ArticlePDF Available

GPER functions as a tumor suppressor in triple-negative breast cancer cells

Authors:

Abstract and Figures

The orphan, membrane-bound estrogen receptor (GPER) is expressed at high levels in a large fraction of breast cancer patients and its expression is favorable for patients' survival. We investigated the role of GPER as a potential tumor suppressor in triple-negative breast cancer cells MDA-MB-231 and MDA-MB-468 using cell cycle analysis and apoptosis assay. The constitutive activity of GPER was investigated. GPER-specific activation with G-1 agonist inhibited breast cancer cell growth in concentration-dependent manner via induction of the cell cycle arrest in G2/M phase, enhanced phosphorylation of histone H3 and caspase-3-mediated apoptosis. Analysis of the methylation status of the GPER promoter in the triple-negative breast cancer cells and in tissues derived from breast cancer patients revealed that GPER amount is regulated by epigenetic mechanisms and GPER expression is inactivated by promoter methylation. Furthermore, GPER expression was induced by stress factors, such as radiation, and GPER amount inversely correlated with the p53 expression level. Overall, our results establish the protective role in breast cancer tumorigenesis, and the cell surface expression of GPER makes it an excellent potential therapeutic target for triple-negative breast cancer.
Content may be subject to copyright.
1 3
J Cancer Res Clin Oncol
DOI 10.1007/s00432-014-1620-8
ORIGINAL ARTICLE - CANCER RESEARCH
GPER functions as a tumor suppressor in triple‑negative breast
cancer cells
Christine Weißenborn · Tanja Ignatov · Hans‑Joachim Ochel · Serban Dan Costa ·
Ana Claudia Zenclussen · Zoya Ignatova · Atanas Ignatov
Received: 5 February 2014 / Accepted: 10 February 2014
© Springer-Verlag Berlin Heidelberg 2014
factors, such as radiation, and GPER amount inversely cor-
related with the p53 expression level.
Conclusions Overall, our results establish the protective
role in breast cancer tumorigenesis, and the cell surface
expression of GPER makes it an excellent potential thera-
peutic target for triple-negative breast cancer.
Keywords GPER · GPR30 · Breast cancer ·
Tumor suppression · TNBC
Introduction
Several distinct subtypes of invasive breast cancers, the
most common cancer types in women, have been identified
using microarray-based approach (Perou et al. 2000). One
of them, the triple-negative breast cancer (TNBC), is nega-
tive for estrogen, progesterone, and HER2 receptors, which
limits the options for optimal adjuvant therapy. The iden-
tification of potential therapeutic targets for this subtype
remains a clinical challenge (Perou et al. 2000; Schneider
et al. 2008).
More than 50 % of breast cancer patients express high
levels of the orphan G-protein-coupled receptor, GPR30,
a membrane-bound estrogen receptor (GPER) (Arias-
Pulido et al. 2010; Filardo et al. 2006; Ignatov et al. 2011;
Kuo et al. 2007; Liu et al. 2009; Tu et al. 2009), whose
expression is favorable for patients’ survival (Arias-Pulido
et al. 2010; Ignatov et al. 2011). Similar results were also
obtained for ovarian cancer patients (Ignatov et al. 2013a).
The function of GPER, however, in the disease pathology
is controversial and still a subject of intense debate. The
identification and first functional studies on GPER pro-
posed rapid nongenomic effects of estrogen (Prossnitz et al.
2008). Studies with high-affinity non-steroidal receptor
Abstract
Background The orphan, membrane-bound estrogen
receptor (GPER) is expressed at high levels in a large frac-
tion of breast cancer patients and its expression is favorable
for patients’ survival.
Methods We investigated the role of GPER as a poten-
tial tumor suppressor in triple-negative breast cancer cells
MDA-MB-231 and MDA-MB-468 using cell cycle analy-
sis and apoptosis assay. The constitutive activity of GPER
was investigated.
Results GPER-specific activation with G-1 agonist inhib-
ited breast cancer cell growth in concentration-dependent
manner via induction of the cell cycle arrest in G2/M phase,
enhanced phosphorylation of histone H3 and caspase-3-me-
diated apoptosis. Analysis of the methylation status of the
GPER promoter in the triple-negative breast cancer cells
and in tissues derived from breast cancer patients revealed
that GPER amount is regulated by epigenetic mechanisms
and GPER expression is inactivated by promoter methyla-
tion. Furthermore, GPER expression was induced by stress
C. Weißenborn · T. Ignatov · S. D. Costa · A. Ignatov (*)
Department of Obstetrics and Gynecology, University
of Magdeburg, Gerhart-Hauptmann Str 35, Magdeburg, Germany
e-mail: atanas.ignatov@gmail.com
C. Weißenborn · A. C. Zenclussen
Department of Experimental Obstetrics and Gynaecology,
University of Magdeburg, Magdeburg, Germany
H.-J. Ochel
Department of Radiotherapy, University of Magdeburg,
Magdeburg, Germany
Z. Ignatova
Department of Biochemistry and Biology, University of Potsdam,
Potsdam, Germany
J Cancer Res Clin Oncol
1 3
agonist G-1 and antagonist G-15 (Bologa et al. 2006; Den-
nis et al. 2009) suggest that GPER mediates the prolifera-
tive effects of estrogen in many estrogen-related cancers
(Albanito et al. 2007; Filardo et al. 2000; Ignatov et al.
2010b, c; Vivacqua et al. 2006). Conversely, studies in vari-
ous cell cultures show that GPER can act as an inhibitor
on cell growth and proliferation (Ariazi et al. 2010; Chan
et al. 2010; Gao et al. 2011; Holm et al. 2011; Wang et al.
2012). Thereby, GPER blocks the cell cycle progression
in G1 or G2/M phase of estrogen receptor-positive breast
cancer cells without any apoptotic effect (Ahola et al.
2002; Ariazi et al. 2010). In vivo, the GPER expression is
down-regulated during breast cancer progression, suggest-
ing the potential role of GPER in tumor suppression (Igna-
tov et al. 2013b). The influence of various environmental
and milieu-specific factors, including the presence or the
absence of estrogen (Fan et al. 2009; Ignatov et al. 2010c;
Leblanc et al. 2007), may explain in part those polarized
observations. An alternative plausible explanation might
be the fact that GPER-induced stimulation of cell prolif-
eration is mainly observed after receptor stimulation with
non-specific GPER agonists such as estrogen and tamox-
ifen (Albanito et al. 2008; Bologa et al. 2006; Filardo et al.
2000, 2002; Girgert et al. 2012; Ignatov et al. 2010c; Mag-
giolini et al. 2004). Importantly, the TNBC is negative for
estrogen receptor implying that underlying mechanisms
that control the GPER expression may significantly differ
than in other type of breast cancer.
Here, we demonstrate that GPER mediates the inhibi-
tory effect of G-1 on triple-negative breast cancer cells via
induction of the cell cycle arrest and cell apoptosis. GPER
expression is regulated by epigenetic mechanisms both in
vitro and in vivo. Moreover, GPER expression is stimulated
via radiation which acts as DNA-damaging agent. Our
results propose that GPER may act as a potential therapeu-
tic target in TNBC.
Materials and methods
Cell culture and treatment
MDA-MB-231, MDA-MB-468, MCF-7, and HEK-293
cells were obtained from Cell Lines Services (Germany)
and routinely cultured in DMEM/F12 (PAN Biotech)
supplemented with 10 % FBS, 100 U/ml penicillin, and
100 μg/μl streptomycin at 37 °C in a humidified 5 % CO2
atmosphere. If necessary, the cells were synchronized by
estrogen withdrawal for 24 h using phenol red-free medium
supplemented with 10 % charcoal-stripped steroid-depleted
FBS. Thereafter, the cells were treated as indicated in the
figure legends, and the cell count was measured with a
Coulter counter as described (Filardo et al. 2006).
MTT viability assay
MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazo-
lium bromide)-viability assay was performed as already
described (Ignatov et al. 2003). Briefly, 2,000 cells per well
were seeded and cultured in a 96-well plate in the growth
medium. After 24 h, the cells were stimulated for 3 days
with G-1 or DMSO as a control. Thereafter, MTT was
added for 3 h in the dark. The supernatants were removed,
cells were lysed in 150 μl lysis buffer (isopropanol con-
taining 4 mM HCl and 0.1 % NP-40), and the absorbance
at 570 nm was recorded.
Cell cycle analysis and apoptosis assay
Cells were treated with 1 μM G-1 or medium for 1–3 days.
Cell cycle distribution was analyzed by propidium iodide
(PI) staining using flow cytometry. Apoptosis was deter-
mined with FITC Annexin V Apoptosis Detection Kit (BD,
Heidelberg) following the manufacturer’s protocol.
Real-time RT-PCR
RNA isolation and quantitative RT-PCR were performed
as previously described (Schumacher et al. 2009). We used
the following primers for GPER identification: forward,
5-AGTCGGATGTGAGGTTCAG-3 and reverse, 5-TCT-
GTGTGAGGAGTACAAG-3. GPER mRNA levels were
normalized to the β-actin transcript levels. All reactions
were carried out on an iCycler (BioRad, Germany).
Western blot analysis
The following primary antibodies were used: sc-48524-R
for GPER (1:500 dilution; Santa Cruz), ab13847 for cas-
pase-3 (1:1,000 dilution; Abcam), OP43 for p53 (1:2,000
dilution; Calbiochem), #4138 for cyclin B1 (dilution
1:1,000; Cell signalling), #9112 for Cdc2 (dilution 1:2,000;
Cell signalling); #9701 phosphoH3 (Ser10) (dilution
1:2,000; Cell signalling), and A5441 for β-actin (dilution
1:10,000; Sigma-Aldrich). Peroxidase-conjugated anti-
rabbit or anti-mouse antibodies (Thermo Scientific), diluted
1:2,000 or 1:5,000, respectively, were used as secondary
antibodies.
siRNA treatment
GPER knockout experiments were performed as already
described (Ignatov et al. 2010c) after transfection of the
cells with GPER-specific siRNA (sc-60743, Santa Cruz),
p53-specific siRNA (sc-29435, Santa Cruz), or scram-
bled control siRNA (sc-37007, Santa Cruz). The trans-
fection was performed with Lipofectamine 2000 (Life
J Cancer Res Clin Oncol
1 3
Technologies) according to the manufacturer’s protocol.
Cells were analyzed 48 h after transfection.
Methylation analysis of GPER promoter region in breast
cancer cells and tissue
The methylation status of the GPER promoter region was
determined by methylation-specific PCR (MSP) as previ-
ously described (Ignatov et al. 2008, 2010a). The following
primers, designed with the MethPrimer software (Li and
Dahiya 2002), were used:
me thylated forward 5-GGTTAGTAGGGGGCGTAT
TC-3;
me thylated reverse 5-TAATTACGAATTTCACAATCT
CGTA-3;
un methylated forward 5-TAAATGGTTAGTAGGGGGT
GTATTT-3;
un methylated reverse 5-CATAATTACAAATTTCACAA
TCTCATA-3.
Breast cancer cells, breast cancer tissues, and normal
breast specimens were analyzed. DNA was isolated with
NucleoSpin® DNA extraction kit (Macherey & Nagel).
The initial bisulfite reaction that converts unmethylated
cytosines to uracils was performed with the CpGenome
DNA modification Kit (S7820, Chemicon). The MSP con-
sists of two individual PCRs with distinct primers specific
for methylated and unmethylated DNA sequences. We used
100 nmol/l specific forward and reverse primers, 2 μl of
2 mmol/l dNTPs, 5 μl PCR buffer, 0.75 U of GoTaq poly-
merase (Promega), and 100 ng of template DNA in a final
volume of 25 μl. The PCR conditions were as follows:
95 °C for 10 min; followed by 40 cycles of denaturation at
94 °C for 1 min, annealing at 55 °C for 1 min, and amplifi-
cation at 72 °C for 1 min. The PCR products were analyzed
on 3 % agarose gel stained with ethidium bromide.
Methylated and unmethylated DNA sequences were
always included in the analysis. Gels were interpreted only
in case of working positive control. The methylation of
GPER promoter was made using yes/no decision.
Radiation treatment of breast cancer cells
MDA-MB-231, MDA-MB-468, and MCF-7 cells were
grown on culture plates to 70–80 % confluence and irra-
diated with 0, 2, 5, and 10 Gy at room temperature. The
experiments were performed after 48-h incubation.
Bioinformatic tools and statistical analysis
The following software tools were used to predict methyla-
tion sites in the 5-regions upstream of the transcription start:
http://www.ebi.ac.uk/Tools/emboss/cpgplot/index.html;
http://bio.dfci.harvard.edu/Methylator/.
The curve fit and nonlinear regression analysis of the
dose–response curves were performed with the GraphPad
Prism software. Data are present as mean ± SD. In all sta-
tistical analyses, two-sided tests were applied. Statistical
significance was determined by Student’s t-test, and p val-
ues <0.05 were considered as statistically significant.
Results
GPER stimulation with G-1 inhibited the growth
of triple-negative breast cancer cells via cell cycle arrest
in the M-phase and caspase-dependent cell apoptosis
Triple-negative MDA-MB-231 and MDA-MB-468 breast
cancer cell lines were stimulated with increasing concen-
trations of GPER-specific agonist G-1 for 3 days. G-1
caused a concentration-dependent inhibition of the cell
growth with an IC50 value of 0.1 μM for MDA-MB-231
and 0.3 μM for MDA-MB-468 cells (Fig. 1a). Notably,
MDA-MB-231 cells expressed lower levels of GPER
mRNA than the MDA-MB-468 cells (Fig. 1b). To con-
firm the specificity of GPER agonist G-1, we next down-
regulated the GPER expression with siRNA. Intriguingly,
the down-regulation of GPER in the absence of G-1 was
associated with increased cell growth in MDA-MB-231
and MDA-MB-468 cells. These data demonstrated the
constitutive activity of GPER. In both MDA-MB-231 and
MDA-MB-468 cells, the specific knockdown of the GPER
expression abrogated the inhibitory effect of G-1 (Fig. 1c).
The effect was specific to the triple-negative MDA-
MB-231 and MDA-MB-468 breast cancer cells; no effect
was observed in the control HEK-293 cells lacking GPER
(Fig. 1c).
To assess the effect of G-1-stimulated GPER on the cell
cycle, we next analyzed the distribution of the cells in dif-
ferent phases of the cell cycle using propidium iodide (PI)
staining. The stimulation of MDA-MB-231 and MDA-
MB-468 cells with 1 μM G-1 led to a significant accumula-
tion of cells in G2/M phase compared to the control cells
(Fig. 2a and b). The cell cycle arrest in the G2/M phase
was already detectable after 24 h of G-1 stimulation and
slightly decreased after 48 h (Fig. 2a and b). Notably, a
subpopulation of cells accumulated in the apoptotic sub-G1
phase upon G-1 treatment; the effect was stronger in MDA-
MB-468 than in MDA-MB-231 cells (Fig. 2a and b).
To disentangle the nature of GPER-induced cell cycle
arrest in more details, we next investigated the expression
of G2/M-phase-specific regulatory proteins cyclin B1 and
Cdc2 and the phosphorylation of histone H3. G-1-induced
cell cycle arrest enhanced the phosphorylation of histone
J Cancer Res Clin Oncol
1 3
H3, which is indicative of cells in mitotic phase. The effect
was detectable even 6 h after G-1 stimulation of the cells
and reached the highest value between 12 and 24 h (Fig. 2c
and d). However, the levels of cyclin B1 and Cdc2 protein
remained unchanged (Fig. 2c and d). GPER knock down
by siRNA abrogated the G-1-induced cell cycle arrest and
apoptosis (data not shown), suggesting again that G-1
effects are mediated by GPER.
The observed accumulation of a sizeable fraction of
cells in the apoptotic sub-G1 phase (Fig. 2a and b) led
us determining the effect of GPER on cell apoptosis.
The number of apoptotic cells significantly increased in
both MDA-MB-231 and MDA-MB-468 cells upon GPER
stimulation with G-1 compared to the background of
apoptotic cells among the non-stimulated cells (Fig. 3a
and b): The number of apoptotic cells increased to
13.95 % in MDA-MB-231 cells (Fig. 3a) and to 24.34 %
in MDA-MB-468 cells (Fig. 3b). In addition, pro-cas-
pase-3 and caspase-3 increased in both MDA-MB-231
and MDA-MB-468 cells (Fig. 3c and d). Taken together,
these results suggest that G-1-induced GPER activation
inhibits the proliferation of triple-negative breast cancer
cells via cell cycle arrest in the M-phase and caspase-
dependent cell apoptosis. These analyses were performed
in four additional cell lines: MCF-7, SK-BR-3, BT-2, and
MDA-MB-453. The G-1 inhibitory effect with cell cycle
arrest and stimulation of cell apoptosis was observed in
all cell lines investigated.
Cell growth (% of control)
0
50
100
150
-4-6-8-10
MDA-MB-231
MDA-MB-468
A
0
1
2
Relative mRNA expression
levels of GPER
MDA-MB-231 MDA-MB-468
B
C
Cell number (x105l)
0
100
300
400
+1µM G-1
HEK-293MDA-MB-231
p= 0.0013 p= 0.0087
MDA-MB-468
+1µM G-1+1µM G-1
G-1 concetration, log [M]
MDA-MB-468
MDA-MB-231
GPER
β-actin
GPER
β-actin
200
p= 0.0413
p= 0.0140
Fig. 1 G-1 inhibits the growth of the triple-negative breast cancer
cells. a MTT viability assay of MDA-MB-231 and MDA-MB-468
cells treated with different concentrations of G-1 for 3 days. The
number of proliferating cells is normalized to the untreated (control)
cells. b qRT/PCR of the mRNA expression level of GPER presented
as means of three independent replicates ±SD. GPER signal was nor-
malized to β-actin transcript levels. c Silencing of the GPER expres-
sion abrogate the inhibitory effect of G-1. MDA-MB-231 and MDA-
MB-486 cells were transiently transfected with GPER-specific siRNA
and followed by treatment with 1 μM G-1 for 72 h. Thereafter, cell
number was counted by cell counter. The expression level was nor-
malized to cells transfected with scrambled siRNA (control) and is
presented as means of three experiments ±SD. HEK-293 cells lack-
ing GPER were treated in the same way and served as control. Inset
cells transfected with scrambled or GPER-specific siRNA. β-Actin
was used as a loading control. Each experiment was repeated at least
three times
J Cancer Res Clin Oncol
1 3
Promoter methylation controls GPER expression
The observed inhibition of cell proliferation of the triple-
negative breast cancer cells raised the intriguing question as
to whether GPER can act as a potential tumor suppressor in
breast cancer. The clear dose-dependent inhibitory effect of
the GPER on the cell growth suggested to us that tuning the
promoter by methylation may determine the GPER expres-
sion levels in the cell. We used 5-Aza-2-deoxycytidine
(5-Aza) to inhibit the DNA-methyltransferase, which is
involved in the methylation of the promoter. Intriguingly,
the treatment with 5-Aza enhanced the GPER mRNA level
in both MDA-MB-231 and MDA-MB-468 cells in a time-
dependent manner (Fig. 4a and b).
To address whether methylation-dependent increase in the
GPER expression has the same effect on cell proliferation as
the G-1-induced GPER expression, we pre-treated the two
cell lines with 5 μM 5-Aza for 24 h and then incubated them
with 1 μM G-1 for 72 h. Pre-treatment with 5-Aza showed
synergistic inhibitory effect to G-1 abrogation of the cell
growth: The growth of MDA-MB-231 and MDA-MB-468
cells was reduced down to ~5 % by combined treatment with
5-Aza and G-1 compared to the treatment with a single drug.
These data clearly suggest that the inhibitory effect of the
GPER is proportional to its expression level and significantly
decreases upon promoter methylation.
Next, we analyzed the 5-region upstream of the transla-
tion start site of GPER using MethPrimer software (Li and
B
A
0
20
40
60
80
Sub G1
G-1 stimulation (h)
024 48 72 0244872024 48 72 0244872
G0/G1S G2/M
p= 0.0032
p= 0.0002
p= 0.0084
p= 0.0404
p= 0.0032
p= 0.0239
p= 0.0129
p= 0.0287
p= 0.0424
p= 0.0369
p= 0.0071
0
20
40
60
80
p= 0.0258
p= 0.0171
C
C
cyclin B1
cdc2
G-1CG-1C G-1
0h 24h 48h72h
β-actin
CG-1 CG-1 CG-1
0h 24h48h 72h
C
phospho-H3
β -actin
G-1C G-1C G-1
0h 1h 6h 24h
CG-1 CG-1 CG-1
0h 1h 6h 24h
Percentage of cells
Percentage of cells
Sub G1
G-1 stimulation (h)
024 48 72 0244872024 48 72 0244872
G0/G1S G2/M
D
cyclin B1
cdc2
β -actin
phospho-H3
β -actin
p= 0.0009
11.06 0.94 0.98 2.73 1.42 6.72 10.90 1.85 0.72 4.16 0.41 2.10
11.11 1.44 0.97 1.17 1.04 1.13
11.16 1.52 0.78 1.13 0.59 0.52
11.11 1.26 0.97 1.17 1.04 1.08
11.09 1.14 0.96 0.94 0.75 0.79
Fig. 2 G-1-stimulated GPER inhibits the cell cycle in triple-negative
breast cancer cells. Flow cytometry analysis of the distribution of
MDA-MB-231 (a) and MDA-MB-468 (b) cells in different phases
of the cell cycle after treatment with 1 μM G-1 or control for dif-
ferent times. The results are presented as means of three independ-
ent experiments ±SD. Western blot analysis of expression of cyclin
B1 and Cdc2 and phosphorylation of histone 3 in MDA-MB-231 (c)
and MDA-MB-468 (d) cell stimulated with 1 μM G-1 or control for
different times. β-Actin was used as a loading control. Each Western
blot is a representative example of three independent experiments.
The numbers above the blots represent the relative expression units
compared to the control. The control was set as 1
J Cancer Res Clin Oncol
1 3
Dahiya 2002) to find any potential CpG island(s). The hyper-
methylation of CpG islands in promoter region of tumor sup-
pressor genes is associated with their inactivation (Esteller
et al. 2001). In the region between 4,365 and 4,797 nt
upstream of the transcription start site, we identified a single
433-bp-long CpG-reach island (Fig. 5a). Methylation-specific
PCR approach confirmed experimentally the methylation
within the GPER promoter region in both breast cancer cell
lines and primary breast cancer tissue (Fig. 5b). We analyzed
the breast tissue of 30 breast cancer patients and compared it
to a cohort of healthy individuals of the same size. In 17 out
of 30 patients (56.7 %), the GPER promoter was methylated,
while the normal breast tissues contained only non-methyl-
ated GPER promoters. Furthermore, we also tested whether
5-Aza alters the expression of GPER because of changing the
methylation status of the GPER promoter. Notably, pre-treat-
ment with 5-Aza led to a significant decrease in GPER meth-
ylation in MDA-MB-231 (p = 0.010) and MDA-MB-468
cells (p = 0.009) (Fig. 5c), thus corroborating the enhanced
GPER expression we observed in both triple-negative breast
cancer cells (Fig. 4b and c).
Stress-induced expression of GPER in breast cancer cells
Important function of the tumor suppressor genes is to
induce cell apoptosis and/or to arrest cell cycle progression
in response to DNA-damaging agents, such as radia-
tion (Sun and Yang 2010). We therefore hypothesized that
GPER as a potential tumor suppressor may also play a role
in this processes. We next exposed the MDA-MB-231 and
MDA-MB-468 cells to various doses of γ-radiation for
48 h and measured the levels of GPER mRNA. In both
cell lines, we observed a clear dose-dependent increase
in the GPER mRNA levels (Fig. 6a); the effect was much
stronger in MDA-MB-231 cells. In addition, the expres-
sion of the tumor suppressor p53 protein was also elevated
upon radiation in both cell lines (Fig. 6b). Note that MDA-
MB-231 and MDA-MB-468 cells express a non-functional
mutant of p53 (Kastan et al. 1991; Lim et al. 2009). Thus,
we included in the analysis MCF-7 breast cancer cells that
express wild-type p53 protein (Lim et al. 2009). Intrigu-
ingly, in MCF-7 cells, we observed the opposite effect:
Upon radiation the, GPER expression decreased progres-
sively with the increasing dosage of radiation (Fig. 6a).
Thus, we hypothesized that p53 may modulate the expres-
sion pattern of GPER in an opposite direction. To test this
hypothesis, p53 gene was silenced with a specific siRNA in
MCF-7 cells, and the expression of GPER upon exposure
of the cells to γ-radiation was monitored. p53 knockdown
led to a significant increase in GPER mRNA expression
after radiation (Fig. 6a), implying the direct role of wild-
type p53 in GPER expression. In addition, incubation with
072
Cell apoptosis (%)
p = 0.0007
p = 0.0064
p = 0.0264
0
15
30
45
A
C
B
Pro-caspase-3
Caspase-3
β-actin
24 48
07
2
Cell apoptosis (%)
p = 0.0026
p = 0.0015 p = 0.0005
0
15
30
45
24 48
Time (h)Time (h)
CG-1 CG-1
0h 24h 48h
MDA-MB-231 MDA-MB-468
D
Pro-caspase-3
Caspase-3
β-actin
CG-1 CG-1
0h 24h48h
Control
G-1
Control
G-1
10.98 1.20.93 2.3111.71.33.6
Fig. 3 G-1 induces caspase-dependent apoptosis in triple-negative
breast cancer cells. The fraction of apoptotic cells increases in MDA-
MB-231 (a) and MDA-MB-468 (b) cells upon treatment with 1 μM
G-1 for different times. The apoptotic cells were stained with annexin
V/propidium iodide, counted by flow cytometry, and normalized to
the number of untreated cells. The results are presented as means of
three independent replicates ±SD. The level of pro-caspase-3 and
caspase-3 was enhanced in MDA-MB-231 (c) and MDA-MB-468 (d)
cells upon treatment with 1 μM G-1 for different times as indicated
by Western blot. Time zero represents the basal expression of cas-
pase-3 before the addition of medium (c) and G-1 to the cells. β-actin
was used as loading control. Each Western blot is a representative
example of three independent experiments. The numbers above the
blots represent the relative expression units compared to the control.
The control was set as 1
J Cancer Res Clin Oncol
1 3
G-1 raised the p53 expression level in all three cell lines
(Fig. 6c), corroborating the functional association between
p53 and GPER.
Discussion
Here, we present an analysis on the effect of GPER on tri-
ple-negative breast cancer cells. Our observations clearly
suggest that (1) GPER-specific agonist inhibited breast can-
cer cell proliferation in concentration-dependent manner,
(2) GPER activation leads to a cell cycle arrest and apop-
tosis in cell-dependent fashion, (3) GPER expression is
down-regulated in breast cancer tumorigenesis, (4) aberrant
GPER expression is caused by a reversible promoter meth-
ylation both in vivo and in vitro, and (5) GPER expression
is induced by stress factors such as radiation.
GPER, a member of the GPCR family, is implicated in
breast, endometrial, and ovarian cancers (Filardo et al.
2006; Fujiwara et al. 2012; Smith et al. 2007, 2009). A large
number of GPCR receptors, which are involved in various
types of human cancers, are associated with an increased
cell proliferation and tumor progression (Dorsam and
Gutkind 2007). Our data clearly suggest that GPER acti-
vation in triple-negative breast cancer cells resembles the
growth-specific alterations described for GPER in other
cancer types (Chan et al. 2010; Chimento et al. 2013; Gao
et al. 2011; Holm et al. 2011; Ignatov et al. 2013a; Wang
et al. 2012). The cell growth suppression through enhanced
GPER expression occurs via specific cell cycle arrest in the
M-phase and caspase-3-mediated apoptosis. The effect is
potentiated by the G-1 agonist, which is highly specific for
the triple-negative breast cancer cells and is not a result of
general cytotoxic effect as no growth defect was observed in
GPER-1-negative HEK293 cells. Although Wang et al. have
very recently demonstrated that G-1-induced ovarian cancer
cell inhibition is GPER-independent (Wang et al. 2013), our
observation suggested a GPER-dependent activity of G-1,
and these results have been confirmed by various resent
observations (Ariazi et al. 2010; Chan et al. 2010; Chimento
et al. 2013; Holm et al. 2011; Ignatov et al. 2013a). Unlike
the estrogen receptor-positive MCF-7 breast cancer cells,
for which G-1-induced growth inhibition through G1-phase
block has been described (Fujiwara et al. 2012; Gao et al.
2011), we did not observe any indications of G1-phase alter-
ations in the triple-negative breast cancer cells. Our results
0
2
4
6
Relative mRNA expression
levels of GPR30
8
B
p= 0.0322
p= 0.0163
p= 0.0116
A
0
2
4
6
Relative mRNA expression
levels of GPR30
0244872
Time (h)
p= 0.0297 p= 0.0313
0
50
100
150
Cell proliferation
(% of control)
p= 0.0338
p= 0.0489
5-Aza -+ -+
G-1 --+ +
D
p= 0.0016
p= 0.0157
C
Control
5-Aza
Control
5-Aza
0244872
Time (h)
0
50
100
150
Cell proliferation
(% of control)
5-Aza -+ -+
G-1 --+ +
Fig. 4 Inhibition of promoter methylation enhances GPER level.
qRT-PCR quantification of the mRNA expression of GPER in MDA-
MB-231 (a) and MDA-MB-468 (b) cells upon treatment with 5 μM
5-Aza for different times. Control cells were treated the same way
in a medium without 5-Aza. MTT-viability assay of MDA-MB-231
(c) and MDA-MB-468 (d) cells treated with 5 μM 5-Aza for 48 h,
followed by stimulation with 1 μM G-1 for 72 h. Non-treated cells
served as a control, and their viability was arbitrarily set as 100 %.
The results are presented as means of three independent replicates
±SD
J Cancer Res Clin Oncol
1 3
clearly show that the growth inhibition in MDA-MB-231
and MDA-MB-468 occurs rather through alterations of
the G2/M phase through an increase in the phosphoryla-
tion of histone H3, an important step occurring during G2
to M transition (Hans and Dimitrov 2001). The expression
of the other G2/M-phase-specific regulatory proteins cyclin
B1 and Cdc2 remains unchanged, which is in contrast to the
findings in estrogen receptor-positive breast cancer cells and
prostate cancer cells (Ariazi et al. 2010; Chan et al. 2010).
The increased mitotic duration, which perturbs mitotic pro-
gression, triggers cell apoptosis via caspase-3 cleavage—an
effect that has been already described for both microtubule
inhibitors with anticancer activity, taxol, and nocodazole
(Choi et al. 2011; Toh et al. 2010). We also observed a
G-1-induced apoptosis in MCF-7, an effect that has not been
described yet for these cells, implying that apoptosis might
be a general mechanism to cease cell growth and prolifera-
tion in cancer cells upon activation of the GPER expression.
GPER is ubiquitously encoded in normal breast tissues,
but its expression is silenced through a hypermethylation of
the promoter. Our systematic analysis on the methylation
status of the promoter in cell culture and primary breast
tissue derived from breast cancer patients revealed that
the GPER expression is tightly regulated by epigenetic
mechanisms through methylation and demethylation of
the promoter. Although we have performed the analysis
with a sizeable cohort of patients, to further confirm the
link between promoter methylation and GPER expression
and extract the clinical significance, much larger cohorts of
patients need to be analyzed.
Further important finding of our study is the radiation-
induced alterations in the GPER expression in different
breast cancer cells; the radiation-induced expression is a
characteristic feature of various tumor suppressor genes
(Fei and El Deiry 2003). Strikingly, GPER expression
showed opposing expression in different cells: upregu-
lated GPER expression in MDA-MB-231 and MDA-
MB-468 cells and down-regulated in MCF-7 cells. This
pattern is linked most likely to the form of the p53 protein
expressed in each cell. While MDA-MB-231 and MDA-
MB-468 cells express a non-functional p53 gene, MCF-7
cells have normal wild-type p53 (Kastan et al. 1991; Tam
B
UM-Primer
A
ATG
-4797
5‘
CpG island (433bp)
3‘
1 3
-4365
2
5‘
M (for) -4659
UM (for) -4664
M (rev) -4543
UM (rev) -4541
C1UM M234 56
M-Primer
M-Primer
UM-Primer
CUMM
MDA-MB-231
MDA-MB-468
C
MDA-MB-468
M-Primer
UM-Primer
C5-Aza
M-Primer
MDA-MB-231
C5-Aza
0
1
2
p=0.010
0
1
2p=0.009
Relative
expression
Relative
expression
200
100
200
100
200
100
200
100
Fig. 5 GPER promoter is methylated in triple-negative breast can-
cer cells and in primary breast cancer tissue. a Schematic of the
GREP gene. The putative 433-bp-long CpG island between positions
4,365 and 4,797 is indicated. Gray boxes delineate exon 1, 2, and
3; arrows denote the positions of the MSP specific primers; verti-
cal lines mark the individual CpG sites. The analysis was performed
with a DNA sequence corresponding to the region between 1126443
to 1133451 nt of the chromosome 7 (NCBI Reference sequence
NC_000007.13). b Representative examples of the methylation
analysis of GPER promoter in breast cancer cells and tissue. c Meth-
ylation status of GPER promoter in MDA-MB-231 (left) and MDA-
MB-468 (right) cells upon pre-treatment with 5 μM 5-Aza for 48 h.
The numbers on the left denote the DNA ladder in bp. M-primer,
primer specific for the methylated CpG sites; UM-primer, primer
specific for unmethylated CpG sites. C, negative control; M, standard
methylated control; UM, standard unmethylated control; lanes 1–6 in
panel C, six randomly chosen exemplary cases of primary breast can-
cer tissues. 1, 3, and 6, examples of methylated GPER promoter; 2, 4,
and 5, examples of unmethylated GPER poromoter
J Cancer Res Clin Oncol
1 3
et al. 1994). siRNA-induced knockdown of the wild-type
p53 in MCF-7 cells reverses the GPER expression phe-
notype to those of the MDA-MB-231 and MDA-MB-468
cells. Thus, intact wild-type p53 exerts a protective role: It
counteracts the GPER-induced expression upon radiation.
Moreover, G-1-induced activation of the breast cancer cell
lines enhances the expression of p53. Therefore, we sug-
gest a feedback mechanism between the expression of p53
and GPER: GPER activation increases the p53 expression,
which in turn down-regulates the expression of GPER. In
case of a non-functional p53, GPER expression remains
high and an alternative emergency mechanism is activated,
which induces cell apoptosis. The cell surface expression
of GPER makes it an excellent target for TNBC.
Acknowledgments This work was supported by Deutsche
Krebshilfe.
Conflict of interest This work was supported by a grant from
Deutsche Krebshilfe to A.I.
References
Ahola TM, Manninen T, Alkio N, Ylikomi T (2002) G protein-cou-
pled receptor 30 is critical for a progestin-induced growth inhibi-
tion in MCF-7 breast cancer cells. Endocrinology 143:3376–3384
Albanito L, Madeo A, Lappano R, Vivacqua A, Rago V, Carpino
A, Oprea TI, Prossnitz ER, Musti AM, Ando S, Maggiolini M
(2007) G protein-coupled receptor 30 (GPR30) mediates gene
expression changes and growth response to 17beta-estradiol and
selective GPR30 ligand G-1 in ovarian cancer cells. Cancer Res
67:1859–1866
Albanito L, Sisci D, Aquila S, Brunelli E, Vivacqua A, Madeo A, Lap-
pano R, Pandey DP, Picard D, Mauro L, Ando S, Maggiolini M
(2008) Epidermal growth factor induces G protein-coupled recep-
tor 30 expression in estrogen receptor-negative breast cancer
cells. Endocrinology 149:3799–3808
Arias-Pulido H, Royce M, Gong Y, Joste N, Lomo L, Lee SJ, Chaher
N, Verschraegen C, Lara J, Prossnitz ER, Cristofanilli M (2010)
GPR30 and estrogen receptor expression: new insights into hor-
mone dependence of inflammatory breast cancer. Breast Cancer
Res Treat 123:51–58
Ariazi EA, Brailoiu E, Yerrum S, Shupp HA, Slifker MJ, Cunliffe
HE, Black MA, Donato AL, Arterburn JB, Oprea TI, Prossnitz
ER, Dun NJ, Jordan VC (2010) The G protein-coupled receptor
GPR30 inhibits proliferation of estrogen receptor-positive breast
cancer cells. Cancer Res 70:1184–1194
Bologa CG, Revankar CM, Young SM, Edwards BS, Arterburn JB, Kise-
lyov AS, Parker MA, Tkachenko SE, Savchuck NP, Sklar LA, Oprea
TI, Prossnitz ER (2006) Virtual and biomolecular screening con-
verge on a selective agonist for GPR30. Nat Chem Biol 2:207–212
Chan QK, Lam HM, Ng CF, Lee AY, Chan ES, Ng HK, Ho SM, Lau
KM (2010) Activation of GPR30 inhibits the growth of prostate
cancer cells through sustained activation of Erk1/2, c-jun/c-fos-
dependent upregulation of p21, and induction of G(2) cell-cycle
arrest. Cell Death Differ 17:1511–1523
Chimento A, Casaburi I, Bartucci M, Patrizii M, Dattilo R, Avena P,
Ando S, Pezzi V, Sirianni R (2013) Selective GPER activation
Fig. 6 γ-Radiation influences
the expression of GPER mRNA.
a qRT-PCR determination of
the expression levels of GPER
mRNA in MDA-MB-231,
MDA-MB-468, MCF-7,
and MCF-7 transfected with
p53-specific siRNA (MCF-
7-p53 neg) cells after different
doses of γ-radiation. b p53
expression in various breast
cancer cells after different
doses of γ-radiation monitored
by Western blot. β-Actin was
used as a loading control. c p53
expression in MDA-MB-231,
MDA-MB-468, and MCF-7
cells is altered upon exposure to
1 μM G-1 as compared to cells
incubated in medium only (con-
trol). The results are presented
as means of three independent
experiments ±SD. The numbers
above the blots represent the
relative expression units com-
pared to the control. The control
was set as 1
C
A
0
2.5
Relative expression of
GPER mRNA
5
p= 0.032
p< 0.0001
p= 0.003
p= 0.005
p= 0.047
p= 0.034
p= 0.003
MCF-7-p53 negMDA-MB-468MDA-MB-231
02510 02510 02510Gy
p= 0.0023
p= 0.0097
MCF-7
02510
B
0 Gy 2 Gy 5 Gy 10 Gy
MDA-MB-468
MCF-7
MDA-MB-231
1.00 0.89 1.83 2.91
1.00 1.79 3.17 3.05
1.00 2.55 4.60 4.46
0h C G-1 C G-1
24h48h
p53
β -actin
p53
β -actin
p53
β-actin
1.00 0.79 0.60 0.88 3.14
1.00 1.18 2.70 0.65 3.65
1.00 1.09 1.24 1.73 3.58
J Cancer Res Clin Oncol
1 3
decreases proliferation and activates apoptosis in tumor Leydig
cells. Cell Death Dis 4:e747
Choi HJ, Fukui M, Zhu BT (2011) Role of cyclin B1/Cdc2 up-regula-
tion in the development of mitotic prometaphase arrest in human
breast cancer cells treated with nocodazole. PLoS One 6:e24312
Dennis MK, Burai R, Ramesh C, Petrie WK, Alcon SN, Nayak TK,
Bologa CG, Leitao A, Brailoiu E, Deliu E, Dun NJ, Sklar LA,
Hathaway HJ, Arterburn JB, Oprea TI, Prossnitz ER (2009) In
vivo effects of a GPR30 antagonist. Nat Chem Biol 5:421–427
Dorsam RT, Gutkind JS (2007) G-protein-coupled receptors and can-
cer. Nat Rev Cancer 7:79–94
Esteller M, Corn PG, Baylin SB, Herman JG (2001) A gene hyper-
methylation profile of human cancer. Cancer Res 61:3225–3229
Fan P, Yue W, Wang JP, Aiyar S, Li Y, Kim TH, Santen RJ (2009) Mecha-
nisms of resistance to structurally diverse antiestrogens differ under
premenopausal and postmenopausal conditions: evidence from in
vitro breast cancer cell models. Endocrinology 150:2036–2045
Fei P, El Deiry WS (2003) P53 and radiation responses. Oncogene
22:5774–5783
Filardo EJ, Quinn JA, Bland KI, Frackelton AR Jr (2000) Estrogen-
induced activation of Erk-1 and Erk-2 requires the G protein-cou-
pled receptor homolog, GPR30, and occurs via trans-activation of
the epidermal growth factor receptor through release of HB-EGF.
Mol Endocrinol 14:1649–1660
Filardo EJ, Quinn JA, Frackelton AR Jr, Bland KI (2002) Estrogen
action via the G protein-coupled receptor, GPR30: stimulation of
adenylyl cyclase and cAMP-mediated attenuation of the epider-
mal growth factor receptor-to-MAPK signaling axis. Mol Endo-
crinol 16:70–84
Filardo EJ, Graeber CT, Quinn JA, Resnick MB, Giri D, DeLellis RA,
Steinhoff MM, Sabo E (2006) Distribution of GPR30, a seven
membrane-spanning estrogen receptor, in primary breast cancer
and its association with clinicopathologic determinants of tumor
progression. Clin Cancer Res 12:6359–6366
Fujiwara S, Terai Y, Kawaguchi H, Takai M, Yoo S, Tanaka Y, Tanaka
T, Tsunetoh S, Sasaki H, Kanemura M, Tanabe A, Yamashita Y,
Ohmichi M (2012) GPR30 regulates the EGFR-Akt cascade and
predicts lower survival in patients with ovarian cancer. J Ovarian
Res 5:35
Gao F, Ma X, Ostmann AB, Das SK (2011) GPR30 activation
opposes estrogen-dependent uterine growth via inhibition of stro-
mal ERK1/2 and estrogen receptor alpha (ERalpha) phosphoryla-
tion signals. Endocrinology 152:1434–1447
Girgert R, Emons G, Grundker C (2012) Inactivation of GPR30
reduces growth of triple-negative breast cancer cells: possi-
ble application in targeted therapy. Breast Cancer Res Treat
134:199–205
Hans F, Dimitrov S (2001) Histone H3 phosphorylation and cell divi-
sion. Oncogene 20:3021–3027
Holm A, Baldetorp B, Olde B, Leeb-Lundberg LM, Nilsson BO
(2011) The GPER1 agonist G-1 attenuates endothelial cell pro-
liferation by inhibiting DNA synthesis and accumulating cells in
the S and G2 phases of the cell cycle. J Vasc Res 48:327–335
Ignatov A, Lintzel J, Kreienkamp HJ, Schaller HC (2003) Sphingo-
sine-1-phosphate is a high-affinity ligand for the G protein-cou-
pled receptor GPR6 from mouse and induces intracellular Ca2+
release by activating the sphingosine-kinase pathway. Biochem
Biophys Res Commun 311:329–336
Ignatov A, Bischoff J, Schwarzenau C, Krebs T, Kuester D, Herrmann
K, Costa SD, Roessner A, Semczuk A, Schneider-Stock R (2008)
P16 alterations increase the metastatic potential of endometrial
carcinoma. Gynecol Oncol 111:365–371
Ignatov A, Bischoff J, Ignatov T, Schwarzenau C, Krebs T, Kuester D,
Costa SD, Roessner A, Semczuk A, Schneider-Stock R (2010a)
APC promoter hypermethylation is an early event in endometrial
tumorigenesis. Cancer Sci 101(2):321–327
Ignatov T, Eggemann H, Semczuk A, Smith B, Bischoff J, Roessner
A, Costa SD, Kalinski T, Ignatov A (2010b) Role of GPR30 in
endometrial pathology after tamoxifen for breast cancer. Am J
Obstet Gynecol 203:595-16
Ignatov A, Ignatov T, Roessner A, Costa SD, Kalinski T (2010c) Role
of GPR30 in the mechanisms of tamoxifen resistance in breast
cancer MCF-7 cells. Breast Cancer Res Treat 123:87–96
Ignatov A, Ignatov T, Weissenborn C, Eggemann H, Bischoff J, Sem-
czuk A, Roessner A, Costa SD, Kalinski T (2011) G-protein-cou-
pled estrogen receptor GPR30 and tamoxifen resistance in breast
cancer. Breast Cancer Res Treat 128:457–466
Ignatov T, Modl S, Thulig M, Weissenborn C, Treeck O, Ortmann O,
Zenclussen A, Costa SD, Kalinski T, Ignatov A (2013a) GPER-1
acts as a tumor suppressor in ovarian cancer. J Ovarian Res 6:51
Ignatov T, Weissenborn C, Poehlmann A, Lemke A, Semczuk A,
Roessner A, Costa SD, Kalinski T, Ignatov A (2013b) GPER-1
expression decreases during breast cancer tumorigenesis. Cancer
Invest 31:309–315
Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW
(1991) Participation of p53 protein in the cellular response to
DNA damage. Cancer Res 51:6304–6311
Kuo WH, Chang LY, Liu DL, Hwa HL, Lin JJ, Lee PH, Chen CN,
Lien HC, Yuan RH, Shun CT, Chang KJ, Hsieh FJ (2007) The
interactions between GPR30 and the major biomarkers in infil-
trating ductal carcinoma of the breast in an Asian population. Tai-
wan J Obstet Gynecol 46:135–145
Leblanc K, Sexton E, Parent S, Belanger G, Dery MC, Boucher V,
Asselin E (2007) Effects of 4-hydroxytamoxifen, raloxifene
and ICI 182 780 on survival of uterine cancer cell lines in the
presence and absence of exogenous estrogens. Int J Oncol
30:477–487
Li LC, Dahiya R (2002) MethPrimer: designing primers for methyla-
tion PCRs. Bioinformatics 18:1427–1431
Lim LY, Vidnovic N, Ellisen LW, Leong CO (2009) Mutant p53 medi-
ates survival of breast cancer cells. Br J Cancer 101:1606–1612
Liu Q, Li JG, Zheng XY, Jin F, Dong HT (2009) Expression of
CD133, PAX2, ESA, and GPR30 in invasive ductal breast carci-
nomas. Chin Med J (Engl) 122:2763–2769
Maggiolini M, Vivacqua A, Fasanella G, Recchia AG, Sisci D, Pezzi
V, Montanaro D, Musti AM, Picard D, Ando S (2004) The G
protein-coupled receptor GPR30 mediates c-fos up-regulation by
17beta-estradiol and phytoestrogens in breast cancer cells. J Biol
Chem 279:27008–27016
Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA,
Pollack JR, Ross DT, Johnsen H, Akslen LA, Fluge O, Perga-
menschikov A, Williams C, Zhu SX, Lonning PE, Borresen-Dale
AL, Brown PO, Botstein D (2000) Molecular portraits of human
breast tumours. Nature 406:747–752
Prossnitz ER, Arterburn JB, Smith HO, Oprea TI, Sklar LA, Hatha-
way HJ (2008) Estrogen signaling through the transmembrane G
protein-coupled receptor GPR30. Annu Rev Physiol 70:165–190
Schneider BP, Winer EP, Foulkes WD, Garber J, Perou CM, Rich-
ardson A, Sledge GW, Carey LA (2008) Triple-negative
breast cancer: risk factors to potential targets. Clin Cancer Res
14:8010–8018
Schumacher A, Brachwitz N, Sohr S, Engeland K, Langwisch S,
Dolaptchieva M, Alexander T, Taran A, Malfertheiner SF, Costa
SD, Zimmermann G, Nitschke C, Volk HD, Alexander H, Gunzer
M, Zenclussen AC (2009) Human chorionic gonadotropin attracts
regulatory T cells into the fetal-maternal interface during early
human pregnancy. J Immunol 182:5488–5497
Smith HO, Leslie KK, Singh M, Qualls CR, Revankar CM, Joste NE,
Prossnitz ER (2007) GPR30: a novel indicator of poor survival
for endometrial carcinoma. Am J Obstet Gynecol 196:386–389
Smith HO, Arias-Pulido H, Kuo DY, Howard T, Qualls CR, Lee SJ,
Verschraegen CF, Hathaway HJ, Joste NE, Prossnitz ER (2009)
J Cancer Res Clin Oncol
1 3
GPR30 predicts poor survival for ovarian cancer. Gynecol Oncol
114:465–471
Sun W, Yang J (2010) Functional mechanisms for human tumor sup-
pressors. J Cancer 1:136–140
Tam SW, Shay JW, Pagano M (1994) Differential expression and
cell cycle regulation of the cyclin-dependent kinase 4 inhibitor
p16Ink4. Cancer Res 54:5816–5820
Toh WH, Nam SY, Sabapathy K (2010) An essential role for p73 in
regulating mitotic cell death. Cell Death Differ 17:787–800
Tu G, Hu D, Yang G, Yu T (2009) The correlation between GPR30
and clinicopathologic variables in breast carcinomas. Technol
Cancer Res Treat 8:231–234
Vivacqua A, Bonofiglio D, Recchia AG, Musti AM, Picard D, Ando S,
Maggiolini M (2006) The G protein-coupled receptor GPR30 medi-
ates the proliferative effects induced by 17beta-estradiol and hydrox-
ytamoxifen in endometrial cancer cells. Mol Endocrinol 20:631–646
Wang C, Lv X, Jiang C, Davis JS (2012) The putative G-protein cou-
pled estrogen receptor agonist G-1 suppresses proliferation of
ovarian and breast cancer cells in a GPER-independent manner.
Am J Transl Res 4:390–402
Wang C, Lv X, He C, Hua G, Tsai MY, Davis JS (2013) The G-pro-
tein-coupled estrogen receptor agonist G-1 suppresses prolifera-
tion of ovarian cancer cells by blocking tubulin polymerization.
Cell Death Dis 4:e869
... Recent research has shown a significant correlation between GPER abundance and high-risk TNBC, characterized by G3 tumors, distant metastasis, stage III, and lymph node metastasis . Differing opinions suggest that GPER expression may suppress tumor initiation and progression (Weißenborn et al., 2014). GPER inhibits tumor cell growth by stimulating G2/M-phase cell cycle arrest, decreasing cyclin B expression, and promoting apoptosis in ERα-negative breast cancer cells (Wei et al., 2014). ...
... Previous studies have suggested that G-1 promotes tumor cell proliferation within the range of 10 nM-1 μM, whereas high concentrations inhibit proliferation. Weißenborn et al. (2014) found that GPER treated with 1 μM G-1 inhibited TNBC cell growth. This inhibition was concentrationdependent and achieved by inducing cell cycle arrest in the G2/M phase, thereby enhancing the phosphorylation of histone h3, and promoting caspase-3-mediated apoptosis (Weißenborn et al., 2014). ...
... Weißenborn et al. (2014) found that GPER treated with 1 μM G-1 inhibited TNBC cell growth. This inhibition was concentrationdependent and achieved by inducing cell cycle arrest in the G2/M phase, thereby enhancing the phosphorylation of histone h3, and promoting caspase-3-mediated apoptosis (Weißenborn et al., 2014). However, Lv et al. (2017) reported that the proliferation of TNBC cell lines was significantly inhibited by G-1 at relatively low concentrations (100 nM). ...
Article
Full-text available
Triple-negative breast cancer (TNBC) is a biologically and clinically heterogeneous disease. The G protein-coupled estrogen receptor (GPER) plays a crucial role in mediating the effect of estrogen and estrogen-like compounds in TNBC cells. Compared with other subtypes, GPER has a higher expression in TNBC. The GPER mechanisms have been thoroughly characterized and analyzed in estrogen receptor α (ERα) positive breast cancer, but not in TNBC. Our previous work revealed that a higher expression of GPER mRNA indicates a better prognosis for ERα-positive breast cancer; however, its effects in TNBC differ. Whether GPER could serve as a predictive prognostic marker or therapeutic target for TNBC remains unclear. In this review, we provide a detailed introduction to the subcellular localization of GPER, the different effects of various ligands, and the interactions between GPER and closely associated factors in TNBC. We focused on the internal molecular mechanisms specific to TNBC and thoroughly explored the role of GPER in promoting tumor development. We also discussed the interaction of GPER with specific cytokines and chemokines, and the relationship between GPER and immune evasion. Additionally, we discussed the feasibility of using GPER as a therapeutic target in the context of existing studies. This comprehensive review highlights the effects of GPER on TNBC, providing a framework and directions for future research.
... For example, in SKBr3 breast cancer cells that do not express the nuclear ER receptor, GPER has been shown to inhibit cell proliferation, an effect typically caused in cells expressing nuclear ERs by traditional ligands such as E2 [5]. Nevertheless, the activation of GPER demonstrates an antiproliferative effect-an outcome contrasting with the impact of ER-α activation [6,7]. The type of response largely depends on multiple factors, such as cell type, the profile of estrogen receptors present, and their expression levels. ...
... These effects have been reported both in cell lines that express ER-α (for example, MCF-7) and in those that do not express it (for example, SKBR3). However, some authors report that GPER activation induces a decrement in cell proliferation [6,7,50]. ...
Article
Full-text available
The high incidence of cancer and the prevalence of chemoresistance are serious problems worldwide, underscoring the urgency of novel research focused on understanding the underlying mechanisms and finding new therapeutic targets. Recently, the G protein-coupled estrogen receptor (GPER) has received increasing attention, and it has been studied in various models, including physiological and pathological conditions, using appropriate pharmacological and molecular biological strategies. Numerous studies indicate that GPER plays an important role in cancer progression and resistance. This review focuses on the structure of GPER, the diversity of its ligands and GPER-activated signaling pathways, the role of GPER in cancer progression, and mechanisms of chemoresistance, with special emphasis on different cancer types and the tumor microenvironment. GPER was evidenced to exhibit conformational plasticity and different ligand binding modes. Therefore , GPER-mediated effects can be triggered by estrogens or various estrogen mimetics, including synthesized compounds, licensed drugs, or exogenous environmental compounds. We found multiple reports evidencing that GPER is differentially expressed in healthy tissues and tumors and plays a protumor role in breast, ovarian, lung, thyroid, and endometrial cancers. Additionally, there are several studies that indicate that GPER expression in cells of the tumor microenvironment may also contribute to cancer progression. Among the major mechanisms of GPER-mediated chemoresistance are the epithelial-mesenchymal transition, the overexpression of multidrug resistance pumps, and autophagy regulation.
... We analyzed the expression of GPER in five UM cell lines and found that GPER was expressed in all cell lines at comparable levels by immunoblotting and densitometry analysis ( Figure 1A and B). GPER was described as a tumor suppressor gene in ovarian and breast cancer (22,23) and its activation was shown to regulate the expression of p53 (24) GPER is involved in estrogen-mediated melanin synthesis (14) and the pre-clinical agonist, G-1, was shown to induce both pigment production (8,9) and melanocyte differentiation (14). ...
... We found that GPER is expressed in UM cells and its downregulation stimulated cell growth. GPER knockdown was reported to increase growth of ovarian cancer cells (35), and there are studies demonstrating that GPER has tumor suppressor activities in breast cancer cells (23). Previous findings demonstrated that G-1 treatment induced p53 expression, and p53 knockdown partially reversed G-1-dependent anti-cancer effects in breast cancer cells (24), suggesting that p53 is a downstream mediator of GPER. ...
Article
Full-text available
Uveal melanoma is the most common primary intraocular malignancy in adults and has a high incidence of metastatic disease. Current treatments have shown limited clinical activity in patients with uveal melanoma with metastasis and there is an urgent need for new effective therapies. Recent findings have shown that women with uveal melanoma have better survival rates than men. The G protein–coupled estrogen receptor-1 (GPER) has distinct functions from those of the classic estrogen receptors ERα/β and its activation by specific agonists has tumor-suppressive roles in several cancers. However, the role of GPER had not previously been investigated in uveal melanoma. We demonstrated that downregulation of GPER in uveal melanoma cells decreased expression of p53 and stimulated cell growth. In contrast, the clinical GPER agonist, LNS8801, upregulated p53 and p21, induced melanocytic differentiation markers, inhibited cell proliferation and cell migration, and induced apoptosis. Furthermore, LNS8801 treatment arrested the cells in G2–M-phase of the cell cycle with concomitant activation of mitotic markers and disruption of the mitotic spindle apparatus. LNS8801 significantly inhibited tumor growth of uveal melanoma xenografts in vivo, suggesting that GPER agonists may be a novel treatment for uveal melanoma. Significance Current treatments against metastatic uveal melanoma have shown limited clinical activity and there is an urgent need for effective therapies. Here, we demonstrate that the GPER agonist LNS8801 induced both GPER-dependent and GPER-independent effects and elicited potent anticancer activities in vitro and in vivo. Our results complement and support the ongoing clinical trial of LNS8801 in advanced uveal melanoma.
... Besides, high expression of GPER is associated with pro-metastatic pathways and worse survival in patients with advanced TNBCs [6]. Nevertheless, other studies suggested a tumor-suppression action of GPER in TNBC, therefore its role in the context of the biological TNBC landscape remains to be fully explored [55][56][57][58]. This controversial scenario prompted us to generate GPER KO TNBC cells through the CRISPR-Cas9 gene editing system, which is widely used in order to contribute to unveil cancer genomics [59][60][61]. ...
Article
Full-text available
The G protein-coupled estrogen receptor (GPER) mediates estrogen action in different pathophysiological conditions, including cancer. GPER expression and signaling have been found to join in the progression of triple-negative breast cancer (TNBC), even though controversial data have been reported. In present study, we aimed at providing new mechanistic and biological discoveries knocking out (KO) GPER expression by CRISPR/Cas9 technology in MDA-MB-231 TNBC cells. GPER KO whole transcriptome respect to wild type (WT) MDA-MB-231 cells was determined through total RNA sequencing (RNA-Seq) and gene ontology (GO) enrichment analysis. We ascertained that anti-proliferative and pro-apoptotic gene signatures characterize GPER KO MDA-MB-231 cells. Thereafter, we determined that these cells exhibit a reduced proliferative, clonogenic and self-renewal potential along with an increased mitochondria-dependent apoptosis phenotype. In addition, we recognized that decreased cAMP levels trigger the JNK/c-Jun/p53/Noxa axis, which in turn orchestrates the pro-apoptotic effects observed in GPER KO cells. In accordance with these data, survival analyses in TNBC patients of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset indicated that high Noxa expression correlates with improved outcomes in TNBC patients. Furthermore, we demonstrated that GPER KO in TNBC cells impairs the expression and secretion of the well-acknowledged GPER target gene named CTGF, thus resulting in the inhibition of migratory effects in cancer-associated fibroblasts (CAFs). Overall, the present study provides novel mechanistic and biological insights on GPER KO in TNBC cells suggesting that GPER may be considered as a valuable target in comprehensive therapeutic approaches halting TNBC progression.
... On a different note, research by Sathya et al. [19] indicated that under low oxygen conditions, estrogen suppresses breast cancer growth via the GPER1/ROS/p38 MAPK/p21 signaling pathway. Weißenborn's findings [34,35] demonstrated that the GPER1-specific agonist G-1 activates GPER1 in a concentration-dependent manner, effectively inhibiting breast cancer cell growth. This suggests that cell surfaceexpressed GPER1 holds promise as a potential therapeutic target for non-triple-negative and triplenegative breast cancers. ...
Article
Full-text available
Background: The G protein-coupled oestrogen receptor (GPER) 1 mediates non-genomic oestrogen-related signalling and plays an important role in the regulation of cell growth and programmed cell death through multiple downstream pathways. Despite the increasing interest in the role of GPER1 in cancer development, no pan-cancer analysis has been available for GPER1. Methods: In this study we performed a comprehensive analysis of the role of GPER1 in pan-cancer via Human Protein Atlas (HPA), The Cancer Genome Atlas (TCGA), University of California, Santa Cruz Xena (UCSC XENA), Genotype-Tissue Expression (GTEx), MethSurv, The University of Alabama at Birmingham CANcer data analysis Portal (UALCAN), cBioPortal, STRING and TISIDB detabases, followed by enrichment analysis using R software. Results: GPER1 was widely expressed in tissues and organs and differed in expression from normal tissue in a variety of cancers. In diagnostic assessment, it's Area Under the Curve (AUC) surpassed 0.9 in nine cancer types. Survival analysis showed that GPER1 was correlated with the prognosis of 11 cancer types. Moreover, GPER1 expression was associated with immune infiltration in multiple cancers. Conclusions: In summary, GPER1 has good diagnostic or prognostic value across various malignancies. Together with its extensive correlation with immune components, the aforementioned results suggests that GPER1 shows promise in tumour diagnosis and prognosis, providing new ideas for precise and personalised anti-tumour strategies.
... As shown in Figure 2B, LTED cells exhibited more aggressive proliferative features than MCF-7 cells. The reduced expression of GPER1 favors cell aggressiveness, such as stimulated cell proliferation, particularly in LTED cells, as indicated in the reports suggesting that GPER1 can act as a tumor suppressor in different types of cancers, including breast cancer [33][34][35][36]. Therefore, the possible negative (antiproliferative) effects of GPER1 agonists (G-1 and 2-MeO-E2) [8,17] on the aggressive behavior of LTED cells were not observed or weakened. ...
Article
Full-text available
To identify effective treatment modalities for breast cancer with acquired resistance, we first compared the responsiveness of estrogen receptor-positive breast cancer MCF-7 cells and long-term estrogen-deprived (LTED) cells (a cell model of endocrine therapy-resistant breast cancer) derived from MCF-7 cells to G-1 and 2-methoxyestradiol (2-MeO-E2), which are microtubule-destabilizing agents and agonists of the G protein-coupled estrogen receptor 1 (GPER1). The expression of GPER1 in LTED cells was low (~0.44-fold), and LTED cells displayed approximately 1.5-fold faster proliferation than MCF-7 cells. Although G-1 induced comparable antiproliferative effects on both MCF-7 and LTED cells (IC50 values of >10 µM), 2-MeO-E2 exerted antiproliferative effects selective for LTED cells with an IC50 value of 0.93 μM (vs. 6.79 μM for MCF-7 cells) and induced G2/M cell cycle arrest. Moreover, we detected higher amounts of β-tubulin proteins in LTED cells than in MCF-7 cells. Among the β-tubulin (TUBB) isotype genes, the highest expression of TUBB2B (~3.2-fold) was detected in LTED cells compared to that in MCF-7 cells. Additionally, siTUBB2B restores 2-MeO-E2-mediated inhibition of LTED cell proliferation. Other microtubule-targeting agents, i.e., paclitaxel, nocodazole, and colchicine, were not selective for LTED cells. Therefore, 2-MeO-E2 can be an antiproliferative agent to suppress LTED cell proliferation.
... The observation that GPER protein is rarely expressed in human breast cancer cases is noteworthy, given the substantial number of reports discussing the role of GPER in preclinical models of breast cancer (16). Although GPER signaling was originally reported to be tumor promoting in some breast cancer models (17), subsequent reports show that GPER signaling inhibits breast cancer (18)(19)(20). Additionally, several studies have documented correlations between GPER expression and prognosis in breast cancer, with contradictory results (21,22). ...
Preprint
GPER (G protein-coupled estrogen receptor) has been reported to play roles in several areas of physiology including cancer, metabolic disorders, and cardiovascular disease. However, the understanding of where this receptor is expressed in human tissue is limited due to limited available tools and methodologies that can reliably detect GPER protein. Recently, a highly specific monoclonal antibody against GPER (20H15L21) was developed and is suitable for immunohistochemistry. Using this antibody, we show that GPER protein expression varies markedly between normal human tissue, and also among cancer tissue. As GPER is an emerging therapeutic target for cancer and other diseases, this new understanding of GPER distribution will likely be helpful in design and interpretation of ongoing and future GPER research.
... The effect of LI-NG-CDs on mice beard breast cancer is shown in Fig. 10A. Two cell lines of MCF-7 and SK-BR-3 were selected [55]. MCF-7 cell line shows a less complex chromosomal pattern than SK-BR-3 cells and also based on previous studies to compare the morphological changes of the cancerous tissues, MCF-7 cells with less chromosomal complexity are better in terms of their simplicity, to be used. ...
Article
Full-text available
Bio-responsive nanocomposites with facile fabrication and rational design are of great importance in the diagnosis and treatment of cancer. Herein, the combination of bioimaging due to the presence of QD and controlled drug delivery via nanogel was employed for cancer treatment. To this aim, we fabricated a traceable and bioresponsive fluorescent active nanogel composite by integrating creatinine-functionalized carbon dots (QD) into a lecithin-inulin nanogel. Subsequently, PEGylation and integration of herceptin on the nanogel were carried out to improve the biofate and its ability to target HER2-positive breast cancer. The assessment of cellular uptake demonstrated that this nanogel was effectively internalized by SK-BR-3 cells and the production of reactive oxygen species was signifcantly boosted. Moreover, the nanogel composite led to high expression of P53 and Bax genes along with a low expression of Bcl-2 gene (as promotors of the apoptosis signaling pathway). The cellular uptake of herceptin was enhanced. It led to inhibiting the proliferation of the breast cancer cells as well as the reduction of viability of SK-BR-3 cancer cells (HER-2 positive) compared with MDA-MB-231 cells (triple-negative). The intraperitoneal injection of the developed formulation to MCF-7 breast cancer cell-bearing BALB/c mice supported the observation of tumor growth inhibition. Overall, herceptin-adorned PEGylated lecithin-inulin nanogel composite can be a promising theranostic candidate for targeting HER-2-positive breast cancer.
Article
Full-text available
The G-protein-coupled estrogen receptor 1 (GPER) has recently been reported to mediate the non-genomic action of estrogen in different types of cells and tissues. G-1 (1-[4-(6-bromobenzo[1,3] dioxol-5yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone) was developed as a potent and selective agonist for GPER. G-1 has been shown to induce the expression of genes and activate pathways that facilitate cancer cell proliferation by activating GPER. Here we demonstrate that G-1 has an anticancer potential with a mechanism similar to vinca alkaloids, the commonly used chemotherapy drugs. We found that G-1 blocks tubulin polymerization and thereby interrupts microtubule assembly in ovarian cancer cells leading to the arrest of cell cycle in the prophase of mitosis and the suppression of ovarian cancer cell proliferation. G-1 treatment also induces apoptosis of ovarian cancer cells. The ability of G-1 to target microtubules to suppress ovarian cancer cell proliferation makes it a promising candidate drug for treatment of ovarian cancer.
Article
Full-text available
We have previously shown that estrogens binding to estrogen receptor (ER) α increase proliferation of Leydig tumor cells. Estrogens can also bind to G protein-coupled ER (GPER) and activation of this receptor can either increase or decrease cell proliferation of several tumor types. The aim of this study was to investigate GPER expression in R2C rat tumor Leydig cells, evaluate effects of its activation on Leydig tumor cell proliferation and define the molecular mechanisms triggered in response to its activation. R2C cells express GPER and its activation, using the specific ligand G-1, is associated with decreased cell proliferation and initiation of apoptosis. Apoptosis after G-1 treatment was asserted by appearance of DNA condensation and fragmentation, decrease in Bcl-2 and increase in Bax expression, cytochrome c release, caspase and poly (ADP-ribose) polymerase-1 (PARP-1) activation. These effects were dependent on GPER activation because after silencing of the gene, using a specific small interfering RNA, cyt c release, PARP-1 activation and decrease in cell proliferation were abrogated. These events required a rapid, however, sustained extracellular regulated kinase 1/2 activation. G-1 was able to decrease the growth of R2C xenograft tumors in CD1 nude mice while increasing the number of apoptotic cells. In addition, in vivo administration of G-1 to male CD1 mice did not cause any alteration in testicular morphology, while cisplatin, the cytotoxic drug currently used for the therapy of Leydig tumors, severely damaged testicular structure, an event associated with infertility in cisplatin-treated patients. These observations indicate that GPER targeting for the therapy of Leydig cell tumor may represent a good alternative to cisplatin to preserve fertility in Leydig tumor patients.
Article
Full-text available
It is known that the new membrane-bound estrogen receptor GPER-1 acts suppressive in breast cancer cells and its expression decreases during disease progression. This study was conducted to evaluate the GPER-1 expression in ovarian cancer and its correlation with progression. Its function was tested in vitro in ovarian cancer cells.Patients and methods: GPER-1 expression was analyzed by immunohistochemistry in 35 benign ovarian tumors, 35 tumors of low-malignant potential and in 124 ovarian cancers. GPER-1 expression was correlated to the prospectively evaluated disease-free survival of ovarian cancer patients. We also tested GPER-1 expression in ovarian cancer cells and the effect of GPER-1 stimulation on cell growth. GPER-1 expression was significantly lower in ovarian cancer tissue than in benign and low-malignant ovarian tumors. GPER-1 expression was observed in 83.1% of malignant tumors and was higher in early stage cancers and tumors with high histological differentiation. GPER-1 expression was associated with favourable clinical outcome. The difference in 2-year disease-free survival by GPER-1 expression was significant, 28.6% for GPER-1 negative and 59.2% for GPER-1 positive cases (p = 0.002). GPER-1 expression was observed in SKOV-3 and OVCAR-3 ovarian cancer cell lines. G-1, a selective GPER-1 agonist, suppressed proliferation of the two cell types via inhibition of cell cycle progression in G2/M phase and stimulation of caspase-dependent apoptosis. The blockade in G2/M phase was associated with increased expression of cyclin B1 and Cdc2 and phosphorylation of histone 3. GPER-1 emerges as a new tumor suppressor with unsuspected therapeutic potential for ovarian cancer.
Article
Full-text available
We are in an era where the potential exists for deriving comprehensive profiles of DNA alterations characterizing each form of human cancer. Such profiles would provide invaluable insight into mechanisms underly- ing the evolution of each tumor type and will provide molecular markers, which could radically improve cancer detection. To date, no one type of DNA change has been defined which accomplishes this purpose. Herein, by using a candidate gene approach, we show that one category of DNA alteration, aberrant methylation of gene promoter regions, can enor- mously contribute to the above goals. We have now analyzed a series of promoter hypermethylation changes in 12 genes (p16INK4a, p15INK4b, p14ARF, p73, APC,5 BRCA1, hMLH1, GSTP1, MGMT, CDH1, TIMP3, and DAPK), each rigorously characterized for association with abnormal gene silencing in cancer, in DNA from over 600 primary tumor samples rep- resenting 15 major tumor types. The genes play known important roles in processes encompassing tumor suppression, cell cycle regulation, apopto- sis, DNA repair, and metastastic potential. A unique profile of promoter hypermethylation exists for each human cancer in which some gene changes are shared and others are cancer-type specific. The hypermethy- lation of the genes occurs independently to the extent that a panel of three to four markers defines an abnormality in 70 -90% of each cancer type. Our results provide an unusual view of the pervasiveness of DNA alter- ations, in this case an epigenetic change, in human cancer and a powerful set of markers to outline the disruption of critical pathways in tumori- genesis and for derivation of sensitive molecular detection strategies for virtually every human tumor type.
Article
Full-text available
GPER-1 protein expression was immunohistochemically examined in 164 primary breast cancer specimens and their matched normal breast epithelium. GPER-1 down-regulation correlated significantly with increased histological grading (p = .015), lymph node metastases (p = .032), and negative estrogen receptor status (p = .018). The decrease of GPER-1 expression in breast cancer tissue, relative to normal tissue, was associated with poor overall survival (p = .043) and disease-free survival (p = .037) and remained a significant unfavorable factor in multivariate analysis for DFS (HR = 1.569; 95% CI, 1.024-2.797; p = .041) and OS (HR = 2.082; 95% CI, 1.248-4.773; p = .039). Thus aberrant GPER-1 expression seems to be an important factor in breast cancer progression.
Article
Full-text available
Objectives G protein-coupled receptor 30 (GPR30) is a 7-transmembrane estrogen receptor that functions alongside traditional estrogen receptors to regulate the cellular responses to estrogen. Recent studies suggest that GPR30 expression is associated with a poor prognosis, and that this is due to the GPR30-mediated transactivation of the EGFR in breast cancer. However, the biological contribution of GPR30 in ovarian cancer remains unclear. The purpose of this study was to elucidate the relationships between GPR30 expression and the clinicopathological findings, and to determine how the signaling cascade influences the prognosis of ovarian cancer. Methods The expression levels of GPR30, EGFR, ERα, and ERβ were analyzed using an immunohistochemical analysis, and their correlations with the clinicopathological features were examined in 10 patients with borderline malignant tumors and 152 patients with epithelial ovarian cancer. We also examined whether GPR30 signaling activates the EGFR-Akt pathway in an ovarian cancer cell line (Caov-3) by a Western blotting analysis. Results The GPR30 expression in ovarian carcinomas was significantly higher than that in borderline malignancies (p=0.0016), and was not associated with the expression of the EGFR, ERα, or ERβ. The expression of GPR30 in clear cell carcinomas was significantly lower than that in other subtypes of cancer (P <; 0.001). The expression of both GPR30 and EGFR was significantly associated with a poor prognosis in terms of the progression-free survival rate. The phosphorylation of the EGFR and Akt could be significantly enhanced by G1 (p <; 0.05) and inhibited by a Src family kinase inhibitor. Conclusion The expression of both GPR30 and EGFR is associated with a poor outcome in ovarian cancer, and GPR30 increases the phosphorylation of Akt via the EGFR in ovarian cancer cells. The regulation of GPR30 might be a potentially useful new therapeutic target in ovarian cancer.
Article
Full-text available
G-protein coupled estrogen receptor 1 (GPER) plays an important role in mediating estrogen action in many different tissues under both physiological and pathological conditions. G-1 (1-[4-(6-bromobenzo[1,3]dioxol-5yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta [c]quinolin-8-yl]-ethanone) has been developed as a selective GPER agonist to distinguish estrogen actions mediated by GPER from those mediated by classic estrogen receptors. In the present study, we surprisingly found that G-1 suppressed proliferation and induced apoptosis of KGN cells (a human ovarian granulosa cell tumor cell line), actions that were not blocked by a selective GPER antagonist G15 or siRNA knockdown of GPER. G-1 also suppressed proliferation and induced cell apoptosis in GPER-negative HEK-293 cells and MDA-MB 231 breast cancer cells. Our results demonstrate that G-1 suppresses proliferation of ovarian and breast cancer cells in a GPER-independent manner. G-1 may be a candidate for the development of drugs against ovarian and breast cancer.
Article
Full-text available
Triple-negative breast cancers lack estrogen receptor α (ERα), progesterone receptor, and do not overexpress human epidermal growth factor receptor 2 (Her-2). They are neither susceptible to endocrine therapy nor to a therapy using the anti-Her-2 antibody, trastuzumab. Therefore, an efficient targeted therapy is warranted. Triple-negative breast tumors frequently express membrane bound estrogen receptor G-protein coupled receptor (GPR30). As proof of principle, we analyzed the consequences of a knock-down of GPR30 expression on the growth regulation of triple-negative breast cancer cell lines. Cells of triple-negative breast cancer cell lines were transfected with siRNA against GPR30 or control siRNA, and cell growth was stimulated either with 10(⁻⁹) M 17β-estradiol or 10(⁻⁶) M 4-hydroxytamoxifen. Cell proliferation was measured using Alamar blue staining. Activation of c-Src and epidermal growth factor (EGF)-receptor was assessed using western blot. Expression of c-fos was quantified by reverse transcription polymerase chain reaction. Seven days after transfection with siRNA, GPR30 mRNA in triple-negative breast cancer cell lines MDA-MB-435 and HCC1806 was reduced by 74 and 90%, respectively. 10(⁻⁸) M 17β-estradiol enhanced proliferation of MDA-MB-435 to 129.6±5.4% of control (p<0.05) and HCC1806 to 156.9±15.4% of control (p<0.05), respectively. 10(⁻⁶) M 4-hydroxytamoxifen increased cell number of MDA-MB-435 to 121.0±6.9% of control (p<0.05) and HCC1806 to 124.5±12.1% of control (n.s.), respectively. This increased proliferation by the two estrogenic compounds was completely prevented by knock-down of GPR30 expression in both cell lines. In control cells, activity of Src kinase was increased 3-fold by estradiol and 3.8-fold using 4-hydroxytamoxifen. Transactivation of the EGF-receptor was similarly increased in both cell lines by 17β-estradiol and 4-hydroxytamoxifen. Both compounds increased c-fos expression 1.5- and 3.1-fold, respectively. Knock-down of GPR30 expression completely abolished activation of all these signaling pathways responsible for enhanced proliferation. A pharmacological inhibition of GPR30 by specific small molecular inhibitors might prove to be an appropriate targeted therapy of triple-negative breast cancer in the future.