ArticlePDF Available

CADPE Inhibits PMA-stimulated gastric carcinoma cell invasion and matrix metalloproteinase-9 expression by FAK/MEK/ERK-mediated AP-1 activation

Authors:

Abstract and Figures

Metastasis is one of the main causes of death for patients with malignant tumors. Aberrant expression of matrix metalloproteinase-9 (MMP-9) has been implicated in the invasion and metastasis of various cancer cells. Here, we found that caffeic acid 3,4-dihydroxy-phenethyl ester (CADPE) could inhibit the migration and invasion of human gastric carcinoma cells in Transwell migration assays. To understand the underlying mechanism, we showed that CADPE significantly inhibited phorbol 12-myristate 13-acetate (PMA)-induced increases in MMP-9 expression and activity in a dose-dependent manner. The inhibitory effect of CADPE on MMP-9 expression correlated well with the suppression of MMP-9 promoter activity and the reduction of MMP-9 mRNA. Reporter gene assay and electrophoretic mobility shift assay showed that CADPE inhibited MMP-9 expression by suppressing the activation of the nuclear transcription factor activator protein-1 (AP-1) and c-Fos, but not NF-κB. Moreover, CADPE inhibited PMA-induced phosphorylation of protein kinases involved in AP-1 activation, such as focal adhesion kinase (FAK), mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK), and ERK1/2, whereas CADPE had little effect on the phosphorylation of p38 and c-jun NH(2)-terminal kinase. Taken together, our findings indicate that CADPE could be a unique antitumor agent that specifically inhibits MMP-9 activity by targeting the activation of FAK/MEK/ERK protein kinases and AP-1 transcription factor.
Chemical structures of CADPE and CA and the effects of CADPE on the viability, migration, and invasion of MGC-803 cells. A, chemical structures of CA and CADPE. B, effect of CADPE on cell viability. MGC-803 cells were treated with 5 to 20 μmol/L CADPE in serum or serum-free medium for 24 h. Cell viability was determined by MTS assay. The percentage of cell viability was calculated as the ratio (A 570 ) of treated cells to control cells. Points, mean of three independent experiments; bars, SE. C, CADPE inhibits tumor cell migration. The Transwell migration assay was carried out in MGC-803 cells. After 8 h of incubation with or without the indicated concentration of CADPE, cells that migrated to the low chamber were fixed, stained, and counted by light microscopy as described in Materials and Methods (left). Random fields were scanned (four fields per filter of the well) for the presence of cells on the lower side of the membrane (right image). Columns, mean number of migrated cells in three wells from three independent experiments; bars, SE. *, P < 0.05. D, the effect of CADPE on cell invasion was determined using the Matrigel invasion assay system. For invasion assay, the upper chamber was coated with Matrigel, and MGC-803 cells with various concentrations of CADPE in the presence of 100 nmol/L PMA were added. After 24 h, cells on the bottom side of filter were fixed, stained, and counted under a light microscope (D, left images). The inhibitory effects of CADPE on two additional gastric cancer cell lines, HGC-27 and AGS, were also examined (D, right columns). Columns, mean number of migrated cells in three wells from three independent experiments; bars, SE. *, P < 0.05.
… 
Content may be subject to copyright.
Angiogenesis, Metastasis, and the Cellular Microenvironment
CADPE Inhibits PMA-Stimulated Gastric Carcinoma Cell
Invasion and Matrix Metalloproteinase-9 Expression by
FAK/MEK/ERKMediated AP-1 Activation
Honghui Han, Bing Du, Xinhua Pan, Junchen Liu, Qufei Zhao, Xiaoyuan Lian, Min Qian, and Mingyao Liu
Abstract
Metastasis is one of the main causes of death for patients with malignant tumors. Aberrant expression of
matrix metalloproteinase-9 (MMP-9) has been implicated in the invasion and metastasis of various cancer cells.
Here, we found that caffeic acid 3,4-dihydroxy-phenethyl ester (CADPE) could inhibit the migration and in-
vasion of human gastric carcinoma cells in Transwell migration assays. To understand the underlying mecha-
nism, we showed that CADPE significantly inhibited phorbol 12-myristate 13-acetate (PMA)induced increases
in MMP-9 expression and activity in a dose-dependent manner. The inhibitory effect of CADPE on MMP-9
expression correlated well with the suppression of MMP-9 promoter activity and the reduction of MMP-9
mRNA. Reporter gene assay and electrophoretic mobility shift assay showed that CADPE inhibited
MMP-9 expression by suppressing the activation of the nuclear transcription factor activator protein-1
(AP-1) and c-Fos, but not NF-κB. Moreover, CADPE inhibited PMA-induced phosphorylation of protein
kinases involved in AP-1 activation, such as focal adhesion kinase (FAK), mitogen-activated protein kinase/
extracellular signalregulated kinase (ERK) kinase (MEK), and ERK1/2, whereas CADPE had little effect on
the phosphorylation of p38 and c-jun NH
2
-terminal kinase. Taken together, our findings indicate that CADPE
could be a unique antitumor agent that specifically inhibits MMP-9 activity by targeting the activation of FAK/
MEK/ERK protein kinases and AP-1 transcription factor. Mol Cancer Res; 8(11); 147788. ©2010 AACR.
Introduction
Gastric carcinoma is the second most common cause of
death and the third most common cancer worldwide (1).
The current overall 5-year survival figures for gastric cancers
in western patients are in the range of 5% to 17%. The inci-
dence is also high in Europe, South America, and Eastern Asia
(2-4). Because of its limited treatment efficiency and poor
prognosis, the therapeutics of gastric cancer remains a major
clinical challenge (5). Loss of control of tumor cell invasion
and metastasis is the main cause of death in gastric cancer
patients. The formation of metastatic nodules to gastric
carcinoma is a multistep and complex process that includes
cell proliferation, digestion of the extracellular matrix
(ECM), cell migration to circulation system or lymph nodes,
and remigration and growth of tumors at metastatic sites. It
is widely believed that the aberrant expression of matrix
metalloproteinases (MMP) is involved in these processes (6).
MMPs are well-known ECM-degrading enzymes, which
comprise a family of 24 members. Based on their sub-
strates, MMPs are divided into four subclasses: collagenase,
gelatinase, stromelysin, and membrane-associated MMPs
(7). As a main ECM-degrading enzyme family, MMPs have
essential roles in physiologic processes such as tissue devel-
opment, remodeling, and wound healing (8). However,
they are also involved in some tissue destructive diseases,
such as atherosclerosis; inflammation; rheumatoid arthritis;
and tumor invasion, metastasis, and neoangiogenesis
(9, 10). Recent studies showed that MMPs were important
regulators of the tumor microenvironment, including
tumor progression, metastatic niche formation, and inflam-
mation in cancer (11). Among human MMPs, MMP-2
(gelatinase-A) and MMP-9 (gelatinase-B) are key enzymes
in the degradation of type IV collagen, which is an impor-
tant component of ECM. These two members are mostly
associated with tumor migration, invasion, and metastasis
for various cancers (12). An enhanced expression of
MMP-9 has been shown to be associated with the progres-
sion and invasion of tumors, whereas MMP-2 is usually
expressed constitutively (13, 14).
Authors' Affiliation: The Institute of Biomedical Sciences and School of
Life Sciences, East China Normal University, Shanghai, China
Note: Supplementary data for this article are available at Molecular Can-
cer Research Online (http://mcr.aacrjournals.org/).
H. Han and B. Du contributed equally to this work.
Corresponding Authors: Mingyao Liu, The Institute of Biomedical
Sciences, East China Normal University, 500 Dongchuan Road, Shanghai
200241, China. Phone: 86-21-5434-5124; Fax: 86-21-5434-4922.
E-mail: myliu@bio.ecnu.edu.cn or Min Qian, East China Normal University
School of Life Science, 3663 North Zhongshan, Shanghai 2000 62, PR
China. Phone: 86-021-62233569; Fax: 011-86-021-62233754. E-mail:
mqian@bio.ecnu.edu.cn
doi: 10.1158/1541-7786.MCR-10-0114
©2010 American Association for Cancer Research.
Molecular
Cancer
Research
www.aacrjournals.org 1477
The expression of MMP-9 can be stimulated by various
agents, such as inflammatory cytokine, growth factor, and
phorbol myristate acetate (PMA). PMA is a well-known
inflammatory stimulator and tumor promoter that acti-
vates almost all protein kinase C (PKC) isozymes and in-
creases the invasiveness of various types of cancer cells by
activating MMP-9 (15). Those stimulators can upregulate
the expression of MMP-9 by modulating the activation of
transcription factors such as activator protein-1 (AP-1) and
NF-κB through the Ras/Raf/extracellular signal-regulated
kinase (ERK), c-jun NH
2
-terminal kinase (JNK), and
phosphoinositide 3-kinase/Akt signaling pathways
(16-19). AP-1 has been shown to regulate the expression
of a number of genes, some of which are involved in tu-
morigenesis (20, 21). Thus, it will be an effective way to
find tumorigenesis and metastasis inhibitors from the
agents that can suppress the activities of AP-1 and MMP-9.
Caffeic acid 3,4-dihydroxyphenethyl ester (CADPE) was
originally isolated from Teucrium pilosum as a substance
named teucrol in 2000 (22) and can be synthesized by a
chemical process (23). Caffeic acid (CA) is a phenolic com-
pound and is largely found in food plants. CA has been
reported to posses a wide spectrum of biological effects
(e.g., antioxidant, anti-inflammatory, antitumor angiogen-
esis and antitumor invasion, and metastasis properties; ref.
24-26). Recent studies showed that both CA and CADPE
could inhibit tumor angiogenesis in human renal carcino-
ma cells by suppressing hypoxia-induced signal transducer
and activator of transcription-3 phosphorylation, signal
transducer and activator of transcription-3 nuclear translo-
cation, hypoxia-inducible factor-1αinduction, and vascu-
lar endothelial growth factor expression (24). However, the
effect and related molecular mechanisms of CADPE in the
regulation of MMPs in cancer cells have not been reported.
In this study, human gastric carcinoma cell line (MGC-
803) was used to investigate the effect of CADPE on
PMA-induced MMPs expression and the underlying mo-
lecular mechanism. We show that CADPE inhibits the
migration and invasion of gastric cancer cells by suppres-
sing MMP-9 expression and blocking the activation of
focal adhesion kinase (FAK)/mitogen-activated protein ki-
nase (MAPK)/ERK kinase (MEK)/ERK1/2 protein kinases
and AP-1 transcription factor.
Materials and Methods
Materials and cells
CADPE was synthesized by Dr. Xiaoyuan Lian (The
Institute of Biomedical Sciences, East China Normal
University, Shanghai, China; ref. 23). A 100 mmol/L
stock solution of CADPE was prepared in DMSO.
CA and PMA were purchased from Sigma-Aldrich.
Matrigel was purchased from BD Biosciences. Kinase
inhibitors PD98059, SB203580, SP600125, and
U0126 were purchased from Calbiochem. Antibodies
against MMP-9, MMP-2, c-Fos, c-Jun, p65, total and
phosphorylated FAK, MEK, ERK1/2, stress-activated
protein kinase/JNK, and p38 MAPK were from Cell
Signaling Technology; phospho-PKC antibody sample
kit (#9921) was obtained from Cell Signaling Tech-
nology; and antibody against β-actin was purchased from
Sigma-Aldrich.
Human gastric carcinoma cell lines MGC-803, HGC-27,
and AGS and human breast carcinoma cell line MDA-MD-
231 were obtained from the China Type Culture Collection
(Shanghai, China). MGC-803 and HGC-27 were cultured
in RPMI 1640 containing 10% fetal bovine serum. AGS
and MDA-MD-231 were cultured in DMEM and L-15
medium containing 10% fetal bovine serum, respectively.
Cell viability assay
For the cell viability assay, 2 × 10
4
MGC-803 cells per
well were treated with different concentrations of CADPE
for 24 hours. Cell viability was determined by the 3-(4,5-
dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-
(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) method,
following the manual of CellTiter 96 Aqueous One Solu-
tion Cell Proliferation assay (Promega). Absorbance was
measured with a VERSAmax microplate reader.
Transwell migration assay
The Transwell migration assay was done according to a
previously reported method with some modifications (27).
Briefly, Transwell membrane (8-μm pore size, 6.5-mm
diameter; Corning Costar Corporation) was used. The bot-
tom chambers of the Transwell were filled with migration-
inducing medium (with 10% fetal bovine serum). The top
chambers were seeded with 2 × 10
4
MGC-803 cells per
well with different concentrations of CADPE (0, 5, 10,
and 20 μmol/L). After 8 to 10 hours, the filters were fixed
with 4% paraformaldehyde for 30 minutes at room tem-
perature; subsequently, the cells on the upper side of the
membrane were scraped with a cotton swab. Filters were
stained with hematoxylin for light microscopy. Images
were taken using an Olympus inverted microscope and
migratory cells were evaluated by manual counting. Per-
centage inhibition of migratory cells was quantified and
expressed based on untreated control wells.
Matrigel-based Transwell invasion assay
Cell invasion assays were carried out as previously re-
ported (27) with slight modifications. Transwell membrane
coated with Matrigel (100 μg/mL, 100 μL per well) was
used for invasion assay. Cells (5 × 10
4
) were seeded onto
the upper wells in the presence of different concentrations
ofCADPEorMMP-9inhibitor(28)withorwithout
PMA. The bottom chambers of the Transwell were filled
with condition medium. The inserts were incubated at
37°C for 24 hours. Cells that had invaded the lower surface
of the membrane were fixed, stained, and counted under a
light microscope. Percentage inhibition of invasive cells was
quantified and expressed based on untreated control wells.
Gelatin substrate gel zymography
Gelatin zymography was carried out as previous reported
(18). The MGC-803 cells were plated onto six-well plates at
Han et al.
Mol Cancer Res; 8(11) November 2010 Molecular Cancer Research1478
a density of 2 × 10
5
cells per well and incubated until they
reached 80% confluence; the medium then was changed to
fresh serum-free medium with or without CADPE or specif-
ic inhibitors of MAPKs (PD98059, U0126, SP600125, and
SB203580). After 2 hours of pretreatment, 100 nmol/L
PMA was added to the medium for 24 hours, and the super-
natant was collected and concentrated. The resultant super-
natant was subjected to SDS-PAGE in 8% polyacrylamide
gels that were copolymerized with 1 mg/mL gelatin. After
the electrophoresis runs, the gels were washed several times
with 2.5% Triton X-100 for 1 hour at room temperature to
remove the SDS and incubated for 12 hours at 37°C in a
buffer containing 5 mmol/L CaCl
2
and 1 μmol/L ZnCl
2
.
The gels were stained with Coomassie brilliant blue R250
(0.25%; Bio-Rad) for 1 hour and then destained for 1 hour
in a solution of acetic acid and methanol. Proteolytic activity
was evidenced as clear bands against the blue background of
the stained gelatin.
Western blot assay
For the Western blot assay, MGC-803 cells were treated
with different concentrations of CADPE in the presence of
100 nmol/L PMA for 24 hours. Cell lysates were prepared
in radioimmunoprecipitation assay buffer (20 mmol/L Tris,
2.5 mmol/L EDTA, 1% Triton X-100, 1% deoxycholate,
0.1% SDS, 40 mmol/L NaF, 10 mmol/L Na
4
P
2
O
7
,and
1 mmol/L phenylmethylsulfonyl fluoride). Aliquots of
cellular protein (40 μg/lane) were electrophoresed on
10% to 12% SDS-PAGE and transferred onto a polyviny-
lidene difluoride membrane (Millipore Corp.). The mem-
brane was blocked with 5% skim milk in PBS containing
0.1% Tween 20 and then reacted with specific antibodies.
Detection of specific proteins was carried out with an en-
hanced chemiluminescence Western blotting kit following
the manufacturer's instructions (Amersham-Pharmacia).
To examine the activation of transcription factors, nuclear
fractions were obtained from cells treated with PMA for
1 hour after 4 hours of pretreatment with CADPE. To
assess the changes in signaling pathway, starved MGC-803
cells were treated with CADPE for 2 hours and then stimu-
lated with 100 nmol/L PMA.
Reverse transcription-PCR
In the reverse transcription-PCR (RT-PCR) analysis, to-
tal RNA was extracted from the treated cells. For reverse
transcription reaction, cDNA was synthesized from 1 μg
of total RNA using Moloney murine leukemia virus reverse
transcriptase (Promega). The PCR primers used are as follows:
MMP-9 sense, 5-TCCCTGGAGACCTGAGAACC-3;
MMP-9 antisense, 5-GGCAAGTCTTCCGAGTAGTTT-
3;MMP-2 sense, 5-GGATGATGCCTTTGCTCG-3;
MMP-2 antisense, 5-ATCGGCGTTCCCATACTT-3;
MMP-7 sense, 5-CTTCCTGTATGCTGCAACTC-3,
MMP-7 antisense, 5-GTGGAGGAACAGTGCTTATC-3.
TIMP-1 sense, 5-GGGGACACCAGAAGTCAACCAGA-
3;TIMP-1 antisense, 5-CTTTTCAGAGCCTTGGAG-
GAGCT-3;TIMP-2 sense, 5-TGCAGCTGCTCCCC-
GGTGCAC-3;TIMP-1 antisense, 5-TTATGGGTCCT
CGATGTCGAG-3;β-actin sense, 5-GCCATCGTCAC-
CAACTGGGAC-3;andβ-actin antisense, 5-CGATTT
CCCGCTCGGCCGTGG-3. PCR products were analyzed
by agarose gel electrophoresis and visualized by treatment
with ethidium bromide.
Construction of human MMP-9 promoter
A 700-bp fragment at the 5-flanking region of the hu-
man MMP-9 gene was amplified by PCR from human ge-
nomic DNA. Specific primers were designed to contain the
appropriate restriction enzyme site: sense 5-CGG
GGTACCTGCTACTGTCCCCTTTACTG-3(KpnI)
and antisense 5-CCCAGATCTGTGAGGGCA-
GAGGTGTCT-3(BglII). The amplified promoter DNA
was digested with KpnIandBglIIandthenclonedup-
stream of the luciferase gene in pGL3 plasmid. The
DNA sequence of the MMP-9 promoter was confirmed,
and the resultant reporter plasmid was named pGL3-
MMP-9-WT. The AP-1-1, AP-1-2, NF-κB, and SP-1 mu-
tants from pGL3-MMP-9-WT were generated using the
QuickChange Site-Directed Mutagenesis Kit (Stratagene);
all the mutants were confirmed by DNA sequencing.
Transient transfection and luciferase reporter
gene assays
MMP-9 wild-type (pGL3-MMP-9-WT), AP-1 site-
mutated (pGL3-MMP-9-Mut-AP-1-2), NF-κB site-mutated
(pGL3-MMP-9-Mut-NF-κB), and SP-1 site-mutated
MMP-9 luciferase promoter constructs (pGL3-MMP-
9-Mut-SP-1) were used in transient transfection assays.
MGC-803 cells were plated onto six-well plates at a density
of 2 × 10
5
cells per well and grown overnight. Cells were
cotransfected with 1 μg of MMP-9 promoter-luciferase
reporter constructs and 0.2 μg of the Renilla reporter plas-
mid for 6 hours using Lipofectamine reagent (Invitrogen)
according to the manufacturer's protocol. After transfec-
tion, the cells were cultured in condition medium with
PMA and incubated with different concentrations of
CADPE for 24 hours. Luciferase and Renilla activities
were determined by following the manufacturer's protocol
(Dual-Luciferase Reporter Assay System, Promega). Lucif-
erase activity was normalized with the Renilla activity in the
cell lysate and expressed as an average of three independent
experiments.
Electophoretic mobility shift assay
Cultured cells were collected by centrifugation, washed,
and suspended in buffer A [10 mmol/L HEPES (pH 7.9),
10 mmol/L KCl, 0.1 mmol/L EDTA, 0.1 mmol/L EGTA,
1 mmol/L DTT, and 0.5 mmol/L phenylmethylsulfonyl
fluoride]. After 15 minutes on ice, the cells were vortexed
in the presence of 0.5% NP40. The nuclear pellet was then
collected by centrifugation and extracted with buffer B
[20 mmol/L HEPES (pH 7.9), 0.4 mol/L NaCl, 1 mmol/L
EDTA, 1 mmol/L EGTA, 1 mmol/L DTT, and 1 mmol/L
PMSF] for 15 minutes at 4°C. Double-stranded oligonu-
cleotides containing the consensus sequences for AP-1 up-
stream (5-GAGGAAGCTGAGTCAAAGAAGGC-3),
CADPE Inhibits Gastric Carcinoma Invasion
Mol Cancer Res; 8(11) November 2010www.aacrjournals.org 1479
AP-1 proximal (5-CTGACCCCTGAGTCAGCACTT-
GC-3), and NF-κB(5-CCCCAGTGGAATTCCCCA
GCCTTG-3) were end- labeled with [γ-
32
P]ATP using
T4 polynucleotide kinase and used as probes for electro-
phoretic mobility shift assay (18). The nuclear extracts
(5 μg) were incubated at 4°C for 30 minutes in 25
mmol/L HEPES buffer (pH 7.9), 0.5 mmol/L EDTA,
0.5 mmol/L DTT, 0.05 mol/L NaCl, and 2.5% glycerol
with 1 μg of poly(deoxyinosinic-deoxycytidylic acid) and
0.5 pmol of labeled probe. The DNA-protein complex
FIGURE 1. Chemical structures of CADPE and CA and the effects of CADPE on the viability, migration, and invasion of MGC-803 cells. A, chemical
structures of CA and CADPE. B, effect of CADPE on cell viability. MGC-803 cells were treated with 5 to 20 μmol/L CADPE in serum or serum-free medium
for 24 h. Cell viability was determined by MTS assay. The percentage of cell viability was calculated as the ratio (A
570
) of treated cells to control cells.
Points, mean of three independent experiments; bars, SE. C, CADPE inhibits tumor cell migration. The Transwell migration assay was carried out in
MGC-803 cells. After 8 h of incubation with or without the indicated concentration of CADPE, cells that migrated to the low chamber were fixed, stained, and
counted by light microscopy as described in Materials and Methods (left). Random fields were scanned (four fields per filter of the well) for the
presence of cells on the lower side of the membrane (right image). Columns, mean number of migrated cells in three wells from three independent
experiments; bars, SE. *, P< 0.05. D, the effect of CADPE on cell invasion was determined using the Matrigel invasion assay system. For invasion assay, the
upper chamber was coated with Matrigel, and MGC-803 cells with various concentrations of CADPE in the presence of 100 nmol/L PMA were added.
After 24 h, cells on the bottom side of filter were fixed, stained, and counted under a light microscope (D, left images). The inhibitory effects of CADPEon
two additional gastric cancer cell lines, HGC-27 and AGS, were also examined (D, right columns). Columns, mean number of migrated cells in
three wells from three independent experiments; bars, SE. *, P< 0.05.
Han et al.
Mol Cancer Res; 8(11) November 2010 Molecular Cancer Research1480
was separated by electrophoresis at 4°C in 6% polyacryl-
amide gels in 0.5× Tris-borate EDTA buffer. For compe-
tition assay to confirm the binding specificity, nuclear
extracts were preincubated at 4°C for 30 minutes with
a 100-fold excess of an unlabeled oligonucleotide. Gels
were dried and imaged using the Personal Molecular Im-
ager system (Bio-Rad).
Statistical analysis
The results are presented as mean ± SE, and statistical
comparisons between groups were done using one-way
ANOVA followed by Student's ttest. P0.05 was consid-
ered statistically significant.
Results
Effects of CADPE on the viability and invasion of
gastric carcinoma cells
As a synthetic derivative of CA, CADPE preserved
the key structure of CA (Fig. 1A). To evaluate the
effect of CADPE on cell proliferation, we treated
MGC-803 cells with different concentrations of
CADPE or CA in serum-free and serum-containing me-
dium, respectively, for MTS assays. Only 9% and 15%
decreases in cell viability were found in cells treated
with 25 μmol/L CADPE in serum-containing and
serum-free medium (Fig. 1B). No significant cytotoxi-
city was found for CA at the concentration of
100 μmol/L (data not shown). To investigate the effects
of CADPE on tumor cell migration and invasion,
the Transwell migration assay and the Matrigel-based
Transwell invasion assay were done using MGC-803
cells at different concentrations of CADPE, ranging
from 5 to 20 μmol/L. As shown in Fig. 1C, CADPE
significantly inhibited MGC-803 cancer cell migration
(Fig. 1C). Moreover, CADPE dramatically inhibited
PMA-induced cell invasion in a dose-dependent
manner (Fig. 1D), suggesting that CADPE is an effec-
tive inhibitor of cancer cell migration, invasion,
and metastasis.
FIGURE 2. Inhibition of MMP-9 activity by
CADPE in tumor cells. A, CADPE inhibits
PMA-induced MMP-9 activity. MGC-803,
HGC-27, AGS, and MDA-MB-231 cells were
preincubated with varying concentrations of
CADPE for 2 h, followed by PMA stimulation for
24 h. Conditional media were collected and
MMP activity was analyzed by gelatin
zymography. B, CADPE inhibits EGF-induced
MMP-9 activity. MGC-803 cells were incubated
with varying concentrations of CADPE in the
presence of EGF for 24 h and gelatin
zymography was done. C, CA inhibits
PMA-induced MMP-9 activity. MGC-803 cells
were incubated with 25 to 100 μmol/L CA in the
presence of PMA, and MMP-9 enzyme activity
in the conditioned medium was analyzed by
zymography. D, MMP-9 and MMP-2 derived
from PMA-treated conditioned medium were
incubated with CADPE (5-20 μmol/L) for 30 min
and then subjected to gelatin zymography.
CADPE Inhibits Gastric Carcinoma Invasion
Mol Cancer Res; 8(11) November 2010www.aacrjournals.org 1481
CADPE inhibits MMP-9 expression and activity
MMP-9 and MMP-2 are important ECM-degrading
enzymes. It has been reported that both enzymes were in-
volved in cancer cell invasion and metastasis (14, 29). The
fact that CADPE inhibited cancer cell invasion prompted
us to examine the effect of CADPE on MMPs activity
using gelatin zymography. The secretion of MMP-9 in
the conditioned medium of MGC-803 was dramatically
induced by PMA (10-100 nmol/L) in a dose-dependent
manner, whereas no detectable change of MMP-2 (data
not shown) was found in the same condition. To examine
the effects of CADPE on MMP-9 expression in cancer
cells, we chose 100 nmol/L PMA to induce the activation
of MMP-9 in different cancer cells. As shown in Fig. 2A,
treatment of MGC-803 cells with CADPE (5-20 μmol/L)
suppressed PMA-induced MMP-9 activity in a dose-
dependent manner, whereas the activity of MMP-2 did
not significantly decrease. Similar results were obtained
in three other cancer cell lines, including two gastric can-
cer cell lines (AGS and HGC-27) and one breast cancer
cell line (MDA-MB-231; Fig. 2A), suggesting that
CADPE significantly inhibits the secretion of MMP-9 in
FIGURE 3. Inhibition of PMA-induced MMP-9
expression by CADPE. A, CADPE inhibits
the protein expression of MMP-9, but not
MMP-2. The expression levels of MMP-9 and
MMP-2 in MGC-803 cells treated with
CADPE in the presence of PMA for 24 h were
evaluated by Western blot analysis with MMP-9
and MMP-2 antibodies. Expression of β-actin
in cell lysates was used as a control. B, CADPE
inhibits MMP-9, but not MMP-2, mRNA level.
MGC-803 cells were treated with or without
CADPE (20 μmol/L) in the presence of PMA for
0, 3, 6, 12, 18, and 24 h, and mRNA levels of
MMP-9 and MMP-2 were examined. C, effects
of CADPE on the mRNA levels of MMPs and
their regulators. MGC-803 cells were incubated
with CADPE for 24 h. The mRNA expression
levels of MMP-9, MMP-2, MMP-3, MMP-7,
TIMP-1, and TIMP-2 were analyzed by RT-PCR;
β-actin expression was included as an internal
control. The expression levels of MMP-9 in
CADPE-treated or untreated MGC-803 cells
were determined by real-time PCR analysis.
D, MMP-9 inhibitor can block PMA-induced
MGC-803 cell invasion in the Matrigel
invasion assay.
Han et al.
Mol Cancer Res; 8(11) November 2010 Molecular Cancer Research1482
invasive cancer cell lines. Compared with CADPE, CA
also decreased MMP-9 activity in MGC-803 cells, but
at a much higher concentration (25-100 μmol/L). CA
had little effect on MMP-2 activity (Fig. 2C). Further-
more, we show that CADPE inhibited epidermal growth
factor (EGF)induced MMP-9 expression and activity in
MGC-803 cells (Fig. 2B). To investigate whether CADPE
directly affects MMP-9 enzyme activity, conditioned me-
dium derived from PMA-treated MGC-803 was incubated
with different concentrations of CADPE in the gelatin zy-
mography assays. As shown in Fig. 2D, there was no vis-
ible difference between the CADPE- treated and untreated
groups (Fig. 2D), suggesting that CADPE has no direct
influence on MMP-9 activity. Taken together, our data
suggest that CADPE inhibits PMA- and EGF-induced
MMP-9 activation in different cancer cell lines.
CADPE suppresses MMP-9 transcription in a
dose-dependent manner
As shown in Fig. 3A, the expression levels of MMP-9
gradually decreased in a dose-dependent manner in
Western blot assay, indicating that CADPE inhibits
MMP-9 enzyme activity by reducing the protein level
of MMP-9 (Fig. 3A). Similar to prior observations,
FIGURE 4. Analysis of CADPE on MMP-9
promoter activity through AP-1 and NF-κB
binding sites. A, CADPE inhibits PMA-induced
MMP-9 luciferase activity. B and C, mutations
at NF-κB and SP-1 binding sites have little
influence on the inhibitory effects of CADPE in
MGC-803 cells. D, mutations at the two AP-1
binding sites of the MMP-9 promoter diminish
the inhibitory effects of CADPE, suggesting that
CADPE suppresses the expression of MMP-9
through AP-1 binding sites. E and F, CADPE
inhibits the AP-1-luciferase, but not the
NF-κB-luciferase, activity. MGC-803 cells were
transfected with reporter vectors containing
binding sites for AP-1 and NF-κB. The cells
were cultured in the presence of CADPE
(5-20 μmol/L) for 24 h, and luciferase activity
was measured. Columns, mean of at least three
independent experiments; bars, SE.
*, P< 0.05.
CADPE Inhibits Gastric Carcinoma Invasion
Mol Cancer Res; 8(11) November 2010www.aacrjournals.org 1483
CADPE had little effect on the protein expression of
MMP-2 (Fig. 3A). Furthermore, we performed RT-
PCR to determine the CADPE regulation of MMPs at
the mRNA level. In MGC-803 cells, the mRNA level of
MMP-9 was induced by PMA after 3 hours, peaked in
12 hours, and persisted for least 24 hours (Fig. 3B).
CADPE significantly inhibited PMA-induced MMP-9
mRNA expression in a time- and dose-dependent man-
ner (Fig. 3B and C). CADPE had no effect on the
mRNA level of MMP-2 (Fig. 3B and C). The inhibitory
effect of CADPE on MMP-9 mRNA expression was fur-
ther confirmed by real-time quantitative PCR (Fig. 3C).
Because the activity of MMP-9 is tightly regulated by
endogenous inhibitors, tissue inhibitors of metalloprotei-
nases (TIMP), we examined the expression levels of
TIMP-1 and TIMP-2 by RT-PCR. As shown in Fig. 3C,
TIMP-1, but not TIMP-2, can be slightly stimulated by
PMA. However, CADPE had no effect on the mRNA
levels of both TIMP-1 and TIMP-2 (Fig. 3C). Further-
more, we examined the expression level of MMP-7 in
CADPE-treated MGC-803 cells and showed that
MMP-7 remained essentially unchanged (Fig. 3C). These
results indicate that CADPE selectively suppressed MMP-
9 expression both at the protein and mRNA levels in a
time- and dose-dependent manner.
To understand the relationship of cell invasion and
MMP-9 in gastric cancer, we performed a Matrigel inva-
sion assay with MMP-9 inhibitor. Results showed that
MMP-9 inhibitor blocked MGC-803 cell invasion in a
dose-dependent manner, suggesting that MMP-9 was
largely responsible for the invasion of MGC-803 cells
(Fig. 3D). The inhibitory effect of MMP-9 inhibitor on
MGC-803 cell invasion was almost same as that of
CADPE (Supplementary Fig. S2B). The specificity of
MMP-9 inhibitor was tested by gelatin zymography assay
in different gastric carcinoma cell lines (Supplementary
Fig. S1). We also detected the effects of MMP-9 inhibitor
on the migration of gastric cancer cells. Results showed
that MMP-9 inhibitor had less inhibitory effects when
compared with CADPE (Supplementary Fig. S2A).
CADPE inhibits MMP-9 expression by suppressing
AP-1 binding and AP-1dependent
transcription activity
To understand the molecular mechanism underlying
the inhibitory effects of CADPE on MMP-9 expression,
we find that there are two AP-1 binding sites (located at
79 bp and 533 bp) and an NF-κB binding site (loca-
ted at 600 bp) in the MMP-9 promoter. It has been
shown that NF-κB and AP-1 play an important role in
controlling basal and cytokine-induced MMP-9 expres-
sion in various cancer cell lines (18). To determine the
effect of CADPE on the promoter activity of MMP-9,
luciferase-report gene that contains the MMP-9 promoter
region was transiently transfected into MGC-803 cells. As
shown in Fig. 4A, MMP-9-luciferase activity was activat-
ed up to 10-fold in cells treated with PMA. CADPE
inhibited the PMA-induced MMP-9-luciferase activity
in a dose-dependent manner (Fig. 4A), suggesting that
CADPE could inhibit MMP-9 expression at the tran-
scriptional level.
To determine which of these transcription factors may
participate in the regulation of MMP-9 transcription in
MGC-803 cells, we mutated the potential binding sites
for different transcription factors found in the MMP-9
promoter, including NF-κB, SP-1, and two AP-1 sites
(Fig. 4B-D). MGC-803 cells were transiently transfected
with MMP-9 reporter genes with mutations in different
transcription binding sites. Mutations at the NF-κB and
SP-1 binding sites have little effect on the inhibitory
effects of CADPE on PMA-induced MMP-9 activity
(Fig. 4B and C). However, mutations at the two AP-1
binding sites completely abolished the inhibitory effects
of CADPE on PMA-induced MMP-9 promoter activity
(Fig. 4D), suggesting that the regulation of CADPE on
the MMP-9 promoter region was facilitated by AP-1
transcription factor. To further confirm this observation,
the luciferase report vectors that contain tandem repeats
of the AP-1 or NF-κB binding sites were used to examine
the effects of CADPE in the luciferase assays. As shown
in Fig. 4E, luciferase activity in the cells transfected with
the AP-1 reporter was significantly reduced by treatment
with CADPE in the range of 5 to 20 μmol/L, whereas no
statistically significant changes were found in the cells
transfected with the NF-κB reporters in the presence of
CADPE (Fig. 4F). These results suggest that AP-1 tran-
scription factor and AP-1 binding sites in the MMP-9
promoter region contribute to the inhibition of PMA-
dependent MMP-9 activation by CADPE.
CADPE decreases transcription factor binding to AP-1
motifs in the MMP-9 promoter region
To determine whether CADPE inhibited the transcrip-
tional binding activity of AP-1 to its DNA motifs, we per-
formed electrophoretic gel mobility shift assay using the
consensus sequences of AP-1 or NF-κBasprobesin
CADPE-treated cells. MGC-803 cells were pretreated with
different concentrations of CADPE for 4 hours, followed
by treatment with 100 nmol/L PMA for 1 hour. Then, nu-
clear extracts were prepared and analyzed for AP-1 and
NF-κB DNA binding activities, respectively. As shown in
Fig. 5A, CADPE significantly decreased PMA-induced
AP-1 DNA binding ability, whereas CADPE has no effect
on PMA-induced NF-κB binding activity (Fig. 5B). These
data were consistent with the reporter gene analysis, sug-
gesting that CADPE blocks MMP-9 expression, at least
in part, by inhibiting the expression or DNA binding ac-
tivity of AP-1 transcription factor. To determine which
subunit of AP-1 transcritpion factor is regulated by
CADPE, we examined the expression levels of c-Fos and
c-Jun with CADPE treatment. Our data showed that
CADPE significantly reduced PMA-induced c-Fos expres-
sion but had little effect on the expression of c-Jun or p65
in Western blot assays (Fig. 5C), suggesting that CADPE
inhibits AP-1 transcription activity by suppressing the
expression of c-Fos in the gastric cancer cells.
Han et al.
Mol Cancer Res; 8(11) November 2010 Molecular Cancer Research1484
CADPE blocks PMA-induced activation of FAK, MEK,
and ERK1/2 in gastric cancer cells
Activation of one or more mitogen-activated protein
kinase (MAPK) pathways is important for the MMP-9 in-
duction by PMA in various cell types (30). As shown in
Fig. 6A, the phosphorylation of ERK, JNK, and p38 was
increased by the stimulation with PMA. Addition of
CADPE decreased only ERK phosphorylation in MGC-
803 cells (Fig. 6B). On the other hand, the levels of phos-
phorylated JNK and p38 had little change after CADPE
treatment (Fig. 6B). We further examined the phosphory-
lation of upstream regulators in the ERK signaling pathway
by Western blot with specific phosphorylation antibodies.
CADPE significantly inhibited MEK and FAK phosphor-
ylation induced by PMA treatment (Fig. 6B). As PKC is
also involved in MMP-9 expression, we examined the effects
of CADPE on PKC activation using specific phosphor-
antibodies for different PKC isoforms as described in
Materials and Methods. Our results showed that CADPE
has little effect on PKC activation (Fig. 6B), suggesting
that the effects of CADPE are mediated by pathways other
than PKC. To further confirm our conclusion, we measured
MMP-9 activity using specific protein kinase inhibitor as-
says. Overnight starved MGC-803 cells were pretreated
with PD98059, U0126, SP600125, and SB203580 (inhi-
bitors of MEK, ERK, JNK, and p38, respectively) for
2hoursandthenstimulatedwithPMAfor24hours;
conditioned media were collected for gelatin zymography
assay of MMP-9 activity. As shown in Fig. 6C, MEK
and ERK inhibitors can significantly suppress MMP-9
expression and activity, whereas JNK and p38 inhibitors
have little inhibitory activity on MMP-9. These results
suggest that CADPE inhibits the PMA-induced activation
of AP-1 by blocking FAK and MEK/ERK1/2 activation in
MGC-803 cells.
Discussion
Metastasis and invasion are major properties of various
malignant tumors that are associated with a poor progno-
sis. It was thought that MMP-9 participated in promoting
these processes. Recent studies show that MMP-9 has
statistically significantly different expression patterns
between well-differentiated and poorly differentiated tissue
samples and may play key roles during the development of
gastric cancer (31). In addition, MMP-9 also correlated
with the invasion, metastasis, and angiogenesis of gastric
cancer cells (32, 33). Therefore, development of various
compounds that can inhibit MMP-9 would be useful in
the treatment of gastric carcinoma.
FIGURE 5. Effects of CADPE on
the DNA binding activities and
expression of AP-1 and NF-κB.
A and B, CADPE suppresses
PMA-induced AP-1 DNA binding
activity but has little effect on
NF-κB activity. MGC-803 cells
were pretreated with CADPE for
4 h and then stimulated with PMA
for 1 h. Nuclear extract (5 μg)
prepared from these treated cells
was mixed with radioactive
oligonucleotides containing the
AP-1 (A) or NF-κB (B) motif of the
MMP-9 promoter. Bound
complexes were analyzed by
electrophoresis. C, effects of
CADPE on the expression levels of
AP-1 and NF-κB subunits.
MGC-803 cells were pretreated
with CADPE for 4 h followed by
PMA stimulation for 1 h. Western
blot analysis was done to
determine the nuclear levels of
AP-1 (c-Fos and c-Jun) and NF-κB
(p65) subunits using specific
antibodies for the proteins; β-actin
was used as an internal control.
CADPE Inhibits Gastric Carcinoma Invasion
Mol Cancer Res; 8(11) November 2010www.aacrjournals.org 1485
As a structure analogue of CADPE, CA has a wide spec-
trum of biological effects, including antitumor invasion by
targeting MMP-9 through the AP-1 and NF-κB signaling
pathways (18). The ability to inhibit tumor cell invasion
was also extended to other caffeic acid derivatives, such
as caffeic acid phenethyl ester, which was originally isolated
from honeybee propolis. However, the inhibitory role of
CADPE against MMP expression and the invasiveness of
gastric carcinoma has not been reported. In this study, we
show that CADPE is a potential anticancer agent due to its
ability to inhibit PMA-induced phosphorylation of protein
kinases (FAK, MEK, and ERK1/2), AP-1 nuclear translo-
cation, and MMP-9 expression in invasive tumor cells.
Gelatin zymography is a classic method to detect the ac-
tivity and expression of gelatinases A and B, and thus we
used this method to detect the secretion of MMP-9 and
MMP-2 in the condition medium. In this study, treatment
of MGC-803 cells with CADPE selectively suppressed the
PMA-induced activity of MMP-9, whereas the activity of
MMP-2 was not affected. A similar result was also ob-
served in two more gastric carcinoma cell lines, AGS and
HGC-27, and a breast cancer cell line, MDA-MB-231.
When compared with its precursor CA, CADPE showed
a better inhibitory effect on MMP-9 at a relatively lower
concentration, suggesting that CADPE inhibits tumor in-
vasion through MMP-9 in these invasive tumor cell lines.
The activity of MMPs is precisely regulated at three
levels: gene transcription, posttranscriptional activation
of zymogens, and endogenous expression of tissue inhi-
bitors of metalloproteinases (34). It was thought that the
key step in the regulation of MMPs was at the transcrip-
tion level. To determine which step was affected by
CADPE, gelatin zymography, RT-PCR, and Western blot
analysis were performed to show that CADPE inhibits
the expression of MMP-9 at both the mRNA and pro-
tein levels but has little effect on the enzymatic activity.
FIGURE 6. CADPE inhibits
the activation of FAK, MEK, and
ERKs. A, time-dependent
phosphorylation of ERK, JNK, and
p38 induced by PMA in MGC-803
cells. B, effects of CADPE on
the phosphorylation of ERK, JNK,
p38, MEK, FAK, and PKC.
MGC-803 cells were pretreated
with CADPE for 4 h and stimulated
with PMA for 15 min; the levels of
phosphorylated protein kinases,
including pERK, p-JNK, p-p38,
p-MEK, p-FAK, and p-PKC, were
determined by Western blotting
with phospho-specific antibodies.
C, inhibition of PMA-induced
MMP-9 activity by protein kinase
inhibitors for MEK and ERK.
MGC-803 cells were pretreated
with U0126 (MEK inhibitor; 10
μmol/L), SP600125 (JNK inhibitor;
10 μmol/L), PD98059 (ERK
inhibitor; 20 μmol/L), and
SB203580 (p38 inhibitor;
10 μmol/L) for 2 h and then treated
with PMA for 24 h; MMP-9
levels were tested by gelatin
zymography. D, possible molecular
mechanisms underlying the
antitumor activities of CADPE in
PMA-treated MGC-803 cells.
Han et al.
Mol Cancer Res; 8(11) November 2010 Molecular Cancer Research1486
MMPs activity is also regulated by tissue-specific inhibi-
tors (TIMPs), of which there are four identified members
(TIMP-1 to TIMP-4; ref. 8). These proteins bind MMPs
in a 1:1 stoichiometry and directly affect the level of
MMP activity. Because TIMP-1 is a major inhibitor of
MMP-9, TIMP-1 and TIMP-2 are differentially regulated
in vivo as well as in cultured cells (35, 36). We tested the
mRNA levels of these two proteins and ruled out the ef-
fects of CADPE on TIMP-1 and TIMP-2. Therefore, the
inhibitory effect of CADPE on MMP-9 activity was
mainly due to the transcriptional regulation and protein
expression of MMP-9, which was further confirmed by
MMP-9-luciferase report gene assay.
AP-1 and NF-κB are two key transcription factors in-
volved in the regulation of MMP-9 gene expression (19).
Activation of these transcription factors is centrally in-
volved in the process of tumor invasion and metastasis
by various agents including PMA, growth factors (e.g.,
EGF, vascular endothelial growth factor, platelet-derived
growth factor, and transforming growth factor-β;refs.
37-39), and inflammatory cytokines (e.g., CXCL12 and
tumor necrosis factor-α; refs. 40, 41). Thus, the regulation
of AP-1 and NF-κB downstream of the FAK, phosphoino-
sitide 3-kinase/Akt, and MAPK pathways might be in-
volved in PMA-induced MMP-9 expression and invasion
in MGC-803 cells. Luciferase reporter gene assay and mu-
tation analysis of the promoter revealed that the major tar-
get of CADPE in the MMP-9 promoter was AP-1, whereas
NF-κB had little effect on the transcriptional regulation of
MMP-9. AP-1 is composed of members of the c-Jun and
c-Fos families (21, 42, 43). They associate to form a variety
of homodimers and heterodimers to regulate gene expres-
sion and localize to the nucleus when AP-1 is activated.
Our results showed that CADPE suppressed nuclear
c-Fos in a dose-dependent manner, whereas c-Jun or p65
was not affected, suggesting that the suppression of AP-1 is
responsible for CADPE-induced inhibition of MMP-9 in-
duction and cell invasion. AP-1 activity, including tran-
scriptional activation, protein stability, and intracellular
localization, was modulated by protein kinases such as
MAPKs. It was reported that the intracellular localization,
protein stability, and chromatin association of c-Fos family
proteins were mainly regulated by ERK phosphorylation
(44, 45). In our experiments, CADPE specifically sup-
pressed PMA-mediated MEK and ERK phosphorylation
without affecting pathways involving p38 and JNK. Acti-
vation of PKC was also little affected by CADPE, suggest-
ing that the effects of CADPE are mediated by pathways
other than PKC. Furthermore, CADPE also inhibits
PMA-induced phosphorylation of FAK. FAK plays a
critical role in contact formation between ECM and
cytoskeleton and has been linked with cancer cell migra-
tion, invasion, proliferation, and survival (46). Therefore,
suppression of FAK may partially explain the inhibition of
cell migration in our assays.
In summary, our studies show that CADPE inhibits tu-
mor cell migration and invasion by suppressing MMP-9
expression by blocking the phosphorylation of protein
kinases (FAK, MEK, and ERK) and the activation of
AP-1 transcriptional factor. Therefore, CADPE is a poten-
tial agent for clinical use in preventing the invasion and
metastasis of human malignant tumors, such as gastric cancer.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Grant Support
Research Platform for Cell Signaling Networks (06DZ22923), Pujiang Program
(09PJ1403900), Key Science and Technology Projects (074319104 and
09JC1405200) from the Science and Technology Commission of Shanghai Muni-
cipality, Chen GuangProject (2008CG27) supported by Shanghai Municipal
Education Commission and Shanghai Education Development Foundation.
The costs of publication of this article were defrayed in part by the payment of
page charges. This article must therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Received 03/22/2010; revised 09/03/2010; accepted 09/05/2010; published
OnlineFirst 10/06/2010.
References
1. Bozzetti F, Marubini E, Bonfanti G, Miceli R, Piano C, Gennari L,
Italian Gastrointestinal Tumor Study Group. Subtotal versus total
gastrectomy for gastric cancer: five-year survival rates in a multi-
center randomized Italian trial. Ann Surg 1999;230:1708.
2. Keighley MR. Gastrointestinal cancers in Europe. Aliment Pharmacol
Ther 2003;18 Suppl 3:730.
3. Dicken BJ, Bigam DL, Cass C, Mackey JR , Joy AA, Hamilton SM.
Gastric adenocarcinoma: review and considerations for future direc-
tions. Ann Surg 2005;241:2739.
4. Yu HG, Ai YW, Yu LL, et al. Phosphoinositide 3-kinase/Akt pathway
plays an important role in chemoresistance of gastric cancer cells
against etoposide and doxorubicin induced cell death. Int J Cancer
2008;122:43343.
5. Kolev Y, Uetake H, Takagi Y, Sugihara K. Lactate dehydrogenase-5
(LDH-5) expression in human gastric cancer: association with hypoxia-
inducible factor (HIF-1α) pathway, angiogenic factors production and
poor prognosis. Ann Surg Oncol 2008;15:233644.
6. Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor
metastasis. Cancer Metastasis Rev 2006;25:934.
7. Yan C, Boyd DD. Regulation of matrix metalloproteinase gene
expression. J Cell Physiol 2007;211:1926.
8. Nagase H, Woessner JF, Jr., Matrix metalloproteinases. J Biol Chem
1999;274:214914.
9. Fingleton B. Matrix metalloproteinases: roles in cancer and metasta-
sis. Front Biosci 2006;11:47991.
10. Egeblad M, Werb Z. New functions for the matrix metalloproteinases
in cancer progression. Nat Rev Cancer 2002;2:16174.
11. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regula-
tors of the tumor microenvironment. Cell 2010;141:5267.
12. Chambers AF, Matrisia n LM. Changing views of the role of matrix
metalloproteinases in metastasis. J Natl Cancer Inst 1997;89:
126070.
13. Scorilas A, Karameris A, Arnogiannaki N, et al. Overexpression
of matrix-metalloproteinase-9 in human breast cancer: a potential
CADPE Inhibits Gastric Carcinoma Invasion
Mol Cancer Res; 8(11) November 2010www.aacrjournals.org 1487
favourable indicator in node-negative patients. Br J Cancer 2001;
84:148896.
14. Nabeshima K, Inoue T, Shimao Y, Sameshima T. Matrix metallopro-
teinases in tumor invasion: role for cell migration. Pathol Int 2002;52:
25564.
15. HanS,RitzenthalerJD,SitaramanSV,RomanJ.Fibronectinin-
creases matrix metalloproteinase 9 expression through activation
of c-Fos via extracellular-regulated kinase and phosphatidylinositol
3-kinase pathways in human lung carcinoma cells. J Biol Chem
2006;281:2961424.
16. Cho HJ, Kang JH, Kwak JY, et al. Ascofuranone suppresses PMA-
mediated matrix metalloproteinase-9 gene activation through the
Ras/Raf/MEK/ERK- and Ap1-dependent mechanisms. Carcinogene-
sis 2007;28:110410.
17. Arai K, Lee SR, Lo EH. Essential role for ERK mitogen-activated pro-
tein kinase in matrix metalloproteinase-9 regulation in rat cortical as-
trocytes. Glia 2003;43:25464.
18. Chung TW, Moon SK, Chang YC, et al. Novel and therapeutic effect
of caffeic acid and caffeic acid phenyl ester on hepatocarcinoma
cells: complete regression of hepatoma growth and metastasis by
dual mechanism. FASEB J 2004;18:167081.
19. Sato T, Koike L, Miyata Y, et al. Inhibition of activator protein-1 bind-
ing activity and phosphatidylinositol 3-kinase pathway by nobiletin, a
polymethoxy flavonoid, results in augmentation of tissue inhibitor of
metalloproteinases-1 production and suppression of production of
matrix metalloproteinases-1 and -9 in human fibrosarcoma HT-
1080 cells. Cancer Res 2002;62:10259.
20. Eferl R, Wagner EF. AP-1: a double-edged sword in tumorigenesis.
Nat Rev Cancer 2003;3:85968.
21. Matthews CP, Colburn NH, Young MR. AP-1 a target for cancer pre-
vention. Curr Cancer Drug Targets 2007;7:31724.
22. El-Mousallamy AM, Hawas UW, Hussein SA. Teucrol, a decarboxyr-
osmarinic acid and its 4-O-tr iglycoside, teucroside from Teucrium
pilosum. Phytochemistry 2000;55:92731.
23. Zhang Z, Xiao B, Chen Q, Lian XY. Synthesis and biological evaluation
of caffeic acid 3,4-dihydro xyphenethyl ester. J Nat Prod 2010;73:2524.
24. Jung JE, Kim HS, Lee CS, et al. Caffeic acid and its synthetic derivative
CADPE suppress tumor angiogenesis by blocking STAT3-mediated
VEGF expression in human renal carcinoma cells. Carcinogenesis
2007;28:17807.
25. Silva FA, Borges F, Guimaraes C, Lima JL, Matos C, Reis S. Phenolic
acids and derivatives: studies on the relationship among structure,
radical scavenging activity, and physicochemical parameters. J Agric
Food Chem 2000;48:21226.
26. Nardini M, Leonardi F, Scaccini C, Virgili F. Modulation of ceramide-
induced NF-κB bindin g activity and apoptotic response by caffeic
acid in U937 cells: comparison with other antioxidants. Free Radic
Biol Med 2001;30:72233.
27. Chen HW, Lee JY, Huang JY, et al. Curcumin inhibits lung cancer cell
invasion and metastasis through the tumor suppressor HLJ1. Cancer
Res 2008;68:742838.
28. Mifune M, Ohtsu H, Suzuki H, et al. G protein coupling and sec-
ond messenger generation are indispensable for metalloprotease-
dependent, heparin-binding epidermal growth factor shedding
through angiotensin II type-1 receptor. J Biol Chem 2005;280:
265929.
29. Hu B, Thirtamara-Rajamani KK, Sim H, Viapiano MS. Fibulin-3 is
uniquely upregulated in malignant gliomas and promotes tumor cell
motility and invasion. Mol Cancer Res 2009;7:175670.
30. Woo JH, Lim JH, Kim YH, et al. Resveratrol inhibits phorbol myristate
acetate-induced matrix metalloproteinase-9 expression by inhibiting
JNK and PKCδsignal transduction. Oncogene 2004;23:184553.
31. Song G, Ouyang G, Mao Y, Ming Y, Bao S, Hu T. Osteopontin pro-
motes gastric cancer metastasis by augmenting cell survival and
invasion through Akt-mediated HIF-1αupregulation and MMP9 acti-
vation. J Cell Mol Med 2008;13:170618.
32. Huang W, Yu LF, Zhong J, et al. Angiotensin II type 1 receptor
expression in human gastric cancer and induces MMP2 and
MMP9 expression in MKN-28 cells. Dig Dis Sci 2008;53:1638.
33. Jin H, Pan Y, He L, et al. p75 neurotrophin receptor inhibits invasion
and metastasis of gastric cancer. Mol Cancer Res 2007;5:42333.
34. Jones CB, Sane DC, Herrington DM. Matrix metalloproteinases: a
review of their structure and role in acute coronary syndrome.
Cardiovasc Res 2003;59:81223.
35. Stetler-Stevenson WG, Brown PD, Onisto M, Levy AT, Liotta LA. Tis-
sue inhibitor of metalloproteinases-2 (TIMP-2) mRNA expression in
tumor cell lines and human tumor tissues. J Biol Chem 1990;265:
139338.
36. Sbai O, Ould-Yahoui A, Ferhat L, et al. Differential vesicular distribu-
tion and trafficking of MMP-2, MMP-9, and their inhibitors in astro-
cytes. Glia 2010;58:34466.
37. Hollborn M, Stathopoulos C, Steffen A, Wiedemann P, Kohen L,
Bringmann A. Positive feedback regulation between MMP-9 and
VEGF in human RPE cells. Invest Ophthalmol Vis Sci 2007;48:
43607.
38. Cowden Dahl KD, Symowicz J, Ning Y, et al. Matrix metalloprotei-
nase 9 is a mediator of epidermal growth factor-dependent E-
cadherin loss in ovarian carcinoma cells. Cancer Res 2008;68:
460613.
39. Zeisberg M, Maeshima Y, Mosterman B, Kalluri R. Renal fibrosis. Ex-
tracellular matrix microenvironment regulates migratory behavior of
activated tubular epithelial cells. Am J Pathol 2002;160:20018.
40. Onodera M, Zen Y, Harada K, et al. Fascin is involved in tumor necrosis
factor-α-dependent production of MMP9 in cholangiocarcinoma. Lab
Invest 2009;89:126174.
41. Redondo-Munoz J, Escobar-Diaz E, Samaniego R, Terol MJ, Garcia-
Marco JA, Garcia-Pardo A. MMP-9 in B-cell chronic lymphocytic
leukemia is upregulated by α
4
β
1
integrin or CXCR4 engagement via
distinct signaling pathways, localizes to podosomes, and is involved
in cell invasion and migration. Blood 2006;108:314351.
42. Milde-Langosch K. The Fos family of transcription factors and their
role in tumourigenesis. Eur J Cancer 2005;41:244961.
43. Ozanne BW, Spence HJ, McGarry LC, Hennigan RF. Transcription
factors control invasion: AP-1 the first among equals. Oncogene
2007;26:110.
44. Vial E, Marshall CJ. Elevated ERK-MAP kinase activity protects the
FOS family member FRA-1 against proteasomal degradation in colon
carcinoma cells. J Cell Sci 2003;116:495763.
45. Burch PM, Yuan Z, Loonen A, Heintz NH. An extracellular signal-
regulated kinase 1- and 2-dependent program of chromatin traffick-
ing of c-Fos and Fra-1 is required for cyclin D1 expression during cell
cycle reentry. Mol Cell Biol 2004;24:4696709.
46. Zhao J, Guan JL. Signal transduction by focal adhesion kinase in
cancer. Cancer Metastasis Rev 2009;28:3549.
Han et al.
Mol Cancer Res; 8(11) November 2010 Molecular Cancer Research1488
... The methylation level of the MMP9 gene remained consistent across the three periods, suggesting that methylation might not be the primary factor influencing MMP9 gene expression in collagen deposition and degradation. Other epigenetic mechanisms, such as histone modifications and transcription factor regulation, may also play roles [49]. ...
Article
Full-text available
Simple Summary This study investigates the impact of DNA methylation on collagen deposition in the skin of Dezhou donkeys, a breed valued for its thick, flexible skin with medicinal properties. Utilizing whole genome bisulfite sequencing (WGBS) and RNA sequencing (RNA-seq), the research analyzes the epigenetic landscape and gene expression profiles across three developmental stages of Dezhou donkeys. The study identifies numerous differentially methylated genes related to collagen deposition, such as COL1A1, COL1A2, and MMPs, highlighting an inverse relationship between gene expression and DNA methylation near transcription start sites. The findings of our study reveal the significant regulatory role of DNA methylation in skin collagen deposition, offering insights for genetic improvement and selective breeding to enhance skin quality in Dezhou donkeys. Our current research adds to the foundational knowledge of collagen deposition mechanisms, contributing to the fields of molecular biology and animal husbandry. Abstract DNA methylation represents a predominant epigenetic modification with broad implications in various biological functions. Its role is particularly significant in the process of collagen deposition, a fundamental aspect of dermal development in donkeys. Despite its critical involvement, the mechanistic insights into how DNA methylation influences collagen deposition in donkey skin remain limited. In this study, we employed whole genome bisulfite sequencing (WGBS) and RNA sequencing (RNA-seq) to investigate the epigenetic landscape and gene expression profiles in the dorsal skin tissues of Dezhou donkeys across three developmental stages: embryonic (YD), juvenile (2-year-old, MD), and mature (8-year-old, OD). Our analysis identified numerous differentially methylated genes that play pivotal roles in skin collagen deposition and overall skin maturation, including but not limited to COL1A1, COL1A2, COL3A1, COL4A1, COL4A2, GLUL, SFRP2, FOSL1, SERPINE1, MMP1, MMP2, MMP9, and MMP13. Notably, we observed an inverse relationship between gene expression and DNA methylation proximal to transcription start sites (TSSs), whereas a direct correlation was detected in regions close to transcription termination sites (TTSs). Detailed bisulfite sequencing analyses of the COL1A1 promoter region revealed a low methylation status during the embryonic stage, correlating with elevated transcriptional activity and gene expression levels. Collectively, our findings elucidate key genetic markers associated with collagen deposition in the skin of Dezhou donkeys, underscoring the significant regulatory role of DNA methylation. This research work contributes to the foundational knowledge necessary for the genetic improvement and selective breeding of Dezhou donkeys, aiming to enhance skin quality attributes.
... 18 In the same year, another research group showed that CADPE impedes cancer metastasis by regulating the expression of a focal adhesion kinase/mitogen-activated kinase/extracellular signal-regulated kinase (FAK/MEK/ERK)-mediated nuclear transcription factor activator protein-1 (AP-1) and matrix metalloproteinase-9 (MMP-9) in gastric carcinoma cell line. 19 However, there is still lack a study to prove that CADPE also has a potent anticancer activity in vivo. In our present study, we further demonstrated its anti-inflammation feature and anticancer activity in the mouse model with predicting mechanisms. ...
... Mechanistically, we have provided evidence that the activation of FAK/MEK-1/ERK1/2 signaling, which in turn induces MMP-9 expression, is involved in HK2-mediated regulation of cell migration and invasion. This pathway is linked to the formation and turnover of focal adhesions, which is vital for tumor cell invasion and metastasis [18], including in ovarian [29,30] and gastric [31] cancers. Moreover, uPA and VEGF expression have both been shown to be associated with tumor stage, metastasis and patient survival in ovarian cancer [32,33]. ...
Article
Full-text available
Metabolic reprogramming is a common phenomenon in cancers. Thus, glycolytic enzymes could be exploited to selectively target cancer cells in cancer therapy. Hexokinase 2 (HK2) converts glucose to glucose-6-phosphate, the first committed step in glucose metabolism. Here, we demonstrated that HK2 was overexpressed in ovarian cancer and displayed significantly higher expression in ascites and metastatic foci. HK2 expression was significantly associated with advanced stage and high-grade cancers, and was an independent prognostic factor. Functionally, knockdown of HK2 in ovarian cancer cell lines and ascites-derived tumor cells hindered lactate production, cell migration and invasion, and cell stemness properties, along with reduced FAK/ERK1/2 activation and metastasis- and stemness-related genes. 2-DG, a glycolysis inhibitor, retarded cell migration and invasion and reduced stemness properties. Inversely, overexpression of HK2 promoted cell migration and invasion through the FAK/ERK1/2/MMP9 pathway, and enhanced stemness properties via the FAK/ERK1/2/NANOG/SOX9 cascade. HK2 abrogation impeded in vivo tumor growth and dissemination. Notably, ovarian cancer-associated fibroblast-derived IL-6 contributed to its up-regulation. In conclusion, HK2, which is regulated by the tumor microenvironment, controls lactate production and contributes to ovarian cancer metastasis and stemness regulation via FAK/ERK1/2 signaling pathway-mediated MMP9/NANOG/SOX9 expression. HK2 could be a potential prognostic marker and therapeutic target for ovarian cancer.
Article
Full-text available
Focal adhesion plaques (FAPs) play an important role in the communication between cells and the extracellular matrix (ECM) and in cells’ migration. FAPs are macromolecular complexes made by different proteins which also interact with matrix metalloproteinases (MMPs). Because of these fundamental properties, FAPs and MMPs are also involved in cancer cells’ invasion and in the metastatic cascade. The most important proteins involved in FAP formation and activity are (i) integrins, (ii) a complex of intracellular proteins and (iii) cytoskeleton proteins. The latter, together with MMPs, are involved in the formation of filopodia and invadopodia needed for cell movement and ECM degradation. Due to their key role in cancer cell migration and invasion, MMPs and components of FAPs are often upregulated in cancer and are thus potential targets for cancer therapy. Polyphenols, a large group of organic compounds found in plant-based food and beverages, are reported to have many beneficial healthy effects, including anticancer and anti-inflammatory effects. In this review, we discuss the growing evidence which demonstrates that polyphenols can interact with the different components of FAPs and MMPs, inhibit various pathways like PI3K/Akt, lower focal adhesion kinase (FAK) phosphorylation and decrease cancer cells’ invasiveness, leading to an overall antitumoral effect. Finally, here we highlight that polyphenols could hold potential as adjunctive therapies to conventional cancer treatments due to their ability to target key mechanisms involved in cancer progression.
Article
Full-text available
Background and aim Gastric cancer (GC) is a common malignant tumor worldwide. Modified Gui-shao-liu-jun-zi decoction (mGSLJZ) is a clinically effective traditional Chinese medicine (TCM) compound in GC treatment. This study aimed to analyze main chemical substances of mGSLJZ and investigate active ingredients and molecular mechanism of mGSLJZ against GC. Experimental procedure HPLC-Q-TOF-MS/MS was used to analyze chemical substances of mGSLJZ, and potential active ingredients were screened from TCMSP. The target set of mGSLJZ for GC was obtained based on SwissTargetPrediction. The PPI network was constructed to screen out core targets. GO and KEGG enrichment analyses were conducted to identify BPs, CCs, MFs and pathways. The "active ingredient-core target-pathway" regulatory network was constructed to obtain core substances. Subsequently, Oncomine, Proteinatlas and molecular docking were performed to validate these findings. The cell experiments were conducted to confirm the anti-GC effects of mGLSJZ. Results and conclusion Forty-one potential active ingredients were filtered out from 120 chemical substances in mGSLJZ, including various organic acids and flavonoids. The top 10 key targets, 20 related pathways and 6 core medicinal substances were obtained based on network pharmacology analysis. Molecular docking results indicated that the core substances and key targets had good binding activities. The cell experiments validated that mGSLJZ and the core substances inhibited the proliferation in multiple GC cells and that mGLSJZ restrained the migration of GC. Meanwhile, the top 5 targets and top 2 pathways were verified. The rescue experiments demonstrated that mGSLJZ suppressed the proliferation and migration of GC through the PI3K/AKT/HIF-1 pathway.
Thesis
Full-text available
Les travaux de thèse exposés dans ce manuscrit portent sur l'étude chimique de plantes du genre Psiadia. Deux axes de recherche ont été menés parallèlement. Le premier a été consacré aux criblages chimique et biologique de cinq espèces du genre Psiadia endémiques de Madagascar. Le criblage chimique avait pour objectif d’identifier les biomarqueurs de ces espèces. Il comprend, d’une part, des analyses par CCM, CLHP-UV-CAD et RMN 1H, pour l’étude du métabolome non volatil, et d’autre part, des analyses par CG-SM et CG-DIF pour l’étude des composés volatils. Le criblage biologique a permis, quant à lui, de déceler les espèces présentant des propriétés biologiques prometteuses. Les cibles biologiques choisies étaient le parasite Plasmodium falciparum, pour l’activité anti-paludique, les macrophages de souris stimulés par un composé pro-inflammatoire pour l’activité anti-inflammatoire et les lignées cellulaires cancéreuses HepG2 (cancer de foie humain) et HT 29 (cancer du côlon) pour l’activité cytotoxique. À l'issue de ce criblage, l’ensemble des extraits, riches en métabolites, a révélé un potentiel antipaludique, anti-inflammatoire et cytotoxique. Le deuxième axe de recherche a été consacré à l’étude phytochimique détaillée d’une espèce endémique de La Réunion (P. dentata) et d’une espèce endémique de Madagascar (P. lucida), sélectionnées en raison de leurs fortes activités biologiques. L'extraction, l'isolement et l'élucidation structurale de leurs métabolites ont été entrepris par différentes techniques chromatographiques (CLMP, CLHP…) et spectroscopiques (UV, IRTF, SMHR, RMN 1D et 2D). Trente-quatre métabolites spécialisés appartenant à la famille des acides hydroxycinnamiques, des polyacétylènes, des flavonoïdes, des diterpènes et des coumarines ont été isolés de ces deux plantes. Seize sont de structures nouvelles. La valorisation des molécules isolées a ensuite été envisagée via l'évaluation de leurs activités biologiques. Vingt-sept composés se sont montrés actifs contre le parasite P. falciparum (isokaempféride,3α-angéoyloxy-19,16(R)-dihydroxy-ent-labda-8(17),13-dién-15,16-olide, … ), dix ont montré une activité anti-inflammatoire et se sont révélés en même temps être non toxiques pour les cellules saines (3α-angéoyloxy-19,15(R)-dihydroxy-ent-andrograpanine, 3(R),8(R)-déhydrofalcarindiol, …) et sept ont manifesté une cytotoxicité prometteuse sur cellules HepG2 et HT 29 (11-hydroxy-3(R),8(R)-déhydrofalcarindiol, ermanine, …). Par ailleurs, une approche bio-informatique par génération de réseaux moléculaires à partir des analyses CLUHP-SM2 des fractions issues des deux espèces a permis de supposer la présence de 28 autres composés tels que le falcarinol et l’acide labdanolique, connus pour être bioactifs.
Chapter
In osteoporosis, the density and quality of bone are reduced and become more fragile, with a significantly increased risk of fracture. Living tissue of bone undergoes continuous dynamic changes maintaining homeostasis by a delicate balance between osteoclasts and osteoblasts. Old bone is removed by osteoclasts (resorption), while new bone is added to the skeleton by osteoblasts (bone formation); an imbalance between these factors increases the risk of osteoporosis. Differentiation of osteoclasts and osteoblasts is strictly regulated by multiple cytokines, such as IL-1, IL-6, TNF-α, and TGF-β. Moreover, monocyte/macrophage colony stimulating factor and receptor activator of nuclear factor κB ligand induce osteoclast differentiation. In addition to these cytokines, it has become clear that reactive oxygen species regulate the differentiation of osteoclasts and osteoblasts. Antioxidant phytochemicals like carotenoids, polyphenols, and organosulfur compounds have been found to reduce the risk of oxidative stress-related diseases, such as cancer, arteriosclerosis, and osteoporosis. We focus on phytochemicals as antiosteoporotic agents in osteoporosis, and their antioxidative role via nuclear factor erythroid–derived related factor 2/Kelch-like ECH-associated protein pathway.
Article
Full-text available
There is an increasing interest in natural products and their derivatives with therapeutic benefits and less side effects compared to steroid therapy. Benzofuran derivatives display biological effects including anti-inflammatory effects. The present study aims to investigate whether (3-(7-methoxy-2-p-tolyl benzofuran-5-yl) propan-1-ol) (DK-1108), new synthetic benzofuran compound exerts anti-asthmatic effects in vitro and in vivo. DK-1108 strongly reduced the production of inflammatory mediators, cytokines and chemokines in RAW264.7 and A549 cells. DK-1108 significantly regulated the levels of AKT/MAPKs/c-Jun activation, AP-1 luciferase activity and ICAM-1 expression. Furthermore, DK-1108 effectively suppressed the adhesion of A549 and EOL-1 cells. In OVA-induced asthmatic mice, DK-1108 decreased the levels of IL-5/IL-13/IgE production, eosinophils/macrophages influx, ICAM-1/MCP-1 expression, mucus secretion and airway hyperresponsiveness (AHR). These effects of DK-1108 were accompanied by downregulation of MAPKs activation. Therefore, we suggest that DK-1108 exerts protective effect against airway inflammation and mucus overproduction, and therefore could be valuable therapeutic agent for treatment in asthma.
Article
Background Caffeic acid 3,4-dihydroxyphenethyl ester (CADPE) is a natural polyphenolic ester isolated as a minor component from a water extract of the Chinese medicine Zhongjiefeng [Sarcandra glabra (Thunb.) Nakai (Chloranthaceae)] and has previously shown to have activity against solid tumors through the modulation of multiple targets or signal pathways. However, the activity and potential mechanism of CADPE against leukemia cells have not yet been characterized. Purpose To investigate whether and how CADPE kills leukemia cells. Method (1) The activity of CADPE inhibiting the growth of different leukemia cell lines was evaluated by MTT assay; (2) Cell cycle arrest and apoptosis induced by CADPE were determined by flow cytometry with FlowJo software for quantification; (3) The protein levels were analyzed by Western blot and ubiquitin-binding c-Myc was acquired by co-immunoprecipitation. Results CADPE exerted potent activity against different leukemia cell lines with low toxicity in normal cells. In terms of mechanism of action, CADPE promoted ubiquitin-proteasome-dependent degradation of c-Myc through activating glycogen synthase kinase-3β (GSK3β) and downregulating deubiquitinating enzyme USP28 to trigger the interaction of c-Myc with ubiquitin ligase Fbw7, resulting in the downregulation of cell cycle regulators and anti-apoptotic proteins and consequently, cell cycle arrest and cell apoptosis. Conclusion CADPE is a novel c-Myc inhibitor with high activity and a unique mechanism for killing leukemia cells.
Article
Full-text available
Human tissue inhibitor of metalloproteinase-2 (TIMP-2) was cloned and sequenced from an A2058 human melanoma cell cDNA library. When the sequence was compared with that of human TIMP-1 at both the nucleotide and deduced amino acid levels, the homology appeared closer at the protein level than at the nucleotide level, suggesting that these inhibitors diverged early in the evolution of this gene family. Comparison of the deduced amino acid sequence for TIMP-2 with that of human TIMP-1 shows that there are two regions in which the similarity is below the overall average of 66%. It is postulated that these regions are responsible for the unique ability of TIMP-2 to bind to the latent form of the 72-kDa type IV collagenase. Polyclonal anti-TIMP-2 antisera recognized TIMP-2 but not TIMP-1 on immunoblotting. Northern blot analysis of RNA from A2058 human melanoma, HT-144 human melanoma, HT-1080 human fibrosarcoma, and WI-38 fetal lung fibroblast cell lines demonstrated two distinct transcripts of 1.0 and 3.5 kilobases (kb) for timp-2 mRNA. Both transcripts are down-regulated in response to transforming growth factor-beta but are unchanged in response to phorbol ester treatment. This is in contrast to the up-regulation of timp-1 transcripts by these agents and indicates that timp-2 and timp-1 are independently regulated in cell culture. Northern blot analyses of matched normal and tumor tissue samples from five cases of human colorectal carcinoma were performed. Normal and tumor tissues contain both the 1.0- and 3.5-kb transcripts. However, in the tissue samples the ratio of the 3.5-kb transcript to the 1.0-kb transcript was markedly elevated. No evidence of down-regulation of timp-2 transcript levels was noted in the tumor tissues. This is in contrast to the elevated timp-1 transcript levels seen in these tumor samples. Thus, timp-2 mRNA transcript levels are differentially regulated from timp-1 levels in vivo as well as in cell culture.
Article
Full-text available
Malignant gliomas are highly invasive tumors with an almost invariably rapid and lethal outcome. Surgery and chemoradiotherapy fail to remove resistant tumor cells that disperse within normal tissue, which are a major cause for disease progression and therapy failure. Infiltration of the neural parenchyma is a distinctive property of malignant gliomas compared with other solid tumors. Thus, glioma cells are thought to produce unique molecular changes that remodel the neural extracellular matrix and form a microenvironment permissive for their motility. Here, we describe the unique expression and proinvasive role of fibulin-3, a mesenchymal matrix protein specifically upregulated in gliomas. Fibulin-3 is downregulated in peripheral tumors and is thought to inhibit tumor growth. However, we found fibulin-3 highly upregulated in gliomas and cultured glioma cells, although the protein was undetectable in normal brain or cultured astrocytes. Overexpression and knockdown experiments revealed that fibulin-3 did not seem to affect glioma cell morphology or proliferation, but enhanced substrate-specific cell adhesion and promoted cell motility and dispersion in organotypic cultures. Moreover, orthotopic implantation of fibulin-3-overexpressing glioma cells resulted in diffuse tumors with increased volume and rostrocaudal extension compared with controls. Tumors and cultured cells overexpressing fibulin-3 also showed elevated expression and activity of matrix metalloproteases, such as MMP-2/MMP-9 and ADAMTS-5. Taken together, our results suggest that fibulin-3 has a unique expression and protumoral role in gliomas, and could be a potential target against tumor progression. Strategies against this glioma-specific matrix component could disrupt invasive mechanisms and restrict the dissemination of these tumors.
Article
Full-text available
Fascin is an actin-binding protein involved in the cell motility. Recently, aberrant expression of fascin in carcinoma cells was reported to participate in their invasive growth in cooperation with proteinases such as matrix metalloproteinases (MMPs). This study examined the participation of fascin in the progression of cholangiocarcinoma (CC) with reference to MMPs and tumor necrosis factor-alpha (TNF-alpha). Expression levels of fascin and MMP2 and 9 were examined immunohistochemically in human non-neoplastic biliary epithelium (13 cases) and CC (87 cases). The relationship between fascin and MMP9-expression levels was examined using two CC cell lines (CCKS-1 and HuCCT1). It was also examined whether or not fascin was involved in TNF-alpha-induced overproduction of MMP9 in CC. Fascin and MMP9 were expressed in 49 and 53% of CC samples, respectively, and the expression of these genes was frequent in intrahepatic CC. Fascin expression was correlated significantly with MMP9 expression. In particular, these two molecules were expressed more intensely at the invasive fronts of CC. Fascin expression was an unfavorable prognostic factor for patients with intrahepatic CC. In vitro studies showed that TNF-alpha could induce the overexpression of fascin and MMP9 in two CC cell lines. A knockdown study of fascin by siRNA showed that TNF-alpha induced the overproduction of fascin, which in turn upregulated MMP9 expression. Overexpression of fascin may have an important function in the progression of CC, and fascin expression might be involved in the signaling pathway in TNF-alpha-dependent production of MMP9 in CC.
Article
The transcription factor activator protein 1 (AP-1) plays a pivotal role in tumorigenesis. AP-1 activity is increased in multiple human tumor types. Inhibitors of AP-1 have been shown to block tumor promotion, trans formation, progression and invasion. Chronic inflammation and tumor development are linked. AP-1 may act, in part, by perpetuating the inflammatory signal. AP-1 is a recognized molecular target of many antioxidant and anti-inflammatory chem,chemopreventive compounds. This review focuses on the AP-1 family proteins as targets for chemoprevention.
Article
Focal adhesion kinase (FAK) has received much attention as a transducer of integrin-mediated signals. FAK becomes autophosphorylated on tyrosine 397 (Y397) in response to cell adhesion to the extracellular matrix (ECM) and in response to certain growth factors. Src-family kinases bind to FAK Y397 and mediate the phosphorylation of several additional tyrosine residues located within FAK. This leads to an enhancement of FAK activity and the recruitment of Grb2-SOS to the FAK signalling complex with subsequent activation of the Ras/ERK growth pathway in some cell types. Other signalling molecules such as Crk-associated substrate, Crk and paxillin can physically associate with FAK in response to adhesion to the ECM. FAK also seems to mediate cell migration and cell survival. Evidence obtained from several laboratories demonstrates that FAK is overexpressed in a variety of human cancers. FAK is not a classical oncogene and its overexpression appears to be a relatively late change in tumour progression. Because FAK can positively regulate the growth, migration and survival of cultured cells, it is likely that overexpression of FAK can contribute to the invasive/metastatic phenotype seen in late stage cancers. Therefore, FAK has received much attention as a point of therapeutic intervention in human cancers. The purpose of this article is to review the biology of FAK to date and to determine whether FAK inhibition might provide the much needed ‘magic bullet’ in cancer therapeutics.
Article
Lactate-dehydrogenase-5 (LDH-5) is an important isoenzyme converting pyruvate to lactate under hypoxic conditions and might play an important role in the development and progression of malignancies. However, the role of LDH-5 in gastric cancer is still unclear. In this study, we investigated the clinical significance of LDH-5 expression in gastric carcinoma. LDH-5 expression in 152 patients with different grade and stage gastric carcinoma was analyzed by immunohistochemistry. In addition, hypoxia-inducible factor 1alpha (HIF-1alpha) as a marker of tumor hypoxia, as well as vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) as angiogenesis parameters were also assessed in this study. Correlations between the expression of investigated proteins and various clinicopathological factors including survival were determined. There were 94 cases (61.8%) showing high LDH-5 expression, and 95 patients (62.5%) had high HIF-1alpha expression. Positive correlation was found between LDH-5 expression and HIF-1alpha, VEGF, and COX-2. The overexpression of LDH-5 was more prevalent in advanced tumors having positive vessel invasion. Patients with overexpression of LDH-5 showed far lower disease-free (63.5% vs 82.7%) and overall (56.3% vs 78.4%) survival rates compared with patients with low LDH-5 expression. HIF-1alpha expression was shown to have no significance on survival. In multivariate analysis, high LDH-5 expression kept its independence as a negative prognostic indicator. The results of the current study show that LDH-5 expression may be a useful prognostic factor for patients with gastric carcinoma.
Article
Extracellular proteolysis mediates tissue homeostasis. In cancer, altered proteolysis leads to unregulated tumor growth, tissue remodeling, inflammation, tissue invasion, and metastasis. The matrix metalloproteinases (MMPs) represent the most prominent family of proteinases associated with tumorigenesis. Recent technological developments have markedly advanced our understanding of MMPs as modulators of the tumor microenvironment. In addition to their role in extracellular matrix turnover and cancer cell migration, MMPs regulate signaling pathways that control cell growth, inflammation, or angiogenesis and may even work in a nonproteolytic manner. These aspects of MMP function are reorienting our approaches to cancer therapy.
Article
A high-yield synthesis of caffeic acid 3,4-dihydroxyphenethyl ester (1) has been achieved through Knoevenagel condensation of 3,4-dihydroxybenzaldehyde and 3,4-dihydroxyphenethyl monomalonate as the key step. Compound 1 was tested against a 56-cell-line cytotoxicity panel and for its free-radical-scavenging activity in the DPPH test.
Article
Astrocytes play an active role in the central nervous system and are critically involved in astrogliosis, a homotypic response of these cells to disease, injury, and associated neuroinflammation. Among the numerous molecules involved in these processes are the matrix metalloproteinases (MMPs), a family of zinc-dependent endopeptidases, secreted or membrane-bound, that regulate by proteolytic cleavage the extracellular matrix, cytokines, chemokines, cell adhesion molecules, and plasma membrane receptors. MMP activity is tightly regulated by the tissue inhibitors of MMPs (TIMPs), a family of secreted multifunctional proteins. Astrogliosis in vivo and astrocyte reactivity induced in vitro by proinflammatory cues are associated with modulation of expression and/or activity of members of the MMP/TIMP system. However, nothing is known concerning the intracellular distribution and secretory pathways of MMPs and TIMPs in astrocytes. Using a combination of cell biology, biochemistry, fluorescence and electron microscopy approaches, we investigated in cultured reactive astrocytes the intracellular distribution, transport, and secretion of MMP-2, MMP-9, TIMP-1, and TIMP-2. MMP-2 and MMP-9 demonstrate nuclear localization, differential intracellular vesicular distribution relative to the myosin V and kinesin molecular motors, and LAMP-2-labeled lysosomal compartment, and we show vesicular secretion for MMP-2, MMP-9, and their inhibitors. Our results suggest that these proteinases and their inhibitors use different pathways for trafficking and secretion for distinct astrocytic functions.