You are on page 1of 9

Research Article

Gonadotropin-Releasing Hormone Promotes Ovarian Cancer Cell


Invasiveness through c-Jun NH2-Terminal Kinase–Mediated
Activation of Matrix Metalloproteinase (MMP)-2 and MMP-9
1 2 1
Lydia W.T. Cheung, Peter C.K. Leung, and Alice S.T. Wong
1
Department of Zoology, University of Hong Kong, Hong Kong and 2Department of Obstetrics and Gynecology, University of British
Columbia, Vancouver, British Columbia, Canada

Abstract However, a role of GnRH in ovarian epithelial cell migration/


invasion has not been established and the mechanism by which
Gonadotropin-releasing hormone (GnRH) receptor is present
GnRH contributes to ovarian cancer invasiveness is also unknown.
in 80% of ovarian cancer, and numerous studies have provided
Ovarian cancer is the most lethal gynecologic malignant tumor
evidence for a role of GnRH in cell proliferation. In this study,
in the Western world. This is in part due to the fact that most (60%)

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


the effect of GnRH on the invasion potential of ovarian cancer
of the patients are diagnosed with already widespread intraperi-
cells was investigated. In vitro migration and cell invasion
toneal metastasis and ascites and the lack of effective therapies
assays with the ovarian cancer cell lines Caov-3 and OVCAR-3
for advanced-stage disease (5). Thus, there is a need for new
revealed the biphasic nature of GnRH; low concentrations of
therapeutic targets and a better understanding of the mechanisms
GnRH agonist (GnRHa) increased the cell motility and
involved in the spread of ovarian carcinoma. The factors that
invasiveness of these cells, but at increased concentrations,
regulate the metastatic process of ovarian cancer, however, are
the stimulatory effect was insignificant. Reverse transcription-
PCR, Western blot, and gelatin zymography showed that the poorly understood.
expression of metastasis-related proteinases, matrix metal- Matrix metalloproteinases (MMP), a family of secreted or
loproteinase (MMP)-2 and MMP-9, was up-regulated and transmembrane enzymes, can collectively digest almost all
activated by GnRHa. Moreover, we observed that GnRHa was extracellular matrix (ECM) and basement membrane components
(6, 7). Thus, MMPs are largely implicated in promoting angiogen-
able to transactivate the MMP-2 and MMP-9 promoters. The
esis and tumor metastasis. Above all, MMP-2 and MMP-9, which
invasive/migratory phenotype activated by GnRHa can be
can degrade collagen IV, the major ECM component of the
blocked by specific inhibitors or neutralizing antibodies to
basement membranes, have been suggested to be critical for the
MMP-2 and MMP-9. Knockdown of the GnRH receptor using
invasive and metastatic potential in ovarian carcinoma. Elevated
small interfering RNA significantly inhibited the GnRH-
expression of MMP-2 and MMP-9 has been detected in ovarian
induced MMP activation, invasion, and migration. In addition,
cancer ascites, tissues, and cancer cells in culture (8–11), and
we showed that the c-Jun NH2-terminal kinase, but not
experimental metastasis is suppressed by a synthetic MMP
extracellular signal-regulated kinase 1/2 or p38 mitogen-
inhibitor (12). MMP activity is often controlled by a specific
activated protein kinase, signaling pathway was critical for
inhibitor known as the tissue inhibitor of metalloproteinases
GnRH-mediated up-regulation of MMP, cell invasion, and
(TIMP). TIMP-1 selectively binds pro-MMP-9, whereas TIMP-2
motility. These results indicate for the first time an expanded
associates with pro-MMP-2 in a crucial step in the cell-mediated
role for GnRH in other aspects of ovarian tumor progression,
activation of MMPs (13). By inhibiting active MMPs, TIMPs
such as metastasis, via activation of MMP and the subsequent
inhibit cell invasion in vitro and tumorigenesis and metastasis
increase in cell migration and invasion. (Cancer Res 2006;
in vivo (14–16).
66(22): 10902-10)
In the present study, we show for the first time a role for GnRH
in the invasive phenotype and motility of human ovarian cancer
Introduction cells. We also present evidence suggesting that the activation of
The hypothalamic decapeptide gonadotropin-releasing hormone metastasis-related proteinases, especially MMP-2 and MMP-9,
(GnRH) plays a key role in the ovary and ovarian cancer (reviewed through the c-Jun NH2-terminal kinase (JNK) mediated signaling
in ref. 1). GnRH receptor expression has been shown in 80% of pathway in this process.
human ovarian tumors and numerous cancer cell lines, and the
analogues inhibit proliferation of the GnRH receptor-bearing Materials and Methods
tumor cells both in vivo and in vitro, supporting evidence for a
Cell culture. The human ovarian epithelial carcinoma cell lines Caov-3
direct antiproliferative effect (1–3). Of particular interest, levels of and OVCAR-3 (kindly provided by Dr. N. Auersperg, University of British
GnRH receptor seem to be associated with cancer grading and have Columbia, Vancouver, British Columbia, Canada) were grown in medium
been reported to be elevated in advanced-stage (stages III and IV) 199:105 (Sigma, St. Louis, MO) supplemented with 5% fetal bovine serum
ovarian carcinomas (4). This opens the possibility that GnRH could and 100 units/mL penicillin-streptomycin (Invitrogen, San Diego, CA) in a
directly regulate tumor progression of ovarian cancer cells. humidified atmosphere of 5% CO2 at 37jC.
Antibodies, reagents, and plasmids. GnRH agonist (GnRHa; D-Ala6)
was purchased from Sigma. PD98059, SB203850, SP600125, MMP-2 inhibitor
Requests for reprints: Alice S.T. Wong, Department of Zoology, University of (OA-Hy), MMP-9 inhibitor (SB-3CT), and monoclonal antibodies to
Hong Kong, 4S-14 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong.
Phone: 852-2299-0865; Fax: 852-2559-9114; E-mail: awong1@hku.hk.
MMP-2 and MMP-9 were purchased from Calbiochem (San Diego, CA).
I2006 American Association for Cancer Research. Anti-TIMP-1 and anti-TIMP-2 were from Chemicon International, Inc.
doi:10.1158/0008-5472.CAN-06-2217 (Temecula, CA). Anti-extracellular signal-regulated kinase 1/2 (ERK1/2),

Cancer Res 2006; 66: (22). November 15, 2006 10902 www.aacrjournals.org
GnRH Stimulates Ovarian Cancer Invasion

Figure 1. GnRH promotes ovarian cancer


cell migration and invasion. A, ovarian
cancer cells (1.5  106/mL) were seeded
on a Matrigel-precoated filter (8-Am pore)
of Transwell chambers in the presence or
absence of increasing concentrations
(0.1 nmol/L to 1 Amol/L as indicated) of
GnRHa. B, cell motility under same
treatment was measured through uncoated
filters, except that conditioned medium
from NIH-3T3 fibroblast culture was used in
the lower chamber as chemoattractant.
After 24 hours of incubation, cells in the

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


upper side of the filter were removed and
the invaded or migrated cells were fixed,
stained, and counted. Left, representative
pictures. Columns, mean number of
invaded or migrated cells of five fields of
triplicate wells from three independent
experiments; bars, SD. C, expression of
GnRH receptor (GnRHR ) in ovarian cancer
cell lines was detected by semiquantitative
PCR using primers specific for GnRH
receptor and h-actin. D, cells were
transiently transfected with vector
expressing short hairpin RNA directed
against GnRH receptor (2 Ag) or
nonspecific siRNA (NS siRNA ) as control.
Transfected cells were collected for
invasion assay through Matrigel (left) and
migration assay (right ) through uncoated
filter in the presence or absence of
0.1 nmol/L GnRHa. *, P < 0.05; **,
P < 0.005, compared with untreated
controls.

anti-p38 mitogen-activated protein kinase (MAPK), anti-JNK, anti- nonspecific siRNA control, scrambled siRNA duplex was used. Transfection
phosphorylated ERK1/2, anti-phosphorylated p38 MAPK, and anti- was done using LipofectAMINE 2000 reagent (Invitrogen) following the
phosphorylated JNK were obtained from Cell Signaling Technology, Inc. manufacturer’s instruction.
(Beverly, MA). The dominant-negative mutant of human JNK was kindly Gelatin zymography. The activities of MMP-2 and MMP-9 in the
provided by Dr. R. Davis (University of Massachusetts Medical School, conditioned medium were determined by gelatin zymography. Briefly, the
Worcester, MA). The MMP-2 promoter region between positions 1,659 and media were collected and clarified by centrifugation to remove cells and
+57 was cloned into promoterless luciferase reporter vector pGL3-Basic as debris. Samples (80 Ag) were loaded under nonreducing conditions onto
described previously (17). The MMP-9 promoter plasmid was a generous SDS-polyacrylamide gel polymerized with 1 mg/mL gelatin. Following
gift of Dr. D. Boyd (M. D. Anderson Cancer Center, Houston, TX). electrophoresis, the gels were washed with 2.5% Triton X-100 to remove SDS
Western blot analysis. Equal amounts of protein (50 Ag) were subjected and then incubated in a developing buffer [50 mmol/L Tris-HCl buffer
to SDS-PAGE (7.5%) and then transferred to nitrocellulose membrane. (pH 7.4), 10 mmol/L CaCl2] overnight at 37jC. Gels were stained with 0.25%
Membranes were blocked using 5% nonfat dry milk in PBS containing 0.05% Coomassie Brilliant Blue R-250 and destained in the same solution without
Tween 20 and incubated overnight at 4jC with the appropriate primary dye. Gelatinase activity was visualized as clear bands against the blue-
antibody. The immunocomplex was detected with horseradish peroxidase– stained gelatin background. Molecular sizes were determined from mobility
conjugated IgG as secondary antibody (Bio-Rad, Hercules, CA), and blots using gelatin zymography standards (Chemicon International). Three
were developed with an enhanced chemiluminescence kit (Amersham individual experiments were conducted with independent protein samples.
Biosciences, Little Chalfont, United Kingdom). The experiment was Semiquantitative reverse transcription-PCR. Total RNA was isolated
repeated at least twice with independent protein samples. from cultured cells using the Trizol reagent (Invitrogen) according to the
Small interfering RNA transfection. The pSUPER vector encoding 19- manufacturer’s procedure. First-strand cDNA synthesis was done from
mer hairpin small interfering RNA (siRNA) duplex specific to GnRH 2.5 Ag of total RNA using SuperScript Reverse Transcriptase (Life
receptor sequence 5¶-AGATCCGAGTGACGGTTAC-3¶ (nucleotides 107-125 Technologies). The cDNA was used for subsequent PCR using primers
downstream of the start codon) has been described elsewhere (18). As a specific for human GnRH receptor, MMP-2, MMP-9, TIMP-1, and TIMP-2 as

www.aacrjournals.org 10903 Cancer Res 2006; 66: (22). November 15, 2006
Cancer Research

reported previously (19, 20). h-Actin was used as internal control to not penetrated the filter were wiped out, and invaded cells on the lower
normalize the relative amount of cDNA in each reaction. The logarithmic surface of the filter were fixed with ice-cold methanol and stained with 0.5%
phase of PCR amplification for each target gene had been determined in crystal violet. Results are presented as the mean number of invaded cells of
initial experiments. The reproducibility of the quantitative measurements five fields FSD of three independent experiments. To assess cellular
was evaluated by three independent cDNA syntheses and PCR ran from migration potential, the protocol described above was used, except that
each preparation of RNA. Matrigel was omitted and 3T3 fibroblast conditioned medium was placed in
Reporter gene assay. Cells were transiently transfected with 1 Ag of the the lower chamber as a chemoattractant.
construct using LipofectAMINE (Invitrogen) as directed by the manufac- Statistical analysis. Statistical analysis was carried out using ANOVA
turer. After 6 hours, the cells were incubated with or without GnRHa followed by Tukey’s post hoc test (GraphPad Software, San Diego, CA).
(0.1 nmol/L) for an additional 24 hours. The pSV-h-galactosidase plasmid P < 0.05 was considered statistically significant.
was cotransfected as internal control. Luciferase units were calculated as
luciferase activity/h-galactosidase activity and are presented as the
mean FSD of three individual experiments with triplication. The fold Results
change was calculated by comparison with the promoterless luciferase
GnRH stimulates ovarian tumor cell migration and inva-
vector (pGL3-Basic).
sion. During metastasis, invasion of basement membrane by tumor
Invasion and migration assays. Twenty-four-well Transwell inserts
with 8-Am pores coated with Matrigel (50 AL/well; BD Biosciences, Palo cells is thought to be a critical event (21). To study whether
Alto, CA) were used to assess cell invasion. Cells (1  105) in serum-free expression of GnRH receptor is associated with metastasis of

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


medium, with or without GnRHa, were seeded in triplicate in the upper ovarian cancer cells, the effect of GnRH on cellular motility and
chamber. Serum-free medium was placed in the lower wells. The chambers invasive activity was evaluated. Because native GnRH has a
were incubated for 24 hours at 37jC in a 5% CO2 atmosphere. Cells that had relatively short half-life both in vivo and in vitro, GnRHa, in

Figure 2. GnRH induces production and


synthesis of MMP-2 and MMP-9. A, cells
were grown for 24 hours in the presence or
absence of increasing doses of GnRHa
(0.1 nmol/L to 1 Amol/L). Equal amounts
of protein (50 Ag) were then analyzed by
Western blot using specific antibodies
recognizing MMP-2 and MMP-9.
Immunoblotting for h-actin was included as
loading controls. The signal intensity was
determined by densitometry, and the levels
of MMP-2 and MMP-9 protein were
normalized against that of h-actin.
B, conditioned medium from treated cells
was also collected and analyzed for MMP
activities by gelatin zymography. Arrows,
gelatinase activities corresponding to
pro-MMP-2, active MMP-2, pro-MMP-9,
and active MMP-9. Lane 1, a human
MMP-2 and MMP-9 standard was used as
a positive control. C, cells were transiently
transfected with vector expressing short
hairpin RNA targeted against GnRH
receptor or nonspecific siRNA as control.
Left, aliquots of conditioned medium
normalized for same total protein
concentration were subjected to gelatin
zymography; right, cell lysates (50 Ag) were
analyzed by Western blot for the presence
of MMP-2 and MMP-9 proteins.
Immunoblotting for h-actin was included as
loading controls. D, cells were grown in
the presence or absence of 0.1 nmol/L
GnRHa for 24 hours and total protein was
harvested for Western blot analysis using
specific antibodies against TIMP-1,
TIMP-2, and h-actin. Experiments were
repeated thrice. Columns, mean; bars, SD.
*, P < 0.05, statistically significant
difference when treated samples were
compared with untreated controls.

Cancer Res 2006; 66: (22). November 15, 2006 10904 www.aacrjournals.org
GnRH Stimulates Ovarian Cancer Invasion

Figure 3. GnRH increases MMP-2 and


MMP-9 mRNA synthesis through modulation
of transcription but not mRNA stability.
A, cells were grown for 24 hours in the
presence or absence of increasing doses
of GnRHa (0.1 nmol/L to 1 Amol/L). Cells
were then harvested for semiquantitative
PCR with specific primers as described in
Materials and Methods. h-Actin served as an
internal control. B, cells were pretreated with
0.1 nmol/L GnRHa for 3 hours followed by
the post-treatment with 4 Ag/mL ActD over a
time course of 0, 1, 2, 4, and 6 hours. Cells
in the absence of GnRHa pretreatment
served as control. Total RNA was then
extracted and reversed transcribed followed
by PCR with MMP-2, MMP-9, and h-actin
sequence-specific primers. The signal
intensity was determined by densitometry,
and the level of MMP-2 and MMP-9 mRNA
was normalized against that of h-actin.

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


The MMP-2 and MMP-9 mRNA levels before
the addition of ActD were arbitrarily set to be
100%. C, cells (5  105) were plated in a
six-well plate and transfected with 1 Ag of
reporter constructs of MMP-2 and MMP-9
promoter. All cells were cotransfected with
0.5 Ag of pSV-h-galactosidase construct.
Eight hours after transfection, cells were
incubated with 0.1 nmol/L GnRHa for a
further 24 hours and harvested. Luciferase
and h-galactosidase activities were assayed,
and the luciferase activity of each sample
was normalized by h-galactosidase activity.
The relative luciferase activities were
calculated relative to the pGL3-Basic vector
(pGL3), which was arbitrarily assigned a
value of ‘‘1.’’ Experiments were repeated
thrice. Columns, mean; bars, SD.
*, P < 0.05; **, P < 0.005, compared with
untreated controls.

which Gly6 has been substituted by a D-amino acid, have been mRNA levels, with a maximal effective dose of 0.1 nmol/L (80.5%
commonly used to resist degradation and to increase potency (22). increase); thereafter, the levels decreased in cells treated with
The GnRHa D-Ala6 used in this study exhibits 3.5 to 4.5 times GnRHa at 100 nmol/L to 1 Amol/L. Cotreatment of cells with the
higher potency and 3 to 8 times higher stability than the native GnRH antagonist antide prevented the biphasic effects of GnRHa,
hormone in vitro (22, 23). Caov-3 and OVCAR-3 cells, which express whereas antide alone had no effect, indicating the specificity of the
functional GnRH receptors (24), were treated with increasing response (data not shown).
concentrations of the GnRHa (D-Ala6), and their invasive capacities To further confirm the role of GnRH receptor on the invasive
were assessed using Transwells with filters coated with Matrigel. properties of ovarian cancer cells, we inhibited the expression of
Our results showed that GnRHa induced a dose-dependent GnRH receptor using vector-based RNA interference assay. The
biphasic regulation on invasion: cells treated with GnRHa at low effectiveness of this siRNA to deplete GnRH receptor expression
doses of 0.1 to 10 nmol/L showed significantly higher numbers of was confirmed by semiquantitative reverse transcription-PCR (RT-
invaded cells (P < 0.05), whereas the response to higher doses of PCR) and Western blot analysis (data not shown) and was
GnRHa (100 nmol/L to 1 Amol/L) was insignificant (Fig. 1A). consistent with the previous study (18). Importantly, siRNA-
We also assessed in vitro migration in response to GnRH mediated depletion of GnRH receptor, but not nonspecific siRNA,
stimulus using a Transwell migration assay. GnRHa dose reduced invasion to near basal levels (80.8% inhibition; Fig. 1D, left).
dependently stimulated the migration of ovarian cancer cells Similarly, we showed that depletion of GnRH receptor inhibited the
through uncoated porous filter, with no significant effect at a cell migration by 85.2% (Fig. 1D, right), confirming a direct
concentration of 100 nmol/L and 1 Amol/L and a maximal effect at involvement of GnRH receptor in the acquisition of a migratory
0.1 nmol/L (P < 0.05; Fig. 1B). and invasive phenotype in ovarian cancer cells.
GnRH receptor siRNA inhibits ovarian tumor cell invasion GnRH induces MMP-2 and MMP-9 expression and enhances
and migration. Numerous studies have shown that alteration of their activities. We next determined whether GnRH-stimulated
GnRH receptor mRNA levels is involved in the regulation of ovarian cell invasion resulted from elevated levels of MMPs, which are well-
responsiveness to GnRH (19, 25, 26). We therefore examined documented ECM-degrading enzymes and whose activity is asso-
whether the effect of GnRH on ovarian cancer invasion was ciated with tumor invasiveness (27). MMP protein and enzymatic
associated with altered GnRH receptor gene expression. As shown activities were measured by Western blotting and gelatin
in Fig. 1C, treatment with the GnRHa induced a biphasic regulation zymography using conditioned medium from Caov-3 (Fig. 2) and
pattern for GnRH receptor mRNA in both Caov-3 and OVCAR-3. OVCAR-3 (data not shown). As shown in Fig. 2A, the effect of
Low doses of GnRHa (0.1-10 nmol/L) increased GnRH receptor GnRHa on MMP production was dose dependent because low

www.aacrjournals.org 10905 Cancer Res 2006; 66: (22). November 15, 2006
Cancer Research

concentrations (0.1-10 nmol/L) of GnRHa resulted in an up- was done. In accordance with changes in protein expression,
regulation of MMP-2/MMP-9 expression (P < 0.05), whereas high MMP-2 and MMP-9 mRNA expression were found to be signi-
concentration (100 nmol/L to 1 Amol/L) had minimal response ficantly enhanced with 0.1 nmol/L GnRHa (5.3-fold for MMP-2 and
(Fig. 2A). Consistently, gelatin zymography showed stronger lytic 2.5-fold for MMP-9 compared with untreated control cells; P < 0.05;
zones at the molecular mass corresponding to latent and active Fig. 3A).
forms of MMP-2 (72-kDa and 66-kDa forms) and MMP-9 (92-kDa To test whether the effect of GnRH on MMP-2 and MMP-9
and 86-kDa forms) in cells treated with 0.1 nmol/L GnRHa with mRNA expression was the result of increased mRNA stability, we
respect to untreated cells, showing their elevated secretion and did actinomycin D (ActD) chase experiments to determine the half-
enzyme activities by GnRHa (Fig. 2B). These results matched well life of MMP-2 and MMP-9 mRNA. Cells were preincubated with
with those observed in cellular invasion (Fig. 1), suggesting that 0.1 nmol/L GnRHa for 3 hours. Then, ActD (4 Ag/mL) was added to
GnRH-mediated induction of MMP-2 and MMP-9 may play a role stop transcription and RNA was isolated after 0, 1, 2, 4, and 6 hours.
in GnRH-enhanced cell invasion/migration. To ascertain whether The amount of gelatinase mRNA compared with h-actin was
GnRH plays a critical role in the induction of MMP-2 and MMP-9, determined by RT-PCR. As shown in Fig. 3B, GnRHa did not affect
we monitored the regulation of these proteinases after transfection the decay rate of the MMP-2 and MMP-9 mRNA.
of a GnRH receptor-targeted siRNA. As shown in Fig. 2C, both the To assess whether GnRHa causes transcriptional activation at
activity and expression of MMP-2 and MMP-9 were clearly reduced the MMP-2 and MMP-9 promoters, human MMP-2 or MMP-9

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


in the presence of the GnRH receptor siRNA. In contrast, control promoter-driven luciferase constructs were transiently transfected
experiments with nonspecific siRNA showed negligible effect. into Caov-3 and OVCAR-3 cells in either the presence or the
MMP expression is often coordinately regulated with production absence of 0.1 nmol/L GnRHa for 24 hours. The results showed
of their endogenous inhibitors, TIMPs (21). However, there was no that exposure of cells to GnRHa increased the activities of MMP-2
change in the expression of TIMP-1 and TIMP-2 irrespective of the and MMP-9 promoters by 10- and 3-fold, respectively (P < 0.005;
presence of different concentration of GnRHa as shown by Western Fig. 3C). Moreover, these increases in promoter activities were
blot analysis (Fig. 2D). completely abolished by GnRH receptor siRNA treatment but not
GnRH modulates transcription activities of MMP-2 and the nonspecific siRNA (Fig. 3C). These results collectively show a
MMP-9. To determine whether the observed alteration in MMP-2 role for GnRH in the transcriptional activation of MMP-2 and
and MMP-9 was the result of changes in gene expression, RT-PCR MMP-9.

Figure 4. Inhibition of MMP-2 and MMP-9


suppresses cell invasion and migration in
response to GnRH. Cells were left untreated
(control) or pretreated with OA-Hy
(15 Amol/L), SB-3CT (10 Amol/L), or
neutralizing MMP-9 antibody (anti-MMP-9 ;
15 Ag/mL) for 30 minutes. A, pretreated
cells (1.5  105) were seeded on
Matrigel-coated (50 Ag/mL) Transwell
chambers (8-Am pore) in the presence or
absence of 0.1 nmol/L GnRHa. After
24 hours, cells on the upper surface were
removed; the invaded cells were stained by
crystal violet and counted. B, in a parallel
experiment, cells that migrated through
uncoated filters were counted. Columns,
mean number of invaded/migrated cells of
five fields of triplicate wells from three
independent experiments; bars, SD.
**, P < 0.005, compared with untreated
controls.

Cancer Res 2006; 66: (22). November 15, 2006 10906 www.aacrjournals.org
GnRH Stimulates Ovarian Cancer Invasion

Inhibition of MMP blocks GnRH-enhanced invasion and GnRH-induced invasion and motility are JNK dependent. To
migration in ovarian cancer cells. To evaluate the potential assess the functional significance of GnRH-activated ERK1/2, JNK,
contribution of MMP-2 and MMP-9 in the GnRH-induced invasion and p38 MAPK in invasion and motility, we asked if interfering with
of ovarian cancer cells, we blocked the gelatinase activities using their activation by specific inhibitors diminishes GnRH-mediated
potent MMP-2 (OA-Hy, 15 Amol/L) or MMP-9 (SB-3CT, 10 Amol/L) in vitro invasiveness and motility. As shown, treatment of 50 Amol/L
specific inhibitors or MMP-9 neutralizing antibody (15 Ag/mL) in SP600125, a specific JNK inhibitor, significantly reduced the number
the presence or absence of 0.1 nmol/L GnRHa. Specific inhibition of of both invaded (Fig. 6A) and migrated (Fig. 6B) cells to near basal
MMP-2 by OA-Hy reversed the stimulatory effect of GnRH on cell levels. In contrast, inhibition of GnRH-stimulated ERK1/2 and p38
invasion (by 81.8% in Caov-3 and 82.9% in OVCAR-3), whereas MAPK activation by 50 Amol/L PD98059 or 10 Amol/L SB203580,
concomitant treatment with MMP-9 inhibitor SB-3CT or anti- respectively, had no effect (Fig. 6A and B). These results indicate a
MMP-9 significantly blocked the increase in invaded cells after critical role for JNK, but not ERK1/2 or p38 MAPK, in GnRH-induced
GnRHa treatment (by 75.6% and 72.7%, respectively, in Caov-3 and invasive phenotype and migration in ovarian cancer cells.
70.3% and 73.1%, respectively, in OVCAR-3; Fig. 4A). Similar results To additionally investigate the role for JNK in cellular invasion
were obtained in migration assays as shown in Fig. 4B. Together, and migration, we transfected Caov-3 and OVCAR-3 cells with a
these experiments show a crucial role for both MMP-2 and MMP-9 dominant-negative JNK (DN-JNK) construct in which the dual
in the GnRH-dependent invasion and migration. phosphorylation motif Thr-Pro-Tyr was mutated to Ala-Pro-Phe

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


Prolonged activation of JNK by GnRH. Because studies have (30). Specific inactivation of JNK in DN-JNK-transfected cells was
shown that the MAPK pathway is critical for the activation of gene confirmed by Western blot analysis (Fig. 6A, inset). Both the
expression by GnRH in some cell types (28, 29), we asked if they invasive phenotype and cell migration were clearly reduced in
also play a role in GnRH-induced human ovarian cancer cell DN-JNK transfectants as shown in Fig. 6A and B, showing the
invasion and/or migration. We first examined the levels of phos- significance of the JNK pathway in GnRH-induced invasion and
phorylated (active) forms of MAPK family members (ERK1/2, migration in Caov-3 and OVCAR-3 cells.
JNK, and p38 MAPK) in Caov-3 and OVCAR-3 cells treated with GnRH-mediated up-regulation of MMP-2 and MMP-9
0.1 nmol/L GnRHa for increasing time course of 0, 5, 15, 30, 60, and involves JNK. Next, we asked whether JNK mediates GnRH
120 minutes. As shown, addition of GnRHa to the cells significantly induction of MMP-2 and MMP-9 activities. As shown in Fig. 6C,
increased the phosphorylation of ERK1/2, JNK, and p38 MAPK both basal and GnRH-stimulated proform and active form of the
(Fig. 5). Interestingly, whereas prominent (15-fold) and sustained MMP-2 and MMP-9 production were inhibited with 50 Amol/L
activation (60 minutes) of JNK was shown in GnRH-activated cells SP600125. GnRH-induced MMP-2 and MMP-9 promoter activities
(P < 0.005), activation of ERK1/2 and p38 MAPK was rapid were also inhibited by targeted inhibition of JNK using SP600125 or
and transient, reaching a maximum of approximately 2- to 3-fold DN-JNK (Fig. 6D). Consistent with the effects on invasion and cell
15 minutes after GnRHa addition and decline thereafter (Fig. 5). migration, pretreatment with either PD98059 or SB203580 showed

Figure 5. Time course activation of


ERK1/2, p38 MAPK, and JNK by GnRH
stimulation. Cells were exposed to 0.1
nmol/L GnRHa for the indicated durations.
Activities of ERK1/2, p38 MAPK, and
JNK were determined by Western
blotting using antibodies specific for
phosphorylated, activated forms of ERK1/2
(A), p38 MAPK (B), and JNK (C). A to C,
bottom, Western blots using antibodies to
total ERK1/2, p38 MAPK, and JNK. The
relative intensity of phosphorylated forms of
ERK1/2, p38 MAPK, and JNK was
normalized to total values of ERK1/2, p38
MAPK, and JNK, which were determined by
densitometric analysis. Three identical
experiments independently done yielded
similar results. *, P < 0.05; **, P < 0.005,
compared with untreated controls.

www.aacrjournals.org 10907 Cancer Res 2006; 66: (22). November 15, 2006
Cancer Research

Figure 6. Inhibition of JNK inhibits


GnRH-induced cell invasion, migration,
and MMP expression. Cells were
pretreated with the ERK1/2 inhibitor
PD98059 (PD ; 50 Amol/L), the p38 MAPK
inhibitor SB203580 (SB ; 10 Amol/L), or the
JNK inhibitor SP600125 (SP ; 50 Amol/L)
for 30 minutes. In some cases, cells were
transiently transfected with either the
control vector (pcDNA-3 ) or DN-JNK
construct in the presence or absence of
GnRHa. Inset, whole-cell lysates were
analyzed for the levels of phosphorylated
and total forms of JNK by Western blot
analysis. A, cells were then allowed to
invade through the Matrigel-coated filters
for 24 hours in the presence or absence of
GnRHa (0.1 nmol/L). B, migration through

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


uncoated filters of the cells was measured.
Columns, mean number of invaded/
migrated cells of five fields of triplicate
wells from three independent experiments;
bars, SD. C, cells were either left untreated
(control) or pretreated with the indicated
kinase inhibitor for 30 minutes followed by
0.1 nmol/L GnRHa addition for another
24 hours. The MMP gelatinolytic activities
secreted into the medium were examined
by gelatin zymography. D, cells were
transiently transfected with either the
MMP-2 or the MMP-9 promoter/reporter
construct. Six hours after transfection, cells
were pretreated with the JNK inhibitor
SP600125 (50 Amol/L) for 30 minutes and
then incubated with 0.1 nmol/L GnRHa.
In some cases, cells were cotransfected
with DN-JNK as indicated. After 24 hours of
incubation, the cells were lysed and
luciferase activities were determined.
Luciferase values were normalized
for transfection efficiency (luciferase/
h-galactosidase ratios). The relative
luciferase activities were calculated relative
to the pGL3-Basic vector, which was
arbitrarily assigned a value of 1. Columns,
mean of three independent experiments;
bars, SD.

no inhibition on GnRH-mediated MMP induction (data not dependent manner, high concentrations were not as efficient.
shown). These results suggest that activation of JNK is required Alteration of GnRH receptor numbers or gene expression is
for enhanced MMP-2 and MMP-9 expression. involved in mechanisms underlying the biphasic effect of GnRH in
the pituitary and ovary (19, 25, 26, 31). In ovarian carcinoma cells,
Discussion we showed previously (25) and in this study that low doses of
GnRH up-regulated its receptor, whereas high doses decreased it.
There is much evidence that implicates an important role for
This also argues for a role for GnRH receptor to activate
GnRH in regulating the proliferation for a wide range of hormone-
intracellular signals necessary for cellular responses. Notably, the
dependent malignancies, including ovarian cancer (1). However,
effects of GnRH on JNK and MMP-2/MMP-9 activation were
little is known about its role in tumor invasion, metastasis, and
consistent with changes in the GnRH receptor mRNA levels. When
other aspects of cancer progression. In this study, we report for the
GnRH receptor mRNA was up-regulated, GnRH enhanced JNK and
first time that GnRH may contribute to the metastatic dissemina-
MMP-2/MMP-9 activities; however, when GnRH receptor mRNA
tion of ovarian cancer cells by promoting the expression and/or
was unaffected or down-regulated, GnRH had no effect on JNK3
processing of metastasis-related MMP, with subsequent down-
and MMP-2/MMP-9. Our finding that siRNA-mediated down-
stream changes in migratory and invasive behavior, providing an
regulation of the GnRH receptor inhibited MMP-dependent
insight into the prospect of developing targeted therapy for ovarian
invasion and cellular migration further upholds this view.
carcinoma.
The present study showed a biphasic effect of GnRH on cellular
migration and invasion. Whereas lower concentrations of the
3
GnRHa stimulated cellular migration and invasion in a dose- Unpublished data.

Cancer Res 2006; 66: (22). November 15, 2006 10908 www.aacrjournals.org
GnRH Stimulates Ovarian Cancer Invasion

Metastasis is a complex phenomenon that requires several The kinetic profile revealed differential regulation of ERK1/2, p38
specific steps, such as decreased adhesion, increased motility, and MAPK, and JNK by GnRH with a sustained signaling through the
proteolysis. In prostate cancer, GnRH has been shown to regulate JNK pathway. The duration of kinase activation is a major
cell motility through its interaction with the small GTPases Rac1, determinant for signal outcome in different cellular contexts (39),
Cdc42, and RhoA, which are involved in the regulation of actin and its influence may have included invasiveness. In support
polymerization (32). Our data show that another mechanism by of this hypothesis, GnRH-stimulated MMP expression and cell
which GnRH may promote tumor cell migration and invasion is invasion were attenuated by specific inhibition of JNK but not
through increased expression and proteolytic activities of MMP-2 ERK1/2 and p38 MAPK. Consistently, a recent report shows that
and MMP-9 that specifically degrade the basement membrane. activated JNK levels are much higher in the malignant pleural
MMP-2 and MMP-9 have been suggested to play critical roles in and peritoneal effusions of patients with advanced-stage ovarian
ovarian cancer metastasis, and up-regulation of MMP-2 and carcinomas, suggesting that activation of JNK may contribute to
MMP-9 is associated with increased invasion and poor prognosis a more invasive phenotype (40). However, unlike ERK1/2 and p38
in late-stage or invasive ovarian cancer patients (8–11). In addition MAPK, a role for JNK in tumor invasion or migration has not
to their enzymatic activities, MMP-2 and MMP-9 can also promote been widely reported. JNK signaling was shown recently to
tumor cell migration by influencing cytoskeletal organization regulate MMP-2 production and invasiveness in human gliomas
through their association with different families of adhesion (41). The JNK pathway targets multiple transcription factors,

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


receptors (33). They have also been reported to promote ascites including c-Jun and c-Fos (42), ATF (43), and PEA (44), and
formation in ovarian cancer cells via vascular endothelial growth putative binding sites for these DNA-binding proteins are present
factor production (34). Surprisingly, activation of MMP-2 and in the MMP promoters (45). Whether these putative regulatory
MMP-9 was not related to a decrease in the specific inhibitor of elements participate in the GnRH-dependent activation of the
TIMP-1 and TIMP-2 in the GnRH-induced ovarian cancer cell MMP-2 and MMP-9 genes remains to be determined.
invasion and motility. There was, however, an increase in MMP/ In summary, our findings emphasize the potential role of GnRH
TIMP ratios, which also in favor of ECM degradation. in promoting cellular migration and metastasis-related proteinase
One of the novel findings of the present study is the demons- activities in GnRH receptor-expressing ovarian tumors. This
tration that GnRH is a direct transcriptional activator of MMP-2 information provides a mechanistic rationale for the observed
and MMP-9 synthesis. MMP-2 and MMP-9 expression are reported GnRH receptor overexpression in advanced-stage ovarian carcino-
to be regulated at many levels, including gene activation, mRNA mas. The promising role for GnRH in invasion of ovarian cancer
stability, proenzyme activation, and inactivation by endogenous cells in vitro merits further investigation in vivo, which may provide
inhibitors in a complex fashion by numerous oncogene and tumor additional insight into its potential as a therapeutic molecular
suppressor pathways and conditions of hypoxia (35–38). In this target to decrease metastasis.
study, we showed that GnRH increased MMP-2 and MMP-9 mRNA
expression, and such expression was not due to changes in mRNA
stabilities. We further showed that GnRH directly induced MMP-2 Acknowledgments
and MMP-9 promoter activities. This is the first demonstration that
Received 6/16/2006; revised 8/24/2006; accepted 9/12/2006.
MMP-2 and MMP-9 are target genes of GnRH receptor activation Grant support: Hong Kong Research Grants Council grants 7484/04M and 7599/
and adding GnRH as a new member of MMP-2 and MMP-9 05M (A.S.T. Wong) and Canadian Institutes of Health Research grant 42441 (P.C.K.
transcriptional modulators. Leung).
The costs of publication of this article were defrayed in part by the payment of page
Our results identify a JNK signaling pathway that mediates charges. This article must therefore be hereby marked advertisement in accordance
GnRH-stimulated MMP expression, secretion, and cell invasion. with 18 U.S.C. Section 1734 solely to indicate this fact.

References proteinases in tumor invasion. Physiol Rev 1993;73: A synthetic matrix metalloproteinase inhibitor decreases
161–95. tumor burden and prolongs survival of mice bearing
1. Kang SK, Choi KC, Yang HS, Leung PC. Potential role 7. Fishman DA, Liu Y, Ellerbroek SM, Stack MS. human ovarian xenografts. Cancer Res 1993;53:2087–91.
of gonadotrophin-releasing hormone (GnRH)-I and Lysophosphatidic acid promotes matrix metalloprotei- 13. Brew K, Dinakarpandian D, Nagase H. Tissue
GnRH-II in the ovary and ovarian cancer. Endocr Relat nase (MMP) activation and MMP-dependent invasion in inhibitors of metalloproteinases: evolution, structure,
Cancer 2003;10:169–77. ovarian cancer cells. Cancer Res 2001;61:3194–9. and function. Biochim Biophys Acta 2000;1477:267–83.
2. Irmer G, Burger C, Muller R, et al. Expression of the 8. Moser TL, Young TN, Rodriguez GC, Pizzo SV, Bast RC, 14. Khokha R, Denhardt DT. Matrix metalloproteinases
messenger RNAs for luteinizing hormone-releasing Jr., Stack MS. Secretion of extracellular matrix-degrading and tissue inhibitor of metalloproteinases: a review of
hormone (LHRH) and its receptor in human ovarian proteinases is increased in epithelial ovarian carcinoma. their role in tumorigenesis and tissue invasion. Invasion
epithelial carcinoma. Cancer Res 1995;55:817–22. Int J Cancer 1994;56:552–9. Metastasis 1989;9:391–405.
3. Volker P, Grundker C, Schmidt O, Schulz KD, Emons G. 9. Young TN, Rodriguez GC, Rinehart AR, Bast RC, Jr., 15. Albini A, Melchiori A, Santi L, Liotta LA, Brown PD,
Expression of receptors for luteinizing hormone-releas- Pizzo SV, Stack MS. Characterization of gelatinases Stetler-Stevenson WG. Tumor cell invasion inhibited by
ing hormone in human ovarian and endometrial linked to extracellular matrix invasion in ovarian TIMP-2. J Natl Cancer Inst 1991;83:775–9.
cancers: frequency, autoregulation, and correlation with adenocarcinoma: purification of matrix metalloprotei- 16. DeClerck YA, Perez N, Shimada H, Boone TC, Langley
direct antiproliferative activity of luteinizing hormone- nase 2. Gynecol Oncol 1996;62:89–99. KE, Taylor SM. Inhibition of invasion and metastasis in
releasing hormone analogues. Am J Obstet Gynecol 10. Fishman DA, Bafetti LM, Banionis S, Kearns AS, cells transfected with an inhibitor of metalloproteinases.
2002;186:171–9. Chilukuri K, Stack MS. Production of extracellular Cancer Res 1992;52:701–8.
4. Chien CH, Chen CH, Lee CY, Chang TC, Chen RJ, Chow matrix-degrading proteinases by primary cultures of 17. Bian J, Sun Y. Transcriptional activation by p53 of the
SN. Detection of gonadotropin-releasing hormone human epithelial ovarian carcinoma cells. Cancer 1997; human type IV collagenase (gelatinase A or matrix
receptor and its mRNA in primary human epithelial 80:1457–63. metalloproteinase 2) promoter. Mol Cell Biol 1997;17:
ovarian cancers. Int J Gynecol Cancer 2004;14:451–8. 11. Davidson B, Goldberg I, Gotlieb WH, et al. The 6330–8.
5. Naora H, Montell DJ. Ovarian cancer metastasis: prognostic value of metalloproteinases and angiogenic 18. An BS, Selva DM, Hammond GL, Rivero-Muller A,
integrating insights from disparate model organisms. factors in ovarian carcinoma. Mol Cell Endocrinol 2002; Rahman N, Leung PC. Steroid receptor coactivator-3 is
Nat Rev Cancer 2005;5:355–66. 187:39–45. required for progesterone receptor trans-activation of
6. Mignatti P, Rifkin DB. Biology and biochemistry of 12. Davies B, Brown PD, East N, Crimmin MJ, Balkwill FR. target genes in response to gonadotropin-releasing

www.aacrjournals.org 10909 Cancer Res 2006; 66: (22). November 15, 2006
Cancer Research

hormone treatment of pituitary cells. J Biol Chem 2006; 28. Bonfil D, Chuderland D, Kraus S, et al. Extracellular sion associated with oncogene-mediated cellular
281:20817–24. signal-regulated kinase, Jun N-terminal kinase, p38, and transformation and metastasis formation. Cell Biol
19. Kang SK, Choi KC, Cheng KW, Nathwani PS, Auersperg c-Src are involved in gonadotropin-releasing hormone- Int 2001;25:411–20.
N, Leung PC. Role of gonadotropin-releasing hormone as stimulated activity of the glycoprotein hormone 37. Nair RR, Boyd DD. Expression cloning of novel
an autocrine growth factor in human ovarian surface follicle-stimulating hormone h-subunit promoter. regulators of 92 kDa type IV collagenase expression.
epithelium. Endocrinology 2000;141:72–80. Endocrinology 2004;145:2228–44. Biochem Soc Trans 2005;33:1135–6.
20. Zhou HY, Wong AS. Activation of p70S6K induces 29. Luo X, Ding L, Chegini N. Gonadotropin-releasing 38. Munoz-Najar UM, Neurath KM, Vumbaca F, Claffey
expression of matrix metalloproteinase 9 associated hormone and TGF-h activate MAP kinase and differen- KP. Hypoxia stimulates breast carcinoma cell invasion
with hepatocyte growth factor-mediated invasion in tially regulate fibronectin expression in endometrial through MT1-MMP and MMP-2 activation. Oncogene
human ovarian cancer cells. Endocrinology 2006;147: epithelial and stromal cells. Am J Physiol Endocrinol 2006;25:2379–92.
2557–66. Metab 2004;287:E991–1001. 39. Marshall CJ. Specificity of receptor tyrosine kinase
21. Liotta LA, Stetler-Stevenson WG. Tumor invasion 30. Derijard B, Hibi M, Wu IH, et al. JNK1: a protein signaling: transient versus sustained extracellular signal-
and metastasis: an imbalance of positive and negative kinase stimulated by UV light and Ha-Ras that binds regulated kinase activation. Cell 1995;80:179–85.
regulation. Cancer Res 1991;51:5054–9s. and phosphorylates the c-Jun activation domain. Cell 40. Davidson B, Konstantinovsky S, Kleinberg L, et al.
22. Karten MJ, Rivier JE. Gonadotropin-releasing hor- 1994;76:1025–37. The mitogen-activated protein kinases (MAPK) p38 and
mone analog design. Structure-function studies toward 31. Kaiser UB, Jakubowiak A, Steinberger A, Chin WW. JNK are markers of tumor progression in breast
the development of agonists and antagonists: rationale Regulation of rat pituitary gonadotropin-releasing carcinoma. Gynecol Oncol 2006;102:453–61.
and perspective. Endocr Rev 1986;7:44–66. hormone receptor mRNA levels in vivo and in vitro . 41. Hu B, Jarzynka MJ, Guo P, Imanishi Y, Schlaepfer DD,
23. Yang X, Rojanasakul Y, Wang L, Ma JY, Ma JK. Endocrinology 1993;133:931–4. Cheng SY. Angiopoietin 2 induces glioma cell invasion
Enzymatic degradation of luteinizing hormone releasing 32. Enomoto M, Utsumi M, Park MK. Gonadotropin- by stimulating matrix metalloprotease 2 expression
hormone (LHRH)/[D-Ala6]-LHRH in lung pneumocytes. releasing hormone induces actin cytoskeleton remodel- through the avh1 integrin and focal adhesion kinase
Pharm Res 1998;15:1480–4. ing and affects cell migration in a cell-type-specific signaling pathway. Cancer Res 2006;66:775–83.

Downloaded from http://aacrjournals.org/cancerres/article-pdf/66/22/10902/2555753/10902.pdf by guest on 21 October 2022


24. Kimura A, Ohmichi M, Kurachi H, et al. Role of manner in TSU-Pr1 and DU145 cells. Endocrinology 42. Cirillo G, Casalino L, Vallone D, Caracciolo A, De
mitogen-activated protein kinase/extracellular signal- 2006;147:530–42. Cesare D, Verde P. Role of distinct mitogen-activated
regulated kinase cascade in gonadotropin-releasing 33. Sanceau J, Truchet S, Bauvois B. Matrix metal- protein kinase pathways and cooperation between Ets-2,
hormone-induced growth inhibition of a human ovarian loproteinase-9 silencing by RNA interference triggers ATF-2, and Jun family members in human urokinase-
cancer cell line. Cancer Res 1999;59:5133–42. the migratory-adhesive switch in Ewing’s sarcoma cells. type plasminogen activator gene induction by interleu-
25. Kang SK, Cheng KW, Nathwani PS, Choi KC, Leung J Biol Chem 2003;278:36537–46. kin-1 and tetradecanoyl phorbol acetate. Mol Cell Biol
PC. Autocrine role of gonadotropin-releasing hormone 34. Belotti D, Paganoni P, Manenti L, et al. Matrix 1999;19:6240–52.
and its receptor in ovarian cancer cell growth. metalloproteinases (MMP9 and MMP2) induce the 43. Gupta S, Campbell D, Derijard B, Davis RJ. Tran-
Endocrine 2000;13:297–304. release of vascular endothelial growth factor (VEGF) scription factor ATF2 regulation by the JNK signal
26. Peng C, Fan NC, Ligier M, Vaananen J, Leung PC. by ovarian carcinoma cells: implications for ascites transduction pathway. Science 1995;267:389–93.
Expression and regulation of gonadotropin-releasing formation. Cancer Res 2003;63:5224–9. 44. O’Hagan RC, Tozer RG, Symons M, McCormick F,
hormone (GnRH) and GnRH receptor messenger 35. Sehgal I, Thompson TC. Novel regulation of type IV Hassell JA. The activity of the Ets transcription factor
ribonucleic acids in human granulosa-luteal cells. collagenase (matrix metalloproteinase-9 and -2) acti- PEA3 is regulated by two distinct MAPK cascades.
Endocrinology 1994;135:1740–6. vities by transforming growth factor-h1 in human Oncogene 1996;13:1323–33.
27. Coussens LM, Werb Z. Matrix metalloproteinases prostate cancer cell lines. Mol Biol Cell 1999;10:407–16. 45. Chakraborti S, Mandal M, Das S, Mandal A,
and the development of cancer. Chem Biol 1996;3: 36. Baruch RR, Melinscak H, Lo J, Liu Y, Yeung O, Chakraborti T. Regulation of matrix metalloproteinases:
895–904. Hurta RA. Altered matrix metalloproteinase expres- an overview. Mol Cell Biochem 2003;253:269–85.

Cancer Res 2006; 66: (22). November 15, 2006 10910 www.aacrjournals.org

You might also like