, Z.W. Original contributions Nonsteroidal anti-inflammatory drugs inhibit cell proliferation and induce apoptosis in various cancer cell lines, which is considered to be an important mechanism for their anti-tumor activity and cancer prevention. However, the molecular mechanisms through which these compounds induce apoptosis are not well understood. Aim: to determine the effects of nonselective cyclooxygenase-2 (COX-2) inhibitor, aspisol on breast cancer cells in vitro and in vivo. Methods: The cytotoxic activity of aspisol was evaluated by MTT assay. The apoptosis index of cells was measured by flow cytometry. Immunohistochemical staining was used to detect expressions of COX-2 and caspase-3 in MDA-MB-231 cells. The expression of bcl-2 and bax was analyzed by Western blot analysis. The content of prostaglandin E2 (PGE2) in MDA-MB-231 cells was estimated by ELISA. In vivo apoptosis of the tumor cells was detected by the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). Results: Our results showed that aspisol reduced viability of MDA-MB-231 cells in time- and dose- dependent fashions and induced apoptosis by increase of caspase-3 and bax expressions while decrease of COX-2 and bcl-2 expression in vitro. In addition, exposure to aspisol decreased the basal release of PGE2. In vivo, aspisol also inhibited the proliferation of breast cancer cells and induced their apoptosis. Conclusions: Our in vitro and in vivo data indicated that the antitumor effects of aspisol on breast cancer cells was probably mediated by the induction of apoptosis, and it could be linked to the downregulation of the COX-2 or bcl-2 expression and up-regulation of caspase-3 or bax expression. Нестероидные противовоспалительные препараты ингибируют пролиферацию клеток и вызывают апоптоз во многих опухолевых клеточных линиях, что считается важным механизмом их противоопухолевой активности и профилактики развития рака. Тем не менее молекулярные механизмы апоптотического действия этих препаратов изучены недостаточно. Цель: изучить действие неспецифического ингибитора циклогексиназы-2 (COX-2) — аспизола — на злокачественные клетки рака молочной железы in vitro и in vivo. Методы: выживаемоть клеток MDA-MB-231 определяли с помощью MTT-теста. Апоптотический индекс измеряли с помощью проточной цитометрии и иммуногистохимическим окрашиванием с антителами против COX-2 и каспазы-3. Экспрессию bcl-2 и bax изучали с помощью Вестерн-блот-анализа. Содержание простагландина E2 (PGE2 ) в клетках MDA-MB-231 оценивали методом ELISA. In vivo апоптоз опухолевых клеток определяли путем выявления разрывов ДНК с помощью концевой дезоксинуклеот-идилтранферазы (метод TUNEL). Результаты: показано, что в зависимости от времени инкубации и дозы аспизол угнетал рост клеток MDA-MB-231 in vitro и вызывал их апоптоз на фоне повышения экспрессии каспазы-3 и bax, а также снижения экспрессии COX-2 и bcl-2. В условиях in vivo аспизол также ингибировал пролиферацию злокачественных клеток рака молочной железы и вызывал их апоптоз. Выводы: данные, полученные in vitro и in vivo, свидетельствуют о противоопухолевом эффекте аспизола на клетки рака молочной железы, что скорее всего опосредовано его проапоптотическим действием и может быть связано со снижением экспрессии COX-2 и bcl-2, а также повышением экспрессии каспазы-3 и bax. 2008 Article Aspisol inhibits tumor growth and induces apoptosis in breast cancer / X.G. Zhu, L. Tao, Z.R. Mei, H.P. Wu, Z.W. Jiang // Experimental Oncology. — 2008. — Т. 30, № 4. — С. 289–294. — Бібліогр.: 25 назв. — англ. 1812-9269 http://dspace.nbuv.gov.ua/handle/123456789/139930 en Experimental Oncology Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України |
institution |
Digital Library of Periodicals of National Academy of Sciences of Ukraine |
collection |
DSpace DC |
language |
English |
topic |
Original contributions Original contributions |
spellingShingle |
Original contributions Original contributions Zhu, X.G. Tao, L. Mei, Z.R. Wu, H.P. Jiang, Z.W. Aspisol inhibits tumor growth and induces apoptosis in breast cancer Experimental Oncology |
description |
Nonsteroidal anti-inflammatory drugs inhibit cell proliferation and induce apoptosis in various cancer cell lines, which is considered to be an important mechanism for their anti-tumor activity and cancer prevention. However, the molecular mechanisms through which these compounds induce apoptosis are not well understood. Aim: to determine the effects of nonselective cyclooxygenase-2 (COX-2) inhibitor, aspisol on breast cancer cells in vitro and in vivo. Methods: The cytotoxic activity of aspisol was evaluated by MTT assay. The apoptosis index of cells was measured by flow cytometry. Immunohistochemical staining was used to detect expressions of COX-2 and caspase-3 in MDA-MB-231 cells. The expression of bcl-2 and bax was analyzed by Western blot analysis. The content of prostaglandin E2 (PGE2) in MDA-MB-231 cells was estimated by ELISA. In vivo apoptosis of the tumor cells was detected by the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). Results: Our results showed that aspisol reduced viability of MDA-MB-231 cells in time- and dose- dependent fashions and induced apoptosis by increase of caspase-3 and bax expressions while decrease of COX-2 and bcl-2 expression in vitro. In addition, exposure to aspisol decreased the basal release of PGE2. In vivo, aspisol also inhibited the proliferation of breast cancer cells and induced their apoptosis. Conclusions: Our in vitro and in vivo data indicated that the antitumor effects of aspisol on breast cancer cells was probably mediated by the induction of apoptosis, and it could be linked to the downregulation of the COX-2 or bcl-2 expression and up-regulation of caspase-3 or bax expression. |
format |
Article |
author |
Zhu, X.G. Tao, L. Mei, Z.R. Wu, H.P. Jiang, Z.W. |
author_facet |
Zhu, X.G. Tao, L. Mei, Z.R. Wu, H.P. Jiang, Z.W. |
author_sort |
Zhu, X.G. |
title |
Aspisol inhibits tumor growth and induces apoptosis in breast cancer |
title_short |
Aspisol inhibits tumor growth and induces apoptosis in breast cancer |
title_full |
Aspisol inhibits tumor growth and induces apoptosis in breast cancer |
title_fullStr |
Aspisol inhibits tumor growth and induces apoptosis in breast cancer |
title_full_unstemmed |
Aspisol inhibits tumor growth and induces apoptosis in breast cancer |
title_sort |
aspisol inhibits tumor growth and induces apoptosis in breast cancer |
publisher |
Інститут експериментальної патології, онкології і радіобіології ім. Р.Є. Кавецького НАН України |
publishDate |
2008 |
topic_facet |
Original contributions |
url |
http://dspace.nbuv.gov.ua/handle/123456789/139930 |
citation_txt |
Aspisol inhibits tumor growth and induces apoptosis in breast cancer / X.G. Zhu, L. Tao, Z.R. Mei, H.P. Wu, Z.W. Jiang // Experimental Oncology. — 2008. — Т. 30, № 4. — С. 289–294. — Бібліогр.: 25 назв. — англ. |
series |
Experimental Oncology |
work_keys_str_mv |
AT zhuxg aspisolinhibitstumorgrowthandinducesapoptosisinbreastcancer AT taol aspisolinhibitstumorgrowthandinducesapoptosisinbreastcancer AT meizr aspisolinhibitstumorgrowthandinducesapoptosisinbreastcancer AT wuhp aspisolinhibitstumorgrowthandinducesapoptosisinbreastcancer AT jiangzw aspisolinhibitstumorgrowthandinducesapoptosisinbreastcancer |
first_indexed |
2025-07-10T09:23:54Z |
last_indexed |
2025-07-10T09:23:54Z |
_version_ |
1837251375258402816 |
fulltext |
Experimental Oncology 30, 289–294, 2008 (December) 289
Breast cancer is the second most common cause
of cancer death in women [1]. The incidence of breast
cancer is increasing but current therapy is unable to
achieve clinical responses in patients with this highly
invasive metastatic disease. There is a consequent
need for more effective approaches to prevention and
treatment of breast cancer. Although many cancers
initially respond to chemotherapy, resistance often
develops. Because many breast cancer patients
treated by standard schemes suffer from undesirable
side effects [2], studies of new approaches of breast
cancer treatment should be continued.
Nonsteroidal anti-inflammatory drugs (NSAIDs) are
well known to inhibit cyclooxygenase (COX) activity, the
key enzyme in prostaglandin biosynthesis. However,
several clinical observations, epidemiological and ex-
perimental studies showed that NSAIDs could be promi-
sing anti-cancer agents. COX-2 overexpression was
found in breast cancer tissues and it was associated with
poorer prognosis [3]. Epidemiological studies as well as
early clinical trials suggest that administration of either
dual COX-1/COX-2 or selective COX-2 inhibitors may
reduce the risk of cancer development [4]. Preclinical
studies also indicated that the inhibition of COX is useful
in animal models of chemoprevention [5]. Inhibition of
COX-2 can decrease breast cancer cell motility, invasion
and matrix metalloproteinase expression [6]. Aspirin has
been shown to be associated with lower risks of cancer
incidence and mortality [7]. It was reported recently that
the use of NSAIDs for 5–9 years for more than 10 years
reduced the incidence of breast cancer by 21% and
28%, respectively [8]. Other studies showed that aspirin
and non-aspirin-NSAIDs contributed to breast cancer
prevention in the general population [9, 10], and NSAIDs
induced apoptosis of tumor cells [11–13]. But the mo-
lecular mechanism of NSAIDs-mediated apoptosis is
still unclear. Preclinical trials are needed to determine
whether NSAIDs could be used for prevention and/or
treatment of breast cancer. In spite of the established
role of COX-2 and NSAIDs in human cancer, little is
known about the effect and mechanism of NSAIDs in
the growth control of breast cancer cells. In the present
investigation, we demonstrated that aspisol reduced
MDA-MB-231 cells viability, induced their apoptosis by
increasing the expression of caspase-3 and bax, and
decreased the expression of COX-2 and bcl-2. Also, we
demonstrated that aspisol inhibited tumor growth of and
induced tumor cells apoptosis in C3H mice model.
MATERIALS AND METHODS
Cell Culture and Drug Treatment. The human
breast cancer cell line MDA-MB-231 was obtained from
the American Type Culture Collection (Rockville, MD,
USA). The cells were grown in Dulbecco’s modified
eagle medium (DMEM) (GIBCO-BRL, Rockville, USA)
supplemented with 10% fetal bovine serum (FBS),
100 U penicillin, 0.1 μg streptomycin and 2 mmol/L
L-glutamine at 37 °C, with 5% CO2. The cells were plated
in the regular medium for 24 h, which was then replaced
by either control fresh FBS-free medium or the medium
containing 1, 5, or 10 mM of aspisol (Fengyaun, Anhui,
China). Drugs were dissolved directly in DMEM.
3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetra-
zolium bromide (MTT) assay. MTT assay was used to
measure cell viability. Briefly, 2 × 104 MDA-MB-231 cells
were seeded in 96-well plates in 180 μl of medium, and
incubated in medium containing different concentrations
of aspisol (1–10 mM) for 24 h, 48 h and 72 h. 20 μl of MTT
ASPISOL INHIBITS TUMOR GROWTH AND INDUCES APOPTOSIS
IN BREAST CANCER
X.G. Zhu, L. Tao, Z.R. Mei, H.P. Wu, Z.W. Jiang*
Department of Pharmacology, Pharmacy Department, Bengbu Medical College, Bengbu 233003, China
Nonsteroidal anti-inflammatory drugs inhibit cell proliferation and induce apoptosis in various cancer cell lines, which is considered to
be an important mechanism for their anti-tumor activity and cancer prevention. However, the molecular mechanisms through which
these compounds induce apoptosis are not well understood. Aim: to determine the effects of nonselective cyclooxygenase-2 (COX-2)
inhibitor, aspisol on breast cancer cells in vitro and in vivo. Methods: The cytotoxic activity of aspisol was evaluated by MTT assay. The
apoptosis index of cells was measured by flow cytometry. Immunohistochemical staining was used to detect expressions of COX-2 and
caspase-3 in MDA-MB-231 cells. The expression of bcl-2 and bax was analyzed by Western blot analysis. The content of prosta-
glandin E2 (PGE2) in MDA-MB-231 cells was estimated by ELISA. In vivo apoptosis of the tumor cells was detected by the terminal
deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). Results: Our results showed that aspisol reduced viability
of MDA-MB-231 cells in time- and dose- dependent fashions and induced apoptosis by increase of caspase-3 and bax expressions
while decrease of COX-2 and bcl-2 expression in vitro. In addition, exposure to aspisol decreased the basal release of PGE2. In vivo,
aspisol also inhibited the proliferation of breast cancer cells and induced their apoptosis. Conclusions: Our in vitro and in vivo data
indicated that the antitumor effects of aspisol on breast cancer cells was probably mediated by the induction of apoptosis, and it could
be linked to the downregulation of the COX-2 or bcl-2 expression and up-regulation of caspase-3 or bax expression.
Key Words: aspisol, NSAIDs, apoptosis, COX-2, breast cancer cells.
Received: August 30, 2008.
*Correspondence: zhengrong1978@yahoo.com.cn
Abbreviations used: COX-2 — cyclooxygenase-2; FCM — flow
cytometry; PGE2 — prostaglandin E2; NSAIDs — nonsteroidal
anti-inflammatory drugs; TUNEL — terminal deoxynucleotidyl
transferase-mediated dUTP nick-end labeling.
Exp Oncol 2008
30, 4, 289–294
290 Experimental Oncology 30, 289–294, 2008 (December)
(5 mg/ml in PBS) (Sigma, USA) was added to each well,
and the cells were incubated for an additional 4 h. Blue
formazans were released from the cells by adding 150 μl
DMSO with gentle shaking at 37 °C, and absorbance was
measured at 570 nm using a microplate reader (Bio-Tek
Instruments, Richmond, USA). Percent of viabile cells
was defined as the relative absorbance of treated cells
vs untreated control cells.
Western blot analysis. Following aspisol treat-
ments, MDA-MB-231 cells were washed twice with
ice-cold PBS and harvested in sample buffer con-
taining 50 mM Tris-HCl, pH 7.4, 1 mM EDTA, 100 mM
NaCl, 0.5% Triton X-100, 1 mM PMSF, 1 mM sodium
orthovanadate, 1 μg/ml leupeptin, 1 μg/ml pepstatin,
and 10 μg/ml aprotinin. Soluble extracts were prepared
by centrifugation at 12 000 rpm for 30 min at 4 °C.
Protein concentrations were determined by the Brad-
ford assay. Equivalent amounts of protein (40 μg) for
each sample were resolved in 12% SDS-PAGE. After
electrophoresis, proteins were transferred to nitrocel-
lulose membranes and blocked by 5% nonfat milk for
1h. Antibodies used for Western blot analysis included
rabbit anti-bcl-2 antibody, rabbit anti-bax antibody
(Cell Signaling, USA) and mouse anti-аlpha-tubulin
antibody (Sigma, USA), horseradish peroxidase-con-
jugated secondary antibodies (Santa Cruz, USA).
Flow cytometry analysis. 24 h after the treatment
of cells with aspisol poptosis was determined by stai-
ning the cells with annexin V and propidium iodide (PI)
using apoptosis kit from BD Pharmingen (San Diego,
USA). The percentage of stained cells in each quadrant
was quantified using Winmdi 2.9 software.
Immunohistochemistry. To evaluate whether
aspisol treatment could modify caspase-3 and
COX-2 expression of, we detected caspases-3 and
COX-2 expression treated and untreated cells by
immunohistochemical staining. 2 × 105 cells were
seeded in triplicates in 6-well plates on coverslips,
and grown for 24 h. Medium was then replaced with
media containing aspisol (1–10 mM) and cells were
grown for the additional 12 h. The cells on coverslips
were fixed by 4% paraformaldehyde solution, and then
were dehydrated in alcohol. Endogenous peroxidase
was blocked by 3% H2O2 in methanol and avidin/biotin
(Vector Laboratories, Burlingame, CA). The coverslips
were incubated overnight at 4 °C with 1 : 500 dilutied
rabbit anti-caspase-3 or rabbit anti-COX-2 (Santa
Cruz, USA) specific antibody. All appropriate controls
were made.Immunoreactive complexes were detected
using tyramide signal amplification (TSA-indirect) and
visualized with the peroxidase substrate, AEC. Cover-
slips were counter stained with hematoxylin.
Detection of PGE2 level in culture media. MDA-
MB-231 cells were treated with increasing concentra-
tions of aspisol (1–10 mM). Level of PGE2 released to
culture media was measured using a PGE2 enzyme im-
munoassay kit (Cayman Chemical, USA). Medium was
sampled, centrifuged to remove floating cells and im-
mediately frozen at –70 °C before analysis. The PGE2 as-
say was performed according to the manufacturer’s
instructions. The results were expressed relatively to the
control. Data presented are the results of at least three
independent experiments done in triplicates.
Tumor proliferation in C3H mice. Female C3H
mice were obtained from the Animal Production Area
of China Medical University. The mice were maintained
under specific-pathogen-free conditions. We used
6-week-old mice weighing 18 to 22 g, acclimatized
for one week before starting the experiments. C3H
mice spontaneous mammary adenocarcinoma cells
were injected subcutaneously into forelimb axillas
of C3H mice (1 × 107 cells per mouse). In 24 h mice
were treated with vehicle (normal sodium), 5-fluo-
rouracil (5-FU, 10 mg/kg), aspisol (300 mg/kg/day)
for 4 weeks. Each group comprised of 10 animals.
The tumor volume (TV) was assessed every 3 days by
using a calliper measuring of the two major diameters
by the formula TV = d1 · d22 / 2. After administration of
the last dose (24 h) mice were killed, the tumors were
excised, fixed and sliced into 2-mm-thick sections for
analysis tumor apoptosis with Terminal dUTP nick-end-
labeling assay (TUNEL) assay. The TUNEL assay was
performed as directed by the manufacturer. Endoge-
nous peroxidase was blocked in scetions, and they
were treated with 0.25% Triton X-100 in PBS at 50 °C
for 20 min, and incubated with terminal deoxytrans-
ferase enzyme with biotin dUTP and cobalt ions for
90 min at 37 °C. Anti-BrdUrd and TUNEL-labeled sec-
tions were visualized with streptavidin peroxidase and
diaminobenzidine (Dako Corp, Carpinteria, CA), fol-
lowed by hematoxylin staining. Apoptotic nuclei were
stained dark brown, and normal cell nuclei were blue.
The animal experiments were approved by the local
Ethics Committee for Animal Research.
Statistical analysis. All data were expressed
as mean ± SD and analyzed by one-way of variance
(ANOVA) or Student’s t-test using SPSS software (ver-
sion 11.0 for Windows). Significance was accepted at
P < 0.05.
RESULTS
Aspisol inhibited MDA-MB-231 cell viability.
Our results suggested that treatment with aspisol re-
duced cell viability in dose-dependent manner. Lower
concentrations of aspisol (5 and 10 mM) significantly
reduced MDA-MB-231 cells viability in 72 h (Fig. 1).
Aspisol induced dose-dependent apoptosis
in MDA-MB-231 cells. Following 24 h of drug treat-
ment, induction of apoptosis was observed in the
MDA-MB-231 cells in a dose-dependent manner
(Fig. 2). Aspisol at 5 and 10 mM caused the increase
in apoptotic cells.
COX-2 and caspase-3 expression in MDA-
MB-231 cells. To determine whether the effect of as-
pisol was associated with COX-2 caspase-3 expression
MDA-MB-231 cells, immunohistochemical analysis was
performed. It was found that COX-2 was consistently
expressed by MDA-MB-213 cells, and there was signifi-
cant down-regulation of COX-2 expression upon aspisol
treatment (Fig. 3). Treatment of MDA-MB-231 cells
Experimental Oncology 30, 289–294, 2008 (December) 291
with 5 and 10 mM aspisol for 12 h caused significant in-
crease in the caspases-3 expression (Fig. 4). Obtained
results suggested that aspisol-induced apoptosis
in MDA-MB-231 cells correlated with COX-2 down-
regulation and caspase-3 in these cells.
0
20
40
60
80
100
120
0 1 5 10
Aspisol concentration (mM)
Ce
ll
vi
ab
ili
ty
(
%
o
f c
on
tr
ol
)
24 h
48 h
72 h
*
*
*
*
* *
*
*
Fig. 1. Aspisol inhibits MDA-MB-231 cell viability. MDA-MB-231 cells
were treated for 24, 48, or 72 h with 0, 1, 5, or 10 mM aspisol. Cells
viability was determined by MTT analysis. Aspisol significantly
inhibits the viability of MDA-MB-231 cells in a dose-dependent
manner. There is a significant difference between control and
aspisol treatment (*P < 0.05). Experiments were repeated three
times, with similar results
0
2
4
6
8
10
12
14
16
0 1 5 10
Aspisol concentration (mM)
Ap
op
to
si
s
ra
te
(
%
)
Pr
op
id
iu
m
io
di
de
Pr
op
id
iu
m
io
di
de
Pr
op
id
iu
m
io
di
de
Pr
op
id
iu
m
io
di
de
100
100
100
100
10
0
10
0
10
0
10
0
101
101
101
101
10
1
10
1
10
1
10
1
102
102
102
102
10
2
10
2
10
2
10
2
103
103
103
103
10
3
10
3
10
3
10
3
104
104
104
104
10
4
10
4
10
4
10
4
ANNEXIN-FITC
ANNEXIN-FITC
ANNEXIN-FITC
ANNEXIN-FITC
1.80
6.95
3.50
1.72
6.69
17.73
10.92
25.85
37.24
22.12
77.24
55.53
7.27
22.31
7.82
10.83
a b
c d
*
*
*
Fig. 2. Aspisol induces apoptosis in MDA-MB-231 cells.
a, control; b, 1 mM aspisol; c, 5 mM aspisol; d, 10 mM aspisol.
MDA-MB-231 cells were treated for 24 h with 0, 1, 5, or 10 mM
aspisol. Apoptosis was then determined by flow cytometry.
Data was analyzed by Student’s t-test. There is a significant
difference between control and aspisol treatment (*P < 0.05);
aspisol increased the number of apoptotic MDA-MB-231 cells
in dose-dependent manner. Experiments were repeated three
times, with similar results
Aspisol induce the decrease of bcl-2/bax
ratio in MDA-MB-231 cells. To determine whether
the effect of aspisol is associated with the changes
of bcl-2 and bax expression in MDA-MB-231 cells,
Western blot analysis was performed. It was shown
that exposure to 10 mM aspisol induced the decrease
of bcl-2 expression and theincrease of bax expression
in treated cells. The bcl-2/bax ratio was decreased to
15.4 ± 5.9% from control (Fig. 5).
Aspisol inhibited COX-2-mediated PGE2 pro-
duction by MDA-MB-231 cells. To determine
whether COX-2 activity was affected by aspisol
treatment, PGE2 production was mesuared using
a PGE2-specific enzymelinked immunosorbent assay.
The results are presented on Fig. 6. Overall, it was
shown that aspisol treatment reduced PGE2 secretion
in MDA-MB-231 cells in a concentration-dependent
manner (Fig. 6).
Aspisol inhibited tumor growth by inducing
cancer cells apoptosis in C3H mice. To assess the
relevance of the in vitro data, we implanted mammary
adenocarcinoma cells subcutaneously into C3H mice.
Proliferation of breast cancer xenografts treated with
aspisol was significantly reduced (Fig. 7). We observed
an increase in TUNEL positive cells in aspisol-treated
tumor sections in situ as compared with control tumor
sections (Fig. 8).
0
20
40
60
80
100
120
0 1 5 10
Aspisol concentration (mM)
CO
X-
2-
po
st
iv
e
ra
te
(
%
)
a b
c d
*
*
*
Fig. 3. Aspisol decreases levels of COX-2 in MDA-MB-231 cells.
a, control; b, 1 mM aspisol; c, 5 mM aspisol; d, 10 mM aspisol.
MDA-MB-231 cells were treated for 12 h with 0, 1, 5, or 10 mM
aspisol. COX-2 expression was determined by immunohis-
tochemical analysis with specific antibodies. Data was analyzed
using one-way ANOVA. *P values represent significant difference
between vehicle control and aspisol treatment (*P < 0.05). Experi-
ments were repeated three times, with similar results. × 400
292 Experimental Oncology 30, 289–294, 2008 (December)
0
20
40
60
80
100
120
0 1 5 10
Aspisol concentration (mM)
Ca
sp
as
e-
3-
po
st
iv
e
ra
te
(
%
)
a b
c d
*
*
*
Fig. 4. Aspisol increases levels of caspase-3 in MDA-MB-231 cells.
a, control; b, 1 mM aspisol; c, 5 mM aspisol; d, 10 mM aspisol.
MDA-MB-231 cells were treated for 12 h with 0, 1, 5, or 10 mM
aspisol. Caspase-3 expression was determined by immunohis-
tochemical analysis with specific antibodies. Data was analyzed
using one-way ANOVA. There is a significant difference between
vehicle control and aspisol treatment (*P < 0.05). Experiments
were repeated three times, with similar results. × 400
0
0.5
1
1.5
2
2.5
3
3.5
4
4.5
0 1 5 10
Aspisol concentration (mM)
R
el
at
iv
e
op
tic
al
d
en
si
ty
bcl-2
bax
bcl-2/bax
20 KDA
28 KDa
50 KDa
bax
bcl-2
a-tubulin
Aspiol (mM) 1 2 3 4
*
*
*
*
*
*
*
*
*
Fig. 5. Aspisol decreases levels of bcl-2 and increases bax level in
MDA-MB-231 cells. After treatment with 0, 1, 5, or 10 mM aspisol
for 12 h, the protein levels of bcl-2 and bax were examined using
Western blotting. Blotting of аlpha-tubulin showed equal loading
of proteins between each lane. Upper panel shows representa-
tive results of three independent experiments. Below panel is bar
graph of gray intensities of the immunoreactive bands analyzed by
software. The ratio of bcl-2/bax was shown as fold of control. Data
were analyzed using one-way ANOVA. There is significant diffe-
rence between vehicle control and aspisol treatment (*P < 0.05).
Experiments were repeated three times, with similar results
0
20
40
60
80
100
120
0 1 5 10
Aspisol concentration (mM)
PG
E2
le
ve
ls
(
%
o
f c
on
tr
ol
)
*
*
*
Fig. 6. Inhibition of production of PGE2 by aspisol.
MDA-MB-231 cells were cultured for 24 h with the indicated con-
centrations of aspisol. The amounts of PGE2 in the conditioned
medium were determined by ELISA and expressed relatively to
the control (*P < 0.05). Experiments were repeated three times,
with similar results
0
200
400
600
800
1000
1200
1400
1600
9 12 15 18 21 24 27
Times (days)
Tu
m
or
v
ol
um
e
(m
m
3 )
Vehicle
Aspisol (300 mg/kg)
5-FU (10 mg/kg)
Fig. 7. Treatment with aspisol inhibits the growth of xenografts
in C3H mice. Tumors were measured one time/3 days
0
2
4
6
8
10
12
Vehicle Aspisol
Tumor cells
Ap
op
to
si
s
in
de
x
(%
)
a b
*
Fig. 8. Effect of aspisol treatment on apoptosis in C3H mice
tumor models. TUNEL assay comparison of vehicle- (a) and
aspisol-treated tumors (b) revealed a marked induction of apop-
tosis in tumor cells in C3H mice models. The apoptosis index of
5% in vehicle-treated tumors increased to 9% in aspisol-treated
tumors. × 400
Experimental Oncology 30, 289–294, 2008 (December) 293
DISCUSSION
COX catalyzes the formation of prostaglandins
from arachidonic acid. Overexpression of COX leads
to increased amounts of prostanoids in tumors. Pros-
tanoids affect numerous mechanisms that have been
implicated in carcinogenesis. PGE2 can stimulate cell
proliferation and motility while inhibiting immune sur-
veillance and apoptosis [14, 15].
NSAIDs are amongst the most commonly used
medications worldwide, which can inhibit COX activity.
They are considered as effective anti-inflammatory,
anti-pyretic and analgesic drugs, and aspirin is also
effective in both the primary and secondary prevention
of cardiovascular diseases. Aspisol, a new generation
of NSAIDs, inhibits both isoforms of COX (COX-1 and
COX-2) followed by the decrease of prostanoids
level. In this study we examined the effect of aspisol
on human breast cancer MDA-MB-231 cells. The ob-
tained results showed that aspisol strongly induced
MDA-MB-231 cells apoptosis.
Apoptosis is important in malignancy for two rea-
sons [16]. First, suppression of apoptosis appears to
be a critical event in both cancer initiation and pro-
gression. Second, most cytotoxic anticancer agents
cause tumor regression, at least in part, by inducing
apoptosis. Induction of tumor cell apoptosis by NSAIDs
is an important mechanism of their antitumor effects
[17]. Apoptosis is a tightly regulated process involv-
ing changes in the expression or activities of distinct
genes [18]. COX inhibitor engages different apoptosis
pathways in cancer cells, stimulating death receptor
signa ling, activating caspases and indu cing apoptosis
via mitochondrial pathway. Evidence suggests that in-
crease in tumorigenic potential by COX-2 overexpres-
sion is associated with resistance to apoptosis. Two
distinct isoforms of COX exist, the constitutively ex-
pressing COX-1, and the inducible COX-2. COX-1 ex-
presses constitutively in most tissues, whereas the ex-
pression of COX-2 is induced by inflammatory factors,
hormones and mitogens. COX-1 and COX-2 might
all be involved in tumorigene sis. Previous study has
shown that COX-1 and COX-2 specific inhibitiors in
combined treatment produced the significantly greater
inhibition as compared to single agents alone [19].
M.A. Kern et al. [20] showed that COX-2 inhibition
induced apoptosis in hepatocellular carcinoma cells.
Our data revealed that treatment with aspisol caused
down-regulation of COX-2 in the cells of breast cancer
cell line MDA-MB-231.
Caspases are aspartate-specific cysteine pro-
teases, which cleave their substrates on the carboxyl
side of the aspartate residue [21, 22]. Currently at least
14 different caspases are found, of which two-thirds
play a role in apoptosis. Caspase-3 is the most widely
studied enzyme among other caspases. It was de-
monstrated to play a key role in both the death receptor
pathway, initiated by caspase-8, and the mitochondrial
pathway, involving caspase-9. Because caspase-3 is
a critical mediator of apoptosis [21] and correlates with
apoptosis in breast cancer, it is regarded as a marker
for prediction of breast cancer cells’ response or resis-
tance to chemotherapeutic agents. We demonstrated
that aspisol caused up-regulation of caspase-3 in the
MDA-MB-231 cells, suggesting that up-regulation of
caspases-3 was involved in aspisol-induced tumor
cell apoptosis.
Bcl-2 and bax are other important factors regula ting
apoptosis. Bcl-2 stabilizes mitochondrial memb rane
integrity by preventing cytochrome c release, and sub-
sequent activation of caspases followed by apoptosis
[23, 24]. It has been proposed that the anti-apoptotic
bcl-2 protein and the pro-apoptotic bcl-2 family bax
proteinare associated with mitochondria-mediated
apoptosis through regulation of mitochondrial mem-
brane permeability. The ratio of bcl-2 to bax may
ultimately determine the fate of cells [25]. Liu et al.
[12] confirmed the relationship between COX-2 and
bcl-2 family proteins in prostate cancer. Our study
showed that aspisol treatment significantly reduced
the bcl-2/bax ratio in MDA-MB-231 cells. We found
that the levels of COX-2 as well as the bcl-2/bax ratio
were decreased in MDA-MB-231 cells upon treatment
with aspisol, suggesting that COX-2 and bcl-2 fami-
ly were involved in aspisol-mediated apoptosis of
MDA-MB-231 breast cancer cells. Up-regulation of
bcl-2 by COX-2 may be the mechanism ofthe reduction
of apoptotic susceptibility in MDA-MB-231 cells.
In conclusion, it could be assumed that the non-se-
lective COX-2 inhibitor, aspisol, can suppress the via-
bility of MDA-MB-231 cells by induction of apoptosis.
This effect of aspisol correlated with down-regulation
of COX-2 and bcl-2 expression and up-regulation of
caspase-3 expression. Therfore, aspisol should be re-
garded as the potential chemotherapeutic and cancer
preventive agent in human breast cancer prevention/
treatment.
REFERENCES
1. Chan K, Morris GJ. Chemoprevention of breast cancer
for women at high risk. Semin Oncol 2006; 33: 642–6.
2. Brown K. Breast cancer chemoprevention: risk-benefit
effects of the antioestrogen tamoxifen. Expert Opin Drug Saf
2002; 1: 253–67.
3. Ristimäki A, Sivula A, Lundin J, et al. Prognostic sig-
nificance of elevated cyclooxygenase-2 expression in breast
cancer. Cancer Res 2002; 62: 632–5.
4. Thun MJ, Namboodiri MM, Calle EE, et al. Aspirin use
and risk of fatal cancer. Cancer Res 1993; 53: 1322–7.
5. Kobayashi H, Uetake H, Higuchi T, et al. JTE-522,
a selective COX-2 inhibitor, inhibits growth of pulmonary
metastases of colorectal cancer in rats. BMC Cancer 2005;
5: 26–33.
6. Larkins TL, Nowell M, Singh S, et al. Inhibition of
cyclooxygenase-2 decreases breast cancer cell motility, inva-
sion and matrix metalloproteinase expression. BMC Cancer
2006; 10: 181–92.
7. Bardia A, Ebbert JO, Vierkant RA, et al. Association of
aspirin and nonaspirin nonsteroidal anti-inflammatory drugs
with cancer incidence and mortality. J Natl Cancer Inst 2007;
99: 881–9.
8. Harris RE, Chlebowski RT, Jackson RD, et al. Breast
cancer and nonsteroidal antiinflammatory drugs: prospective
294 Experimental Oncology 30, 289–294, 2008 (December)
results from the Women's health initiative. Cancer Res 2003;
63: 6096–101.
9. Swede H, Mirand AL, Menezes RJ, et al. Association
of regular aspirin use and breast cancer risk. Oncology 2005;
68: 40–7.
10. Zhang Y, Coogan PF, Palmer JR, et al. Use of non-
steroidal antiinflammatory drugs and risk of breast cancer: the
Case-Control Surveillance Study revisited. Am J Epidemiol
2005; 162: 165–70.
11. Li M, Lotan R, Levin B, et al. Aspirin induction of
apoptosis in esophageal cancer: a potential for chemopreven-
tion. Cancer Epidemiol Biomark Prev 2000; 9: 545–9.
12. Liu XH, Yao S, Kirschenbaum A, et al. NS398, a se-
lective cyclooxygenase-2 inhibitor, induces apoptosis and
downregulates bcl-2 expression in LNCaP cells. Cancer Res
1998; 58: 4245–9.
13. Sheng H, Shao J, Kirkland SC, et al. Inhibition of
human colon cancer cell growth by selective inhibition of
cyclooxygenase-2. J Clin Invest 1997; 99: 2254–9.
14. Cohen EG, Almahmeed T, Du B, et al. Microsomal
prostaglandin E synthase-1 is overexpressed in head and
neck squamous cell carcinoma.Clin Cancer Res 2003;
9: 3425–30.
15. Sheng H, Shao J, Washington MK, et al. Prostaglandin
E2 increases growth and motility of colorectal carcinoma cells.
J Biol Chem 2001; 276: 18075–81.
16. Evan GI, Vousden KH. Proliferation, cell cycle and
apoptosis in cancer. Nature 2001; 411: 342–8.
17. Roy HK, Karoski WJ, Ratashak A, et al. Chemopre-
vention of intestinal tumorigenesis by nabumetone: induction
of apoptosis and Bcl-2 downregulation. Br J Cancer 2001;
84: 1412–16.
18. Israels LG, Israels ED. Apoptosis. Oncologist 1999;
4: 332–9.
19. McFadden DW, Riggs DR, Jackson BJ, et al. Additive
effects of Cox-1 and Cox-2 inhibition on breast cancer in vitro.
Int J Oncol 2006; 29: 1019–23.
20. Kern MA, Haugg AM, Koch AF, et al. Schulze-Bergka-
men H, Friess H, Stremmel W, Krammer PH, Schirmacher P,
Müller M. Cyclooxygenase-2 inhibition induces apoptosis
signaling via death receptors and mitochondria in hepatocel-
lular carcinoma. Cancer Res 2006; 66: 7059–66.
21. Stennicke HR, Salvesen GS. Properties of the caspases.
Biochim. Biophys. Acta 1998; 1387: 17–31.
22. Thornberry NA, Lazebnik Y. Caspases: enemies within.
Science 1998; 281: 1312–6.
23. Kluck RM, Bossy-Wetzel E, Green DR, et al. The
release of cytochrome c from mitochondria: A primary site
for Bcl-2 regulation of apoptosis. Science 1997; 275: 1132–6.
24. Yang J, Liu X, Bhalla K, et al. Prevention of apoptosis
by Bcl-2: release of cytochrome c from mitochondria blocked.
Science 1997; 275: 1129–32.
25. Ferri KF, Kroemer G. Organelle-specific initiation of
cell death pathways. Nat Cell Biol 2001; 3: E255–63.
Copyright © Experimental Oncology, 2008
АСПИЗОЛ ИНГИБИРУЕТ РОСТ И ВЫЗЫВАЕТ АПОПТОЗ КЛЕТОК
РАКА МОЛОЧНОЙ ЖЕЛЕЗЫ
Нестероидные противовоспалительные препараты ингибируют пролиферацию клеток и вызывают апоптоз во многих
опухолевых клеточных линиях, что считается важным механизмом их противоопухолевой активности и профилактики
развития рака. Тем не менее молекулярные механизмы апоптотического действия этих препаратов изучены недостаточно.
Цель: изучить действие неспецифического ингибитора циклогексиназы-2 (COX-2) — аспизола — на злокачественные клетки
рака молочной железы in vitro и in vivo. Методы: выживаемоть клеток MDA-MB-231 определяли с помощью MTT-теста.
Апоптотический индекс измеряли с помощью проточной цитометрии и иммуногистохимическим окрашиванием с анти-
телами против COX-2 и каспазы-3. Экспрессию bcl-2 и bax изучали с помощью Вестерн-блот-анализа. Содержание про-
стагландина E2 (PGE2) в клетках MDA-MB-231 оценивали методом ELISA. In vivo апоптоз опухолевых клеток определяли
путем выявления разрывов ДНК с помощью концевой дезоксинуклеот-идилтранферазы (метод TUNEL). Результаты:
показано, что в зависимости от времени инкубации и дозы аспизол угнетал рост клеток MDA-MB-231 in vitro и вызывал
их апоптоз на фоне повышения экспрессии каспазы-3 и bax, а также снижения экспрессии COX-2 и bcl-2. В условиях
in vivo аспизол также ингибировал пролиферацию злокачественных клеток рака молочной железы и вызывал их апоптоз.
Выводы: данные, полученные in vitro и in vivo, свидетельствуют о противоопухолевом эффекте аспизола на клетки рака
молочной железы, что скорее всего опосредовано его проапоптотическим действием и может быть связано со снижением
экспрессии COX-2 и bcl-2, а также повышением экспрессии каспазы-3 и bax.
Ключевые слова: аспизол, NSAIDs, апоптоз, COX-2, рак молочной железы.
|