ArticlePDF Available

Stable transfection of extrinsic Smac gene enhances apoptosis-inducing effects of chemotherapeutic drugs on gastric cancer cells

Authors:

Abstract and Figures

To explore the feasibility of enhancing apoptosis-inducing effects of chemotherapeutic drugs on human gastric cancer cells by stable transfection of extrinsic Smac gene. After Smac gene was transferred into gastric cancer cell line MKN-45, subclone cells were obtained by persistent G418 selection. Cellular Smac gene expression was determined by RT-PCR and Western blotting. After treatment with mitomycin (MMC) as an apoptotic inducer, in vitro cell growth activities were investigated by trypan blue-staining method and MTT colorimetry. Cell apoptosis and its rates were determined by electronic microscopy, annexin V-FITC and propidium iodide staining flow cytometry. Cellular caspase-3 protein expression and its activities were assayed by Western blotting and colorimetry. When compared with MKN-45 cells, the selected subclone cell line MKN-45/Smac had significantly higher Smac mRNA (3.12+/-0.21 vs 0.82+/-0.14, t = 7.52, P<0.01) and protein levels (4.02+/-0.24 vs 0.98+/-0.11, t = 8.32, P<0.01). After treatment with 10 microg/mL MMC for 6-24 h, growth inhibition rate of MKN-45/Smac (15.8+/-1.2-54.8+/-2.9%) was significantly higher than that of MKN-45 (5.8+/-0.4- 24.0+/-1.5%, t = 6.42, P<0.01). Partial MKN-45/Smac cancer cells presented characteristic morphological changes of apoptosis under the electronic microscope with an apoptosis rate of 36.4+/-2.1%, which was significantly higher than that of MKN-45 (15.2+/-0.8%, t = 9.25, P<0.01). Compared with MKN-45, caspase-3 expression levels in MKN-45/Smac were improved significantly (3.39+/-0.42 vs 0.96+/-0.14, t = 8.63, P<0.01), while its activities were 3.25 times as many as those of MKN-45 (0.364+/-0.010 vs 0.112+/-0.007, t = 6.34, P<0.01). Stable transfection of extrinsic Smac gene and its over-expression in gastric cancer cell line can significantly enhance cellular caspase-3 expression and activities, ameliorate apoptosis-inducing effects of mitomycin C on cancer cells, which is a novel strategy to improve chemotherapeutic effects on gastric cancer.
Content may be subject to copyright.
GASTRIC CANCER
Stable transfection of extrinsic Smac gene enhances apoptosis-inducing
effects of chemotherapeutic drugs on gastric cancer cells
Li-Duan Zheng, Qiang-Song Tong, Liang Wang, Jun Liu, Wei Qian
PO Box 2345, Beijing 100023, China World J Gastroenterol 2005;11(1):79-83
Fax: +86-10-85381893 World Journal of Gastroenterology ISSN 1007-9327
E-mail: wjg@wjgnet.com www.wjgnet.com © 2005 The WJG Press and Elsevier Inc. All rights reserved.
ELSEVIER
Li-Duan Zheng, Department of Pathology, Union Hospital of Tongji
Medical College, Huazhong University of Science and Technology,
Wuhan 430022, Hubei Province, China
Qiang-Song Tong, Liang Wang, Department of Surgery, Union
Hospital of Tongji Medical College, Huazhong University of Science
and Technology, Wuhan 430022, Hubei Province, China
Jun Liu, Wei Qian, Department of Gastroenterology, Union Hospital
of Tongji Medical College, Huazhong University of Science and
Technology, Wuhan 430022, Hubei Province, China
Correspondence to: Dr. Qiang-Song Tong, Department of Surgery,
Union Hospital of Tongji Medical College, Huazhong University of
Science and Technology, Wuhan 430022, Hubei Province,
China.
qs_tong@hotmail.com
Telephone: +86-27-85726129
Received: 2004-03-03 Accepted: 2004-04-05
Abstract
AIM: To explore the feasibility of enhancing apoptosis-
inducing effects of chemotherapeutic drugs on human gastric
cancer cells by stable transfection of extrinsic Smac gene.
METHODS:
After Smac gene was transferred into gastric
cancer cell line MKN-45, subclone cells were obtained by
persistent G
418
selection. Cellular Smac gene expression
was determined by RT-PCR and Western blotting. After
treatment with mitomycin (MMC) as an apoptotic inducer,
in vitro cell growth activities were investigated by trypan
blue-staining method and MTT colorimetry. Cell apoptosis
and its rates were determined by electronic microscopy,
annexin V-FITC and propidium iodide staining flow cytometry.
Cellular caspase-3 protein expression and its activities were
assayed by Western blotting and colorimetry.
RESULTS:
When compared with MKN-45 cells, the selected
subclone cell line MKN-45/Smac had significantly higher
Smac mRNA (3.12±0.21 vs 0.82±0.14, t = 7.52, P<0.01) and
protein levels (4.02±0.24 vs 0.98±0.11, t = 8.32, P<0.01).
After treatment with 10 µg/mL MMC for 6-24 h, growth
inhibition rate of MKN-45/Smac (15.8±1.2-54.8±2.9%)
was significantly higher than that of MKN-45 (5.8±0.4-
24.0±1.5%, t = 6.42, P<0.01). Partial MKN-45/Smac cancer
cells presented characteristic morphological changes of
apoptosis under the electronic microscope with an apoptosis
rate of 36.4±2.1%, which was significantly higher than that
of MKN-45 (15.2±0.8%, t = 9.25, P<0.01). Compared with
MKN-45, caspase-3 expression levels in MKN-45/Smac were
improved significantly (3.39±0.42 vs 0.96±0.14, t = 8.63,
P<0.01), while its activities were 3.25 times as many as those
of MKN-45 (0.364±0.010 vs 0.112±0.007, t = 6.34, P<0.01).
CONCLUSION:
Stable transfection of extrinsic Smac gene
and its over-expression in gastric cancer cell line can
significantly enhance cellular caspase-3 expression and
activities, ameliorate apoptosis-inducing effects of mitomycin
C on cancer cells, which is a novel strategy to improve
chemotherapeutic effects on gastric cancer.
© 2005 The WJG Press and Elsevier Inc. All rights reserved.
Key words: Gastric cancer; Mitomycin C; Extrinsic Smac
gene; Apoptosis; Transfection
Zheng LD, Tong QS, Wang L, Liu J, Qian W. Stable transfection
of extrinsic Smac gene enhances apoptosis-inducing effects
of chemotherapeutic drugs on gastric cancer cells. World J
Gastroenterol 2005; 11(1):79-83
http://www.wjgnet.com/1007-9327/11/79.asp
INTRODUCTION
Up to now, chemotherapy is still the important adjuvant treatment
for postoperative and advanced gastric cancer. Its research
focuses on how to reduce the side effects of chemotherapeutic
drugs and improve the sensitivities of tumor cells to them
[1-4]
. A
series of researches indicate chemotherapeutic drugs, radiotherapy
and thermotherapy could all induce apoptosis to various extents
by exerting their anti-tumor effects
[5-7]
. The second mitochondria-
derived activator of caspases (Smac) or DIABLO gene is a recently
identified and novel proapoptotic molecule, which is released from
mitochondria into the cytosol when cell apoptosis undergoes, to
enhance activities of caspase-3 through eliminating the functions
of inhibitors of apoptosis proteins (IAPs)
[8]
. It was reported that
the apoptosis-inducing effects of chemotherapeutic drugs on ovary
cancer and leukemia could be significantly enhanced by gene
transfer of Smac into cancer cells
[9,10]
. In this study, we
investigated the effects of stable transfection of extrinsic Smac
gene on apoptosis of gastric cancer cells induced by
chemotherapeutic drugs, to explore a novel strategy to improve
chemotherapeutic effects on gastric cancers.
MATERIALS AND METHODS
Genes and main reagents
Eukaryotic vector pcDNA3.1-Smac containing a full-length
human Smac cDNA (719 bp) was kindly provided by Professor
Xiao-Dong Wang (USA). Blank vector pcDNA3.1 was
preserved
by our central laboratory. Polyclonal rabbit anti-human Smac
was a kind gift from Professor Emma (Ireland). Monoclonal mouse
anti-human caspase-3 and its activity detection kit were purchased
from Santa Cruz Biotechnology and Clontech Company
respectively. Annexin V-FITC reagent kit was purchased from
Jingmei Biotech Company. Liposome GeneSHUTTLE-40 was
purchased from Q-bio Gene Limited Company. Newborn calf
serum, RPMI 1640, Trizol
TM
reagent kit and G
418
were all
purchased from Gibco Company. Dimethyl sulfoxide (DMSO)
and 3, [4,5- dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide
(MTT) were both purchased from Clontech Company. Mitomycin
C (MMC) was a product from Japan Union Ferment Industry
Company, which was prepared into 1 g/L stock solution with
PBS, preserved at 4 and kept from light.
Cell culture
Human gastric cancer cell line MKN-45 was kindly provided by
Professor Ji-Hua Dong, Department of Virology, Union Hospital
of Tongji Medical College, Huazhong University of Science
and Technology, China. Cells were cultured in RPMI 1640 medium
supplemented with penicillin/streptomycin (100 units/mL and
100 µg/mL respectively) and 10% neonatal bovine serum at 37
in a humidified atmosphere of 50 mL/L CO
2
and passaged every
three days.
Gene transfection and selection of subclone cell line
The MKN-45 cells at exponential phases of growth were
inoculated into a 24-well plate. At the same time, non-
transfection, pcDNA3.1 and pcDNA3.1-Smac transfection
groups were designed for this experiment. Gene transfection was
conducted according to the protocol of liposome GeneSHUTTLE-
40 (G40). In brief, 4-6 µL G40 was mixed with 1 µg pcDNA3.1-Smac
or pcDNA3.1 and the mixture was incubated for 30 min, then
DNA/liposome complex solutions were added into the wells.
After incubated at 37
in a mosphere containing 5 mL/L CO
2
for 48 h, the cells were digested with 0.01%EDTA, then seeded
into a 6-well plate (35 mm in diameter) at 1×10
8
/L density, and
then selected with 600 mg/L G
418
for 2 wk. When most of the
non-transfected cells were dead, the concentration of G
418
was
decreased to 300 mg/L and maintained for another 2 wk. After
cellular clones were formed, subclones were chosen at random
and amplified. The subclone cells expressing Smac and neo genes
were named as MKN-45/Smac and MKN-45/neo respectively.
Detection of cellular Smac mRNA expression
Cellular Smac mRNA expression levels were assayed by reverse
transcription polymerase chain reaction (RT-PCR). Total cellular
RNA extraction was conducted with Trizol
TM
reagent kit,
according to the protocol of the manufacturer. The reverse
transcription was conducted at 42 for 30 min in 25 µL total
volume containing 2 µg template RNA, 1 µL 10 mmol/L dNTP,
20 U RNasin, 1 µL Oligo dT
18
, 200 U AMV, 5 µL 5×AMV
buffer. The PCR primers for Smac gene were designed by Primer
5.0 software: upstream 5’-CTGTGACGATTGGCTTTG-3’,
downstream 5’-GTGATT CCTGGCGGTTAT-3’, which were
synthesized by Shanghai GeneBase Company. The anticipated
product length was 425 bp. α-tubulin (310 bp) served as an
internal control. Touchdown PCR (TD PCR) was used for
amplification reaction. Amplified products were separated with
2% agarose electrophoresis. The brightness ratio between Smac
and α-tubulin was evaluated with a gel computer image system
(MGIAS-1000, Bio-Rad Company).
Detection of cellular Smac protein expression
Cellular Smac protein expression levels were assayed by
Western blotting. The extraction, quantification and separation
of proteins were conducted as previously described in Molecular
Cloning. Blots were incubated sequentially with 1% nonfat
dry milk, rabbit polyclonal anti-Smac antibody and goat radish
peroxidase-conjugated immunoglobulin G, and evaluated using
an ECL Western blotting kit. Smac protein band intensities
were determined densitometrically using the video imaging
CMIASWIN system.
Cell growth curve
MKN-45, MKN-45/neo and MKN-45/Smac cells were seeded at
2×10
8
/L density into the 24-well chamber slides (each group had
five wells). After cells were attached to the wells, 10 mg/L MMC
was added into each well. Then cells were incubated with RPMI
1640 at 37 in at mospbere containg 5 mL/L CO
2
for 0 h, 6 h,
12 h, 18 h and 24 h respectively. Then cells were digested by
0.125% trypsinase + 0.01% EDTA, stained with trypan blue and
counted under an inversion microscope. For each well, cell count
was repeated 3 times to draw the cell growth curve.
Detection of cellular chemotherapeutic sensitivity
MTT colorimetry was used. MKN-45, MKN-45/neo and MKN-
45/Smac cells were seeded at 3×10
4
/L density into 96-well
chamber slides. For each cell line, untreated control group,
0.1 mg/L MMC group, 1 mg/L MMC and 10 mg/L MMC group
were designed, with each group having five wells. After treating
with MMC for 24 h, 20 µL MTT (5 g/L) was added into each well
and cultured for another 4 h, the supernatant was discarded,
then 100 µL DMSO was added. When the crystals were dissolved,
the optical density A values of the slides were read on the enzyme-
labeled minireader II at the wavelength of 490 nm. Cell proliferation
inhibitory rate (%) = (1 - average A
490 nm
value of experimental
group/average A
490 nm
value of control group) ×100%. For each
detection, the total procedure was repeated 3 times.
Cell ultrastructure observation
After treated with 10 mg/L MMC for 24 h, three kinds of cancer
cells were collected, sequentially rinsed in PBS, fixed with 2.5%
glutaraldehyde for 30 min, and then washed with PBS. After
routine embedment and section, cells were observed under an
electron microscope.
Detection of cell apoptosis
After treatment with 10 mg/L MMC for 24 h, apoptotic ratios of
three kinds of cells were determinated by annexin V-FITC and
propidium iodide staining flow cytometry. Cells from the above
groups were collected, washed twice with cold PBS,
resuspended with 100 µL binding buffer (10 mmol/L HEPES,
140 mmol/L NaCl, 2.5 mmol/L CaCl
2
, pH 7.4) into 2 - 5×10
5
cells /mL
density, and incubated with annexin V-FITC at room temperature
for 10 min. After washing with binding buffer, the cells were
resuspended with 400 µL binding buffer containing 10 µL
propidium iodide (20 µg/mL), and incubated on ice for 15 min.
Apoptosis was analyzed by flow cytometry (BD Company,
USA) at a wavelength of 488 nm.
Detection of caspase-3 protein expression and activities
After treatment with 10 µg/mL MMC for 24 h, cellular caspase-3
protein expression levels were assayed by Western blotting
(as the same method above). The caspase-3 expression levels
were analyzed with the computer imaging system. 2×10
5
cells
from above groups were respectively collected, added into
50 µL cellular lysis buffer, then incubated on ice for 10 min.
After centrifugation (12 000 r/min) at 4 for 3 min, the
supernatant was collected and added sequentially into 50 µL
2×reaction buffer, 5 µL 1.0 mmol/L caspase-3 substrate DEVD-
pNA, and incubated at 37 for 1 h. After
transferred into 96
wells, the optical density A values of the slides were read on
the enzyme-labeled minireader II at the wavelength of 405 nm
(A
405 nm
), which stood for the relative activities of caspase-3.
Statistical analysis
Data was expressed as mean±SD and analyzed using SPSS10.0
statistical software.
RESULTS
Establishment of Smac stably-transfected subclone cells
All the untransfected MKN-45 cells were dead after G
418
(600 µg/mL) selection for 1 wk. The pcDNA3.1 and pcDNA3.1-
Smac transfected cells were continuously selected with G
418
for
4 wk, until magnificent clones could be observed. The clones
were respectively amplified. The subclone MKN-45/neo and
MKN-45/Smac cells were obtained, stably expressing neo and
Smac genes respectively.
80 ISSN 1007-9327 CN 14-1219/ R World J Gastroenterol January 7, 2005 Volume 11 Number 1
Zheng LD
et al. Smac enhances chemotherapeutic effects 81
Cellular Smac mRNA expression
As shown in Figure 1A, after electrophoresis of RT-PCR
products, Smac (425 bp) amplification bands could be observed
in MKN-45, MKN-45/neo and MKN-45/Smac cells. There were
only weak bands in MKN-45 and MKN-45/neo cells, and
brighter bands in MKN-45/Smac cells, amplified with the same
amount of RNA template. MGIAS-1000 gel computer image
system proved that the Smac/α-tubulin ratio in MKN-45/Smac
was 3.8 times, 3.7 times as many as that of MKN-45 (3.12±0.21 vs
0.82±0.14, t = 7.52, P<0.01), MKN-45/neo (3.12±0.21 vs 0.84±0.11,
t = 8.26, P<0.01) respectively. The brightness of Smac bands
between MKN-45 and MKN-45/neo had no significant
difference (P>0.05).
Figure 1
Cellular Smac expression detected by RT-PCR and
Western blotting. A: Cellular Smac mRNA expression detected
by RT-PCR. Lane 1: MKN-45 cells; lane 2: MKN-45/neo cells;
lane 3: MKN-45/Smac cells; lane 4: PCR marker (100 bp, 200 bp,
300 bp, 400 bp, 500 bp, 600 bp, 700 bp, 800 bp, 900 bp, 1000 bp).
B: Cellular Smac protein expression detected by Western
blotting. Lane 1: MKN-45/Smac cells; lane 2: MKN-45/neo
cells; lane 3: MKN-45 cells.
Figure 2 In vitro growth curves of gastric cancer cells after
treatment with 10 mg/L MMC.
Cellular Smac protein expression
As shown in Figure 1B, M
r
27 000 protein bands could be detected by
Western blotting in MKN-45, MKN-45/neo and MKN-45/Smac cells.
Computer image system indicated that Smac protein band brightness
of MKN-45/Smac cells was 4.1 times and 4.2 times as many as
that of MKN-45 (4.02±0.24 vs 0.98±0.11, t = 8.32, P<0.01) and
MKN-45/neo (4.02±0.24 vs 0.96±0.13, t = 8.84, P<0.01) respectively.
There was no significant difference in Smac protein band brightness
between MKN-45 and MKN-45/neo cells (P>0.05).
Cell growth curve
After treated with 10 mg/L MMC for 6-24 h, the in vitro growth
activities of MKN-45, MKN-45/neo and MKN-45/Smac cells
were all decreased. The growth inhibitory rates were 5.8±0.4-
24.0±1.5%, 7.1±0.6-26.8±1.2% and 15.8±1.2-54.8±2.9%
respectively. The differences in growth activities between
MKN-45 and MKN-45/neo cells were not significant (P>0.05),
while the growth activities of MKN-45/Smac cells were reduced
by 10.0±0.9-30.8±1.5% (t = 6.42, P<0.01), when compared with
those of MKN-45 cells (Figure 2).
Cellular sensitivity to MMC
After treated with 0.1 mg/L, 1 mg/L, 10 mg/L MMC, the growth
activities of MKN-45, MKN-45/neo and MKN-45/Smac cells
were reduced in a time- and dose-dependent manner. After
treatment with 10 mg/L MMC for 24 h, the inhibitory rate of
MKN-45 cells was 21.85±1.64%, while that of MKN-45/Smac
cells reached 43.71±3.12%, and the difference between these two
groups was significant (t = 7.56, P<0.01). The difference in growth
inhibitory rate of MMC between MKN-45 and MKN-45/neo
cells was not significant (P>0.05) (Figure 3).
Cellular morphological features
After treating with 10 mg/L MMC for 24 h, some cells had
characteristic morphological changes of apoptosis under an
electron microscope, such as cellular volume reduction, nuclear
shrinkage, chromatin congregation around the nuclear
membrane and integrity of cellular membranes (Figure 4).
Cell apoptosis detection
After treated with 10 mg/L MMC for 24 h, the apoptotic rates of
MKN-45 and MKN-45/neo cells were 15.2±0.8% and 16.5±1.1%
respectively, with no significant difference (P>0.05). The
apoptotic MKN-45/Smac cells increased and the apoptotic rate
reached 36.4±2.1%, with a significant difference compared to
that of MKN-45 (t = 9.25, P<0.01) and MKN-45/neo (t = 7.72,
P<0.01) (Figure 5).
Figure 3 Growth inhibitory effects of various concentrations
of MMC on gastric cancer cells.
Caspase-3 protein expression and activities
As shown in Figure 6, after treated with 10 mg/L MMC for
24 h, 17 ku (p17) and 20 ku (p20) protein bands, subunits of
caspase-3, could all be detected in MKN-45, MKN-45/neo and
MKN-45/Smac cells. Computer image system indicated that there
was a significantly higher p20 expression in MKN-45/Smac
compared to MKN-45 (3.39±0.42 vs 0.96±0.14, t = 8.63, P<0.01)
and MKN-45/neo (3.39±0.42 vs 0.94±0.11, t = 9.43, P<0.01). There
was no significant difference in p20 expression levels between
MKN-45 and MKN-45/neo cells (P>0.05). The A
405 nm
values of
MKN-45, MKN-45/neo and MKN-45/Smac cells were 0.055±0.008,
0.052±0.012 and 0.060±0.011 respectively, while differences
among them were not significant (P>0.05). After treatment
with 10 µg/mL MMC for 24 h, the A
405 nm
value reached 0.364±0.010
in MKN-45/Smac cells, which was 3.25 times as many as that in
MKN-45 cells (0.112±0.007, t = 6.34, P<0.01) (Figure 7).
1 2 3 4
Smac (425 bp)
α-tubulin (310 bp)
600 bp
500 bp
400 bp
300 bp
200 bp
100 bp
1 2 3
Smac 27 ku
A
B
6 12 18 24
12
10
8
6
4
2
0
MKN-45
MKN-45/neo
MKN-45/Smac
Treatment time (h)
Survival cells (10
4
/mL)
50
45
40
35
30
25
20
15
10
5
0
MKN-45
MKN-45/neo
MKN-45/Smac
0.1 µg/L 1 µg/L 10 µg/L
MMC MMC MMC
Cellular growth
inhibition rates (%)
Figure 6 Cellular caspase-3 expression detected by Western
blotting. Lane 1: MKN-45 cells untreated with MMC; lane 2:
MKN-45/neo cells untreated with MMC; lane 3: MKN-45/Smac
cells untreated with MMC; lane 4: MKN-45 cells treated with
10 mg/L MMC; lane 5: MKN-45/neo cells treated with 10 mg/L
MMC; lane 6: MKN-45/Smac cells treated with 10 mg/L MMC.
DISCUSSION
Gastric cancer is one of the most common malignant neoplasms
in alimentary tract. Because of the side effects of chemotherapy
on normal cells and drug resistance of tumor cells, it has been
a research focus on how to ameliorate the chemotherapeutic
effects on gastric cancer
[11]
. Recent researches indicate that
abnormal blockage of apoptosis is an important factor for the
occurrence and development of cancer
[12-14]
. To understand
apoptosis mechanisms is hopeful for improving the sensitivities
of tumor cells to chemotherapeutic drugs, and overcoming drug
resistance
[15,16]
.
The mechanisms of apoptosis are highly conserved in all
sorts of species, including a series of processes. Apoptosis
usually has three phases: initiation, effectors and execution
[17-19]
.
Figure 7
Caspase-3 activities detected in gastric cancer cells
before and after treatment with 10 mg/L MMC.
When external stimuli induce cell apoptosis by different pathways,
such as death receptor-mediated and stress-dependent pathways,
imbalance between activators and inhibitors of apoptosis occurs,
thus activating caspase family and changing mitochondrial outer
membrane permeability, finally resulting in cell apoptosis
[20]
.
Mitochondria are regarded as the key regulation element of cell
death and the target of many proapoptotic signal pathways
[21,22]
.
Some researches demonstrate that there are inhibitors of
apoptosis proteins (IAPs) in mammalian cells, which suppress
apoptosis by inhibiting procaspase activation and catalytic
activity of mature caspases
[23,24]
. IAPs, including MIHA
(mammalian IAP homolog A, or called XIAP), c-IAP1, c-IAP2
and survivin, could bind directly to caspase-3, caspase-7 and
caspase-9, and inhibit their activities, which are the downstream
effectors during cascades of caspase family. MIHA could also
suppress apoptosis induced by chemotherapeutic agents,
UV-irradiation and Bax
[25-27]
. It is suggested that cellular IAPs
accumulation is one of the reasons for cancer cells to escape
Figure 4 Cellular morphological changes of MKN-45/Smac before and after treatment with MMC under electron microscope,
×5 000. A: Cellular ultrastructure before treatment with MMC; B: Cellular chromatin congregating around nuclear membrane
after treatment with 10 mg/L MMC for 24 h; C: Cellular nuclear shrinking after treatment with 10 mg/L MMC for 24 h.
Figure 5 Apoptosis determination in gastric cancer cells by annexin V-FITC and propidium iodide staining flow cytometry. A:
MMC untreated control; B: MKN-45 cells treated with 10 mg/L MMC for 24 h; C: MKN-45/neo cells treated with 10 mg/L MMC
for 24 h; D: MKN-45/Smac cells treated with 1 0 mg/L MMC for 24 h.
ABC
10
0
10
1
10
2
10
3
10
4
10
4
10
3
10
2
10
1
10
0
A
Annexin Y FITC
PIPE
10
0
10
1
10
2
10
3
10
4
10
4
10
3
10
2
10
1
10
0
Annexin Y FITC
PIPE
10
0
10
1
10
2
10
3
10
4
10
4
10
3
10
2
10
1
10
0
Annexin Y FITC
PIPE
10
0
10
1
10
2
10
3
10
4
10
4
10
3
10
2
10
1
10
0
Annexin Y FITC
PIPE
BCD
1 2 3 4 5 6
p32
p20
p17
MKN-45 MKN-45/neo MKN-45/Smac
0.40
0.35
0.30
0.25
0.20
0.15
0.10
0.05
0.00
A
405nm
Value
Untreated control
Treated with MMC
82 ISSN 1007-9327 CN 14-1219/ R World J Gastroenterol January 7, 2005 Volume 11 Number 1
the killing effects of anti-cancer agents
[28]
.
Smac or DIABLO gene is a proapoptotic molecule, which is
released from mitochondria into the cytosol, along with
cytochrome C (cyt-C) during apoptosis. As an important apoptotic
modulator, Smac functions as eliminating the caspase-inhibitory
properties of IAPs
[29]
. Some researches have found that Smac
could promote apoptosis via two pathways, which are dependent
on the interaction between Smac and IAPs
[30]
. One hydrolyzes
caspase-3 protein, and the other enhances the catalytic activity
of mature caspase-3. McNeish et al.
[9]
constructed the adenoviral
vector for Smac gene, and transfected it into ovarian cancer
cells. They found that apoptotic cancer cells increased remarkably,
and cell apoptosis mediated by Smac was not dependent on
cyt-C and Bcl-2, but realized through Caspase-9. Jia et al.
[10]
stably transfected full-length (FL) and mature (MT) Smac genes
into K562 and CEM leukemic cell lines, and they concluded that
both FL and MT Smac transfection could promote the sensitivity
of leukemic cells to UV light, and activate cellular caspase-9 and
caspase-3.
In this study, an extrinsic Smac gene was transfected into
gastric cancer cells which induced its over-expression. We found
that Smac overexpression could enhance the apoptosis-
inducing effects of MMC, by electronic microscopy and annexin
V-FITC and propidium iodide staining flow cytometry. These
results are consistent with those recently reported by Guo et al.
[31]
(2002) and Fulda et al.
[32]
(2002), in which Smac could enhance
apoptosis in leukemia and malignant neuroglioma cells induced
by chemistry or immunology in vivo. Western blotting and
colorimetry were sequentially used to assay the cellular caspase-
3 protein expression and its activities, and it was found that
stable transfection of an extrinsic Smac gene could increase the
cellular activity levels of caspase-3 after treating with MMC,
which accords with the functional mechanisms of Smac. These
results provide a novel strategy to improve chemotherapeutic
sensitivity in gastric caner patients and reduce their side effects,
thus establishing a basis for further exploring the roles of Smac
gene in apoptosis regulation of gastric cancer.
REFERENCES
1 Van Cutsem E, Haller D, Ohtsu A. The role of chemotherapy
in the current treatment of gastric cancer. Gastric Cancer 2002;
5(Suppl 1): 17-22
2 Janunger KG, Hafstrom L, Glimelius B. Chemotherapy in gas-
tric cancer: a review and updated meta-analysis. Eur J Surg
2002; 168: 597-608
3 Hu JK, Chen ZX, Zhou ZG, Zhang B, Tian J, Chen JP, Wang L,
Wang CH, Chen HY, Li YP. Intravenous chemotherapy for
resected gastric cancer: meta-analysis of randomized controlled
trials. World J Gastroenterol 2002; 8: 1023-1028
4 Yao JC, Ajani JA. Adjuvant and preoperative chemotherapy
for gastric cancer. Curr Oncol Rep 2002; 4: 222-228
5 Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between
cancer genetics and chemotherapy. Cell 2002; 108: 153-164
6 Szostak MJ, Kyprianou N. Radiation-induced apoptosis: pre-
dictive and therapeutic significance in radiotherapy of pros-
tate cancer. Oncol Rep 2000; 7: 699-706
7 Schulze PC, Adams V, Busert C, Bettag M, Kahn T, Schober R.
Effects of laser-induced thermotherapy (LITT) on proliferation
and apoptosis of glioma cells in rat brain
transplantation
tumors. Lasers Surg Med 2002; 30: 227-232
8 Du C, Fang M, Li Y, Li L,Wang X. Smac, a mitochondrial
protein that promotes cytochrome c-dependent caspase acti-
vation by eliminating IAP inhibition. Cell 2000; 102: 33-42
9 McNeish IA, Bell S, McKay T, Tenev T, Marani M, Lemoine
NR. Expression of Smac/DIABLO in ovarian carcinoma cells
induces apoptosis via a caspase-9-mediated pathway. Exp
Cell Res 2003; 286: 186-198
10 Jia L, Patwari Y, Kelsey SM, Srinivasula SM, Agrawal SG,
Alnemri ES, Newland AC. Role of Smac in human leukaemic
cell apoptosis and proliferation. Oncogene 2003; 22: 1589-1599
11 Marcus SG, Cohen D, Lin K, Wong K, Thompson S, Rothberger
A, Potmesil M, Hiotis S, Newman E. Complications of gastrec-
tomy following CPT-11-based neoadjuvant chemotherapy for
gastric cancer. J Gastrointest Surg 2003; 7: 1015-1022
12 Schulze-Bergkamen H, Krammer PH. Apoptosis in cancer-
implications for therapy. Semin Oncol 2004; 31: 90-119
13 Brown JM, Wilson G. Apoptosis genes and resistance to cancer
therapy: what does the experimental and clinical data tell us?
Cancer Biol Ther 2003; 2: 477-490
14 Kasibhatla S, Tseng B. Why target apoptosis in cancer
treatment? Mol Cancer Ther 2003; 2: 573-580
15 Westphal S, Kalthoff H. Apoptosis: targets in pancreatic
cancer. Mol Cancer 2003; 2: 6
16 Reed JC. Apoptosis-targeted therapies for cancer. Cancer Cell
2003; 3: 17-22
17 Sun SY, Hail N Jr, Lotan R. Apoptosis as a novel target for
cancer chemoprevention. J Natl Cancer Inst 2004; 96: 662-672
18 Derradji H, Baatout S. Apoptosis: a mechanism of cell suicide.
In Vivo 2003; 17: 185-192
19 Debatin KM. Apoptosis pathways in cancer and cancer
therapy. Cancer Immunol Immunother 2004; 53: 153-159
20 Shi Y. Mechanisms of caspase activation and inhibition during
apoptosis. Mol Cell 2002; 9: 459-470
21 Gulbins E, Dreschers S, Bock J. Role of mitochondria in
apoptosis. Exp Physiol 2003; 88: 85-90
22 Hockenbery DM, Giedt CD, O’Neill JW, Manion MK, Banker
DE. Mitochondria and apoptosis: new therapeutic targets. Adv
Cancer Res 2002; 85: 203-242
23 Martin SJ. Destabilizing influences in apoptosis: sowing the
seeds of IAP destruction. Cell 2002; 109: 793-796
24 Liston P, Fong WG, Korneluk RG. The inhibitors of apoptosis:
there is more to life than Bcl2. Oncogene 2003; 22: 8568-8580
25 Lotocki G, Keane RW. Inhibitors of apoptosis proteins in in-
jury and disease. IUBMB Life 2002; 54: 231-240
26 Vaziri SA, Grabowski DR, Tabata M, Holmes KA, Sterk J,
Takigawa N, Bukowski RM, Ganapathi MK, Ganapathi R. c-
IAP1 is overexpressed in HL-60 cells selected for doxorubicin
resistance: effects on etoposide-induced apoptosis. Anticancer
Res 2003; 23: 3657-3661
27 Altieri DC. Survivin, versatile modulation of cell division and
apoptosis in cancer. Oncogene 2003; 22: 8581-8589
28 Notarbartolo M, Cervello M, Dusonchet L, Cusimano A,
D’Alessandro N. Resistance to diverse apoptotic triggers in
multidrug resistant HL60 cells and its possible relationship to
the expression of P-glycoprotein, Fas and of the novel anti-
apoptosis factors IAP (inhibitory of apoptosis proteins). Can-
cer Lett 2002; 180: 91-101
29 Verhagen AM, Vaux DL. Cell death regulation by the mamma-
lian IAP antagonist Diablo/Smac. Apoptosis 2002; 7: 163-166
30 Vaux DL, Silke J. Mammalian mitochondrial IAP binding
proteins. Biochem Biophys Res Commun 2003; 304: 499-504
31 Guo F, Nimmanapalli R, Paranawithana S, Wittman S, Griffin
D, Bali P, O’Bryan E, Fumero C, Wang HG, Bhalla K. Ectopic
overexpression of second mitochondria-derived activator of
caspases (Smac/DIABLO) or cotreatment with N-terminus of
Smac/DIABLO peptide potentiates epothilone B derivative-
(BMS 247550) and Apo-2L/TRAIL-induced apoptosis. Blood
2002; 99: 3419-3426
32 Fulda S, Wick W, Weller M, Debatin KM. Smac agonists sensi-
tize for Apo2L/TRAIL- or anticancer drug-induced apoptosis
and induce regression of malignant glioma in vivo. Nat Med
2002; 8: 808-815
Assistant Editor Guo SY
Edited by Zhang JZ and Wang XL
Zheng LD
et al. Smac enhances chemotherapeutic effects 83
... Previous studies have indicated that decreased levels of Smac/DIABLO correlate with cancer progression. Smac/DIABLO is considered a potent therapeutic target (8)(9)(10)(11). However, the clinical significance of Smac/DIABLO in various cancers remains unclear. ...
... Correlation between Smac/DIABLO expression and clinicopathological parameters in gastric adenocarcinomas. Clinicopathological -------------------------------------parameters Total, Smac/DIABLO expression is associated with a good prognosis and low tumor stage (8)(9)(10)(11). In the present study, a trend towards an association between decreased Smac/DIABLO expression and pathological stage in gastric and colorectal carcinomas was observed, however, no statistically significant difference was identified. ...
Article
Full-text available
Lack of apoptosis is a key factor in carcinogenesis and tumor progression. Survivin is a member of the inhibitor of apoptosis protein (IAP) family. Second mitochondria-derived activator of caspases/direct inhibitor of apoptosis-binding protein with low pI (Smac/DIABLO) is an antagonist of IAPs. Recently, Smac/DIABLO was identified as a potent therapeutic target. However, the clinical significance of Smac/DIABLO in gastrointestinal carcinomas remains unclear. In the present study, Smac/DIABLO expression was analyzed by immunohistochemistry in 72 gastric adenocarcinomas and 78 colorectal adenocarcinomas. The expression of Smac/DIABLO was significantly higher in colorectal carcinoma than in gastric carcinoma. Additionally, a correlation was found between the expression of Smac/DIABLO and nuclear survivin in well- to moderately-differentiated colorectal adenocarcinomas (r=0.245; P<0.01). Based on these results, it was hypothesized that gastric and colorectal carcinomas differ in the level of Smac/DIABLO expression. Our previous studies revealed that the expression of cleaved caspase-9 was significantly lower in colorectal carcinoma than in gastric carcinoma (P<0.0001). Conversely, the expression levels of microtubule-associated protein 1 light chain 3 (LC3), an autophagy marker, and survivin were significantly higher in colon cancer than in gastric cancer (P<0.0001 and P<0.01, respectively). Taken together, these results indicate that not only LC3 and survivin expression, but also Smac/DIABLO expression, are significantly higher in colorectal carcinoma than in gastric carcinoma. We hypothesize that the analysis of Smac/DIABLO, survivin and LC3 expression in colorectal carcinoma is likely to aid cancer therapy due to the involvement of these markers in apoptosis and/or autophagy.
... The level of expression of SMAC and its role in treatment sensitization has been studied in relation to several types of cancers. SMAC levels were seen to correlate well with survival in lung cancer (19), while overexpression of it increased chemosensitivity in gastric cancer cells (20), hepatocellular carcinoma (21) and osteosarcoma (22). Low levels of SMAC resulted in early resistance to chemotherapy in thyroid cancer (23) and its expression was downregulated in renal cell carcinoma, predicting a poor prognosis (24). ...
Article
Full-text available
Second mitochondria-derived activator of caspases/direct inhibitor of apoptosis-binding protein with low pI (Smac/DIABLO) is released by mitochondria in response to apoptotic stimuli and is thought to regulate apoptosis by antagonizing inhibitors of apoptosis proteins, which play an important role in sensitization of cancer cells to various therapeutic regimens. The expression of Smac/DIABLO has been demonstrated in various cancer cells, though little is known about its clinical significance with respect to colorectal cancer. The current study was designed to evaluate the relationship between prognosis and Smac/DIABLO expression by clinicopathological analysis of patients with colorectal cancer. Smac/DIABLO expression was evaluated using immunohistochemical staining in 121 consecutive patients with colorectal cancer and the relationship between Smac/DIABLO expression and clinicopathological factors was analyzed. Smac/DIABLO-positive expression was detected in 80 of the 121 patients (66%). The incidence of lymph node and distant metastasis in Smac/DIABLO-negative cancer was significantly higher than that in Smac/DIABLO-positive cancer (P=0.0004 and P=0.003, respectively). While univariate analysis showed that survival in patients with Smac/DIABLO-negative expression was significantly poorer than in Smac/DIABLO-positive cases (P<0.0001), Smac/DIABLO-negative expression was a prognostic indicator independent of Dukes' staging and lymph node metastasis by multivariate analysis. This study proposes that the decrease of Smac/DIABLO expression is an independent factor determining the poorer prognosis of patients with colorectal cancer.
Data
Full-text available
Effects of the histone-deacetylases inhibitor trichostatin A (TSA) on the growth of three different human pancreatic endocrine carcinoma cell lines (CM, BON, and QGP-1) have been assessed via dosage-dependent growth inhibition curves. TSA determined strong inhibition of cell growth with similar IC50 values for the different cell lines: 80.5 nM (CM), 61.6 nM (BON), and 86 nM (QGP-1), by arresting the cell cycle in G2/M phase and inducing apoptosis. 2DE and nano-RP-HPLC-ESI-MS/MS analysis revealed 34, 33, and 38 unique proteins differentially expressed after TSA treatment in the CM, BON, and QGP-1 cell lines, respectively. The most important groups of modulated proteins belong to cell proliferation, cell cycle, and apoptosis classes (such as peroxiredoxins 1 and 2, the diablo protein, and HSP27). Other proteins pertain to processes such as regulation of gene expression (nucleophosmin, oncoprotein dek), signal transduction (calcium-calmodulin), chromatin, and cytoskeleton organization (calgizzarin, dynein, and lamin), RNA splicing (nucleolin, HNRPC), and protein folding (HSP70). The present data are in agreement with previous proteomic analyses performed on pancreatic ductal carcinoma cell lines (Cecconi, D. et al.., Electrophoresis 2003; Cecconi, D. et al., J. Proteome Res. 2005) and place histone-deacetylases inhibitors among the potentially most powerful drugs for the treatment of pancreatic tumors.
Article
The effect of Smac gene on the TRAIL-induced apoptosis of the prostate cancer cell line PC-3 and the molecular mechanism were investigated. The Smac gene was transfected into PC-3 cells under the induction of liposome. The intrinsic Smac gene expression was detected by Western blotting. After treatment with TRAIL as an apoptosis inducer, in vitro cell growth activity was assayed by MTT colorimetry. The apoptosis rate of PC-3 cells was determined by annexin V-FITC and propidium iodide staining flow cytometry. The expression of cellular XIAP and caspase-3 genes was examined by Western blotting. Smac-transfected cells (PC-3/Smac group) had significantly increased Smac protein level as compared with PC-3 controls (P<0.01). After induction with 100-200 ng/mL TRAIL for 12-36 h, cellular proliferation rate in PC-3/Smac group was significantly lower than in PC-3 controls (P<0.05). After induction with 100 ng/mL TRAIL for 24 h, the apoptosis rate in PC-3/Smac group was significantly enhanced as compared with that of PC-3 controls (P<0.05). Accordingly, the XIAP expression level was down-regulated significantly (P<0.05) and caspase-3 subunit P20 was up-regulated significantly (P<0.05). It is suggested that the over-expression of cellular Smac can inhibit inhibitor of apoptosis proteins (IAPs), enhance caspases activity and the apoptosis rate of PC-3 cells induced by TRAIL, which may provide a useful experimental basis for prostate cancer therapy.
Article
The recent discovery of Smac and the elucidation of its structure and function have led to the rapid development of Smac mimetics, comprising Smac derivative and mimicking molecules, for use in cancer treatment. Smac is an endogenous proapoptotic protein that resides in the mitochondria and is released when a cell is triggered to undergo programmed cell death. One of the mechanisms by which Smac promotes apoptosis is through its ability to inhibit inhibitors of apoptosis (IAPs), by direct inhibition and/or proteasomal degradation of some members of the IAP family, and therefore disinhibit caspases. Thus, the use of Smac mimetics as anticancer agents follows a rational approach in cancer therapeutics. This approach directly targets dysregulated, neoplastic cells that overexpress IAPs or underexpress Smac. Although Smac mimetics are able to elicit an anticancer response when used alone, these molecules can also function effectively and synergistically when combined with other therapeutic agents. A variety of Smac mimetic types comprising peptides, polynucleotides, and compounds have been studied both in vitro and in vivo. This discussion addresses the current status of Smac mimetics in cancer research.
Article
Our research group recently reported that pancreatic endocrine cancer cell lines are sensitive to the HDAC inhibitor trichostatin A (TSA). In the present paper, we show that the combined treatment of pancreatic endocrine tumour cell lines with TSA and the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (DAC) determines a strong synergistic inhibition of proliferation mainly due to apoptotic cell death. Proteomic analysis demonstrates that the modulation of specific proteins correlates with the antiproliferative effect of the drugs. A schematic network clarifies the most important targets or pathways involved in pancreatic endocrine cancer growth inhibition by single or combined drug treatments, which include proteasome, mitochondrial apoptotic pathway and caspase related proteins, p53 and Ras related proteins. A comparison between the patterns of proteins regulated by TSA or DAC in endocrine and ductal pancreatic cancer cell lines is also presented.
Article
Dysregulation of apoptosis is a key factor in carcinogenesis and tumor progression. X-linked inhibitor of apoptosis (XIAP) is the most potent member of the inhibitor of apoptosis protein (IAP) family, which directly inhibits apoptosis by binding to caspases. Antagonists of XIAP have recently been identified: second mitochondria-derived activator of caspase/direct IAP-binding protein with low PI (Smac/DIABLO) and XIAP-associated factor 1 (XAF1). However, little research has been conducted on the association between gastric cancer survival and the mechanism of apoptosis involving XIAP and its antagonists, Smac/DIABLO and XAF1. XIAP, Smac/DIABLO, and XAF1 expression was analyzed by immunohistochemistry (IHC) in 187 gastric adenocarcinomas. Correlations between XIAP, Smac/DIABLO or XAF1 expression and clinicopathological factors were analyzed. Disease-specific survival after surgery was examined. Of 187 samples, XIAP was overexpressed in 140, Smac was overexpressed in 117, and XAF1 was overexpressed in 106. Individually, XIAP, Smac, and XAF1 were not significantly associated with disease-specific survival. However, patients showing high expression of XIAP and low expression of XAF1 had significantly poorer survival when compared with other groups (P = 0.024). The expression balance of XIAP and XAF1 is an independent prognostic factor in gastric adenocarcinoma.
Article
Smac/DIABLO is a mitochondrial protein that participates in apoptotic cell death by means of sequestering several members of the inhibitor of apoptosis protein family. This action allows caspase activation, cleavage of key cellular substrates and death. Release from mitochondria is considered the main regulatory step of Smac/DIABLO activity. Nevertheless, the fact that at least one isoform, Smac-beta, does not reside in this organelle implies that transcriptional regulation could also be important. cAMP is a well known second messenger with important apoptotic effects. To analyze if cAMP could be involved in Smac/DIABLO gene regulation, we analyzed 2903 base pairs upstream of the coding sequence and characterized the minimal promoter, which contains a consensus CRE site. We found that cAMP/PKA/CREB pathway is indeed an important regulator of Smac/DIABLO transcription, since exposure to the cAMP analog 8-CPT-cAMP, the adenylyl cyclase activator forskolin, the inhibitor of phosphodiesterase isobutylmethylxanthine or by hindering PKA activation with H89, regulated the promoter activity, as shown by gene reporter and RT-PCR assays. Additionally, the results of site-directed mutagenesis revealed that the consensus CRE site was biologically functional and required for cAMP-induced promoter activity in human HeLa cells. Supporting these results, a negative dominant version of the protein kinase A responsive factor, KCREB, reduced basal Smac/DIABLO expression and rendered the promoter unresponsive to cAMP. Reducing Smac expression using an antisense approach blocked the apoptosis effects of cAMP in cervical cancer cells. These results show that cAMP is an important modulator of the apoptotic threshold in cancer cell by means of regulating Smac/DIABLO expression.
Article
Effects of the histone-deacetylases inhibitor trichostatin A (TSA) on the growth of three different human pancreatic endocrine carcinoma cell lines (CM, BON, and QGP-1) have been assessed via dosage-dependent growth inhibition curves. TSA determined strong inhibition of cell growth with similar IC(50) values for the different cell lines: 80.5 nM (CM), 61.6 nM (BON), and 86 nM (QGP-1), by arresting the cell cycle in G2/M phase and inducing apoptosis. 2DE and nano-RP-HPLC-ESI-MS/MS analysis revealed 34, 33, and 38 unique proteins differentially expressed after TSA treatment in the CM, BON, and QGP-1 cell lines, respectively. The most important groups of modulated proteins belong to cell proliferation, cell cycle, and apoptosis classes (such as peroxiredoxins 1 and 2, the diablo protein, and HSP27). Other proteins pertain to processes such as regulation of gene expression (nucleophosmin, oncoprotein dek), signal transduction (calcium-calmodulin), chromatin, and cytoskeleton organization (calgizzarin, dynein, and lamin), RNA splicing (nucleolin, HNRPC), and protein folding (HSP70). The present data are in agreement with previous proteomic analyses performed on pancreatic ductal carcinoma cell lines (Cecconi, D. et al.., Electrophoresis 2003; Cecconi, D. et al., J. Proteome Res. 2005) and place histone-deacetylases inhibitors among the potentially most powerful drugs for the treatment of pancreatic tumors.
Article
Dysregulation of apoptosis plays an important role in carcinogenesis and tumor progression. Whereas x-linked inhibitor of apoptosis (XIAP) is a potent inhibitor of apoptosis, its antagonists second mitochondria-derived activator of caspases/direct IAP binding protein with low PI (Smac/DIABLO), and XIAP-associated factor 1 (XAF1) promote apoptosis. To explore the relevance of XIAP, Smac/DIABLO, and XAF1 for carcinogenesis and tumor progression, we analyzed 46 primary gastric adenocarcinomas and non-neoplastic gastric mucosa samples by quantitative real-time polymerase chain reaction. XIAP, Smac/DIABLO, and XAF1 expression was found in all non-neoplastic gastric mucosa samples and all adenocarcinomas. XIAP expression levels did not change between non-neoplastic gastric mucosa and adenocarcinomas or between carcinomas of early and advanced stages. Although Smac/DIABLO expression was significantly (P=0.01) higher in carcinomas, the ratio of XIAP to Smac/DIABLO expression remained stable between non-neoplastic mucosa and carcinomas. XAF1 expression had the tendency to decrease from non-neoplastic mucosa to advanced adenocarcinomas. Importantly, the ratio of XIAP to XAF1 expression significantly (P=0.03) increased from non-neoplastic mucosa to adenocarcinomas and the increase was even higher in carcinomas of advanced stage (P=0.01). Moreover, expression of the XAF1 splice variants differing in the zinc-finger domain essential for XIAP-binding was analyzed and revealed a significant higher (P=0.03) variant-2/variant-1 ratio in advanced carcinomas. In conclusion, an increased expression ratio of XIAP to XAF1 in combination with a disturbed expression of the XAF1 splice variants could be shown in gastric adenocarcinomas. These marked imbalances probably result in an impaired ability for XAF1 to antagonize the effects of XIAP thereby contributing to apoptosis-resistance and generating an important growth advantage.
Article
Full-text available
AIM: To assess the safety and efficacy of different intravenous chemotherapeutic regimens in patients with gastric carcinomas who had undergone gastrectomy. METHODS: A meta-analysis of all the relevant randomized controlled trials (RCTs) was performed. Language was restricted to Chinese and English. RCTs were identified from Medline and Embase (1980-2001/4), and Chinese Bio-medicine Database (1990-2001/1). Literature references were checked at the same time. We included randomized and quasi-randomized trials comparing the efficacy of intravenous chemotherapy after gastrectomy with that of surgery alone in patients with confirmed gastric carcinomas who had undergone gastrectomy. Selection criteria were: randomized or quasi-randomized trials with following-up results; Trials could be double-blind, single-blind or not blind; Chemotherapy groups were given intravenous chemotherapy after gastrectomy without neo-adjuvant chemotherapy, intraperitoneal hyperthermic perfusion, radiotherapy or chemoimmunotherapy; Controlled group included those receiving gastrectomy alone. The following data were extracted: the number of survival and death by the end of the follow-up; the different agents and doses of the intravenous chemotherapy; the baseline of the chemotherapy group and the controlled arm; the serious adverse events; the statistical consideration; cost-effectiveness analysis. The statistical analysis was performed by RevMan4.1 software which was provided by the Cochrane Collaboration. A P value of < 0.05 was considered statistically significant. Meta-analysis was done with random effects model. Heterogeneity was checked by chi-square test. Sensitivity analysis was performed by excluding the trials in which Jadad-scale was only 1 score. The result was expressed with odds ratio (OR) for the categorical variable. RESULTS: Fourteen trials involving 4543 patients were included. Meta-analysis was done with random effects model Heterogeneity and sensitivity analysis were performed also. The effect of intravenous chemotherapy after gastrectomy was better than surgery alone (odds ratio 0.56, 95%CI 0.40-0.79). There was a significant difference between the two groups by u-test (P = 0.0008). Sensitivity analysis revealed the same difference (odds ratio 0.81, 95%CI 0.70-0.94). Of fourteen trials, only three studies were of high quality according to the Jadad-scale (with three score). There was one meta-analysis trial and the others, about ten trials, were of low quality. There was no trial which mentioned sample-size calculation, allocation concealment, intention-to-treat analysis. Most of the trials didn’t describe the blind-procedure. There were five trials which detailed the side-effects according to the toxicity grade by WHO standard. The side-effects halting treatment were haematologic and biochemical toxicity, debilitating nausea and vomiting. There were two patients died of chemotherapy toxicity. CONCLUSION: Based on the review, intravenous chemotherapy after gastrectomy may have positive treatment effect on gastric cancer. However, the evidence is not strong because of the general low methodologic quality of the RCTs. Therefore, we can’t make the conclusion that intravenous chemotherapy after gastrectomy may have better treatment effect on gastric cancer than that of surgery alone. Rigorously designed, randomised, double-blind, placebo-controlled trials are required.
Article
Full-text available
Smac (or DIABLO) is a recently identified, novel proapoptotic molecule, which is released from mitochondria into the cytosol during apoptosis. Smac functions by eliminating the caspase-inhibitory properties of the inhibitors of apoptosis proteins (IAP), particularly XIAP. In this study, we stably transfected both full-length (FL) and mature (MT) Smac genes into the K562 and CEM leukaemic cell lines. Both FL and MT Smac transfectants increased the sensitivity of leukaemic cells to UV light-induced apoptosis and the activation of caspase-9 and caspase-3. Purified cytosol from the mature Smac transfectants, or the addition of human recombinant Smac protein or N-7 peptide into nontransfected cytosol, showed an increased sensitivity to cytochrome c-induced activation of caspase-3. The mature Smac enhanced the susceptibility of both K562 and CEM cells to TRAIL-induced apoptosis. Overexpression of the mature Smac protein also inhibited proliferation, as detected by reduced colony formation and Ki-67 expression in leukaemic cells. Cell cycle analysis revealed that Smac transfectants displayed significant G0/G1 arrest and reduction in 5-bromo-2'-deoxyuridine (BrdU) incorporation. Smac sensitized human acute myeloid leukaemia blasts to cytochrome c-induced activation of caspase-3. However, Smac failed to overcome Apaf-1-deficiency-mediated resistance to cytochrome c in primary leukaemic blasts. In summary, this study reveals that Smac/DIABLO exhibits a potential role in increasing apoptosis and suppressing proliferation in human leukaemic cells. Importantly, it also indicates that it is crucial to evaluate the levels of Apaf-1 and XIAP proteins in patient samples before using Smac peptide therapy in the treatment of human leukaemia.Keywords: apoptosis, Apaf-1, caspases, IAPs, Smac
Article
Potential benefits of neoadjuvant therapy for locally advanced gastric cancer include tumor downstaging and an increased R0 resection rate. Potential disadvantages include increased surgical complications. This study assesses postoperative morbidity and mortality by comparing patients undergoing gastrectomy with and without neoadjuvant chemotherapy. From October 1998 to July 2002, a total of 34 patients with locally advanced gastric cancer were placed on a phase II neoadjuvant chemotherapy protocol consisting of two cycles of CPT-11 (75 mg/m2) with cisplatin (25 mg/m2). Demographic, clinical, morbidity, and mortality data were compared for these patients (CHEMO) versus 85 patients undergoing gastrectomy without neoadjuvant chemotherapy (SURG). The CHEMO patients were more likely to be less than 70 years of age (P≤0.01), have proximal tumors (P≤0.01), and undergo proximal gastrectomy (P≤0.025). Fifty-two percent of SURG patients had T3/T4 tumors compared to 19% of CHEMO patients, consistent with tumor downstaging. The R0 resection rate was similar (80%). Morbidity was 41% in CHEMO patients and 39% in SURG patients. There were five postoperative deaths (4.4%), two in the CHEMO group and three in the SURG group (P = NS). It was concluded that neoadjuvant chemotherapy with CPT-11 and cisplatin is not associated with increased postoperative morbidity compared to surgery alone. CPT-11–based neoadjuvant chemotherapy should be tested further in combined-modality treatment of gastric cancer.
Article
Activation of apoptosis pathways is a key mechanism by which cytotoxic drugs kill tumor cells. Also immunotherapy of tumors requires an apoptosis sensitive phenotype of target cells. Defects in apoptosis signalling contribute to resistance of tumors. Activation of apoptosis signalling following treatment with cytotoxic drugs has been shown to lead to activation of the mitochondrial (intrinsic) pathway of apoptosis. In addition, signalling through the death receptor (extrinsic) pathways, contributes to sensitivity of tumor cells towards cytotoxic treatment. Both pathways converge finally at the level of activation of caspases, the effector molecules in most forms of cell death. In addition to classical apoptosis, non-apoptotic modes of cell death have recently been identified. Mechanisms to overcome apoptosis resistance include direct targeting of antiapoptotic molecules expressed in tumors as well as re-sensitization of previously resistant tumor cells by re-expression of caspases and counteracting apoptotis inhibitory molecules such as Bcl-2 and molecules of the IAP family of endogenous caspase inhibitors. Molecular insights into regulation of apoptosis and defects in apoptosis signalling in tumor cells will provide novel approaches to define sensitivity or resistance of tumor cells towards antitumor therapy and provide new targets for rational therapeutic interventions for future therapeutic strategies.
Article
Current therapy for advanced prostate cancer is hampered by the propensity of the disease to progress from an androgen-dependent state to an androgen-independent state. Current treatment for advanced disease is palliative. Therefore, the therapeutic goal for prostate cancer treatment today is to arrest the disease at an early state when it is still localized to the gland. The standard treatment for clinically localized disease is radical prostatectomy or radiation therapy by way of external beam irradiation or local radioactive seed implants (brachytherapy). In advanced disease, the use of radiation therapy is limited to palliation of pain secondary to bone metastases and for spinal cord compression. Tracking residual disease and predicting outcome is limited to following the level of prostate specific antigen (PSA) production, evaluating for bone or solid organ metastasis, and analyzing their preoperative clinical stage, PSA and Gleason's score. Apoptosis as a molecular process of genetically regulated cell death has a critical endpoint that coincides with the goal of successful treatment of human malignancies. Since in cancer treatment the therapeutic goal is to trigger tumor-selective cell death, activation of the apoptotic pathway in prostatic tumor cells offers attractive and potentially effective therapeutic targets. As our understanding of the vital role of apoptosis in the development and growth of the prostate gland has expanded, numerous genes that encode apoptotic regulators have been identified that are severely impaired in prostate tumors. Human prostate cancer cells undergo apoptosis in response to androgen ablation, chemotherapeutic agents and ionizing irradiation. The expression of apoptotic modulators within individual prostate tumors appears to correlate with the cancer cell's sensitivity to traditional therapeutic modalities, including radiotherapy. No strict correlation between radiation-induced apoptosis and longevity of prostate cancer patients has emerged, possibly because the ability to achieve an initial remission alone does not adequately predict long-term outcome and patient survival. In this review we summarize the current understanding of the effects of radiation therapy on prostatic tumor cells within the context of the therapeutic significance of radiation-induced apoptosis in the effective elimination of androgen independent prostate cancer cells. As we enter a new millenium, identification of distinct molecular markers predictive of therapeutic response of prostatic tumors to radiation therapy may afford alternative prognostic indicators in optimizing our treatment protocols for advanced disease.
Article
We report here the identification of a novel protein, Smac, which promotes caspase activation in the cytochrome c/Apaf-1/caspase-9 pathway. Smac promotes caspase-9 activation by binding to inhibitor of apoptosis proteins, IAPs, and removing their inhibitory activity. Smac is normally a mitochondrial protein but is released into the cytosol when cells undergo apoptosis. Mitochondrial import and cleavage of its signal peptide are required for Smac to gain its apoptotic activity. Overexpression of Smac increases cells' sensitivity to apoptotic stimuli. Smac is the second mitochondrial protein, along with cytochrome c, that promotes apoptosis by activating caspases.
Article
Defects in apoptosis underpin both tumorigenesis and drug resistance, and because of these defects chemotherapy often fails. Understanding the molecular events that contribute to drug-induced apoptosis, and how tumors evade apoptotic death, provides a paradigm to explain the relationship between cancer genetics and treatment sensitivity and should enable a more rational approach to anticancer drug design and therapy.
Article
In Drosophila, the genetic locus 75CI1,2 is essential for all developmental cell death. Within this region are the genes for three pro-death proteins, Grim, Reaper and HID. These proteins are transcriptionally regulated and their expression tightly associated with cell death in the developing fly embryo. When ectopically expressed in the retina, Grim, Reaper and HID cause apoptosis and eye ablation. They have a short region of similarity at their N-termini through which they can interact with inhibitor of apoptosis (IAP) proteins, and it is by antagonising IAP inhibition of caspases that Grim, Reaper and HID promote cell death. The observation that Grim, Reaper and HID can interact with mammalian IAPs and induce apoptosis in mammalian cells suggested that mammalian IAP antagonists might also exist. Diablo/Smac, identified six years after the first description of a Drosophila IAP antagonist, is the only mammalian protein identified to date that is clearly functionally related to the Drosophila proteins. Since its discovery, there have been numerous studies investigating how Diablo/Smac interacts with IAPs and promotes cell death. Here we review what is currently known about Diablo/Smac and speculate on other mammalian IAP antagonists.
Article
Laser-induced thermotherapy (LITT) is an approach to the treatment of brain tumors especially in poorly accessible regions. Its clinical applicability with tumor cell destruction has been shown in several studies. However, no data are known about specific effects on tumors cells due to LITT in the time course of the lesion. LITT was performed in adult Lewis rats with implanted glioma cells in the brain using a standard exposure of 3 W for 30 seconds. Before and following LITT, neoplastic lesions were monitored by MRI. Proliferation of implanted cells and gliosis were assessed by several histological techniques and immunohistochemistry. Apoptosis was detected by TUNEL staining. Our experiments show a destruction of neoplastic cells by LITT but surviving tumor cells at the margin of the lesion. Apoptosis was detected following LITT restricted to residual neoplastic cells. Marginal survival of tumor cells lead to a secondary outgrowth into the necrotic lesion adjacent to sprouting capillaries. LITT is a suitable technique for the treatment of brain neoplasms. However, further investigations are necessary to prevent tumor recurrences after LITT.
Article
We studied the human HL60 leukemia cell line and its multidrug resistant (MDR) variant HL60R. In contrast to the HL60, HL60R showed an inability to undergo apoptosis from doxorubicin (Dox) or other different stimuli, including cisplatin, Fas ligation and serum withdrawal. HL60R cells lost surface Fas expression, but we found no evidence that Fas/FasL mediates the apoptotic effects of Dox in HL60. P-glycoprotein (P-gp) did not seem to play a major role as a specific inhibitor of apoptosis. In fact, the P-gp inhibitor verapamil reversed only partially the resistance to Dox-induced apoptosis of the MDR cells. In addition, it did not modify the rate of apoptosis induced from the other stimuli in the same cells. The expression of p53 or Bcl-2 was not different between HL60 and HL60R. However, in HL60R there was an increase in the mRNAs of inhibitory of apoptosis proteins (IAPs) like neuronal apoptosis inhibitory protein (NAIP), c-IAP-2 and survivin. Treatment with Dox or serum starvation strongly down-regulated X-linked IAP and survivin mRNAs in HL60. Cisplatin decreased NAIP and survivin mRNAs in the same cells. However, in HL60R the levels of these IAP mRNAs were much less affected by the treatments. These results support that IAPs may be involved in tumor resistance to chemotherapeutic drugs or other apoptotic agents.