ArticlePDF Available

Baicalein suppresses 17-β-estradiol-induced migration, adhesion and invasion of breast cancer cells via the G protein-coupled receptor 30 signaling pathway

Authors:

Abstract and Figures

Flavonoids are structurally similar to steroid hormones, particularly estrogens, and therefore have been studied for their potential effects on hormone-dependent cancers. Baicalein is the primary flavonoid derived from the root of Scutellaria baicalensis Georgi. In the present study, we investigated the effects of baicalein on 17β-estradiol (E2)-induced migration, adhesion and invasion of MCF-7 and SK-BR-3 breast cancer cells. The results demonstrated that baicalein suppressed E2-stimulated wound-healing migration and cell‑Matrigel adhesion, and ameliorated E2-promoted invasion across a Matrigel-coated Transwell membrane. Furthermore, baicalein interfered with E2-induced novel G protein-coupled estrogen receptor (GPR30)-related signaling, including a decrease in tyrosine phosphorylation of epidermal growth factor receptor (EGFR) as well as phosphorylation of extracellular signal-regulated kinase (ERK) and serine/threonine kinase Akt, without affecting GPR30 expression. The results also showed that baicalein suppressed the expression of GPR30 target genes, cysteine-rich 61 (CYR61) and connective tissue growth factor (CTGF) induced by E2. Furthermore, baicalein prevented GPR30-related signaling activation and upregulation of CYR61 and CTGF mRNA levels induced by G1, a specific GPR 30 agonist. The results suggest that baicalein inhibits E2-induced migration, adhesion and invasion through interfering with GPR30 signaling pathway activation, which indicates that it may act as a therapeutic candidate for the treatment of GPR30-positive breast cancer metastasis.
Content may be subject to copyright.
ONCOLOGY REPORTS
Abstract. Flavonoids are structurally similar to steroid
hormones, particularly estrogens, and therefore have been
studied for their potential effects on hormone-dependent
cancers. Baicalein is the primary avonoid derived from the
root of Scutellaria baicalensis Georgi. In the present study,
we investigated the effects of baicalein on 17β-estradiol
(E2)-induced migration, adhesion and invasion of MCF-7
and SK-BR-3 breast cancer cells. The results demonstrated
that baicalein suppressed E2-stimulated wound-healing
migration and cell-Matrigel adhesion, and ameliorated
E2-promoted invasion across a Matrigel-coated Transwell
membrane. Furthermore, baicalein interfered with E2-induced
novel G protein-coupled estrogen receptor (GPR30)-related
signaling, including a decrease in tyrosine phosphorylation
of epidermal growth factor receptor (EGFR) as well as phos-
phorylation of extracellular signal-regulated kinase (ERK) and
serine/threonine kinase Akt, without affecting GPR30 expres-
sion. The results also showed that baicalein suppressed the
expression of GPR30 target genes, cysteine-rich 61 (CYR61)
and connective tissue growth factor (CTGF) induced by E2.
Furthermore, baicalein prevented GPR30-related signaling
activation and upregulation of CYR61 and CTGF mRNA levels
induced by G1, a specic GPR 30 agonist. The results suggest
that baicalein inhibits E2-induced migration, adhesion and
invasion through interfering with GPR30 signaling pathway
activation, which indicates that it may act as a therapeutic
candidate for the treatment of GPR30-positive breast cancer
metastasis.
Introduction
Breast cancer is the most common malignancy affecting
women and is the leading cause of cancer-related mortality
among females worldwide (1). Despite advances in the diag-
nosis and treatment of breast cancer, approximately a third of
patients develop metastatic disease, with a median survival
time of less than 3 years (2). Preventing metastasis is impor-
tant for effectively overcoming breast cancer. Estrogen plays
an important role, not only in the initiation and proliferation
of breast cancer, but also in cancer metastasis (3). Most of the
biological effects of estrogen are mediated by binding and
activating classic estrogen receptors (ERs), ERα and ERβ,
to stimulate the transcription of target genes via genomic
and non-genomic mechanisms (4). Based on this knowledge,
hormonal therapies that interfere with ERα function a re
currently applied in patients with ERα-positive breast cancer,
and beneficial effects have been achieved. Although there
is general success with ERα expression and responsiveness
to ERα-targeted agents, some patients do not respond to
endocrine therapy, and most patients appear to have acquired
resistance (5). In addition, some types of ERα-negative breast
cancer cells remain estrogen responsive, indicating the exis-
tence of an alternative receptor for estrogen.
G protein-coupled estrogen receptor (GPR30), as a member
of the 7-transmembrane GPR family, mediates both rapid
signaling and transcriptional events in response to estrogen.
It mediates estrogenic-rapid non-genomic actions, through
the Gβγ subunit downstream, release of heparin-bound EGF
(HB-EGF) and metalloproteinase (MMP)-dependent transac-
tivation of epidermal growth factor receptors (EGFRs), as well
as activation of mitogen-activated protein kinases (MAPKs),
adenyl cyclase, and phosphoinositide 3-kinase (PI3K) (6).
Subsequently, it also triggers transcriptional responses by acti-
vating various transcription factors, such as FOS, JUN, EGR1
Baicalein suppresses 17-β-estradiol-induced migration, adhesion
and invasion of breast cancer cells via the G protein-coupled
receptor 30 signaling pathway
DANDAN SHANG1, ZHENG LI1, ZHUXIA ZHU1, HUAMEI CHEN1, LUJUN ZHAO2,3,
XUDONG WANG1 and YAN CHEN1-3
1Department of Physiology/Cancer Research Group, Guiyang Medical University School of Basic Medicine;
2Department of Pharmacology of Chinese Material Medica and 3Key Laboratory of Optimal Utilization
of Natural Medicine Resources, Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
Received October 4, 2014; Accepted January 26, 2015
DOI: 10.3892/or.2015.3786
Correspondence to: Dr Yan Chen, Department of Pharmacology
of Chinese Material Medica, Guiyang Medical University, 9 Beijing
Road, Guiyang, Guizhou 550004, P.R. China
E-mail: s0710189@sina.com
Professor Xudong Wang, Department of Physiology, Guiyang Medical
University, 9 Beijing Road, Guiyang, Guizhou 550004, P.R. China
E-ma il: xdwang@gmc.edu.cn
Key word s: baicalein, estrogen, G protein-coupled receptor 30,
migration, adhesion, invasion, breast cancer
SHANG et al: BAICALEIN INHI BITS ESTROGEN-INDUCED MIGRATION, ADHESION AN D INVASION
2
and ATF3 following the protein kinase signaling cascade (7).
GPR30 expression is positively associated with the features
of breast tumor progression, including tumor size, HER2/neu
status, and metastases. Yet, ERα is inversely associated with
HER2/neu and tumor size (8). ERα in breast cancer generally
indicates good prognosis and treatment responsiveness with
anti-estrogen agents (9); yet, GPR30 is overexpressed in inva-
sive breast cancer and is positively associated with a metastatic
phenotype (10). GPR30 is implicated in breast cancer metas-
tasis and it may provide a new potential target for endocrine
th e r a py.
The incidence of breast cancer is much higher in Western
countries than in Asian ones. Epidemiological studies
contribute these differences to dietary factors, particularly
soy consumption. Flavonoids are considered to have a
signicant potential in breast cancer chemoprevention (11).
Their polyphenolic ring is structurally similar to the steroid
nucleus of 17β-estradiol (Fig. 1A) and may have estrogenic
or anti-estrogenic activity (12). Baicalein is the primary
avonoid derived from the root of Scutellaria baicalensis
Georgi and is also found in soybeans and fruit, which bears
the three-ring structure of the avone backbone with phenolic
hydroxyl at the 5', 6', and 7' position (Fig. 1A). It possesses
several health benets including anti-inammatory, antioxi-
dant, antiviral, neuroprotective, and anticancer effects (13).
It has also been shown to have antitumor effects in various
hormone-dependent cancers such as breast, colon, and
prostate cancer. Previous studies have found that baicalein
suppresses 17β-estradiol (E2)-induced ER transactivation in
MCF-7 cells (14) and displaces >85% of estradiol binding in
mouse uterine cytosol (15). Additionally, baicalein exhibits
effective inhibitory activity against E2/insulin-like growth
factor 1 (IGF-1)-induced proliferative events in breast cancer
cells (16). These investigations propose that baicalein may
exert anti-estrogenic activity.
The present study investigated the inhibitory activity of
baicalein on E2-induced migration, adhesion, and invasion
of MCF-7 and SK-BR-3 breast cancer cells. Furthermore, we
investigated its effect on GPR30-related signaling, including
phosphorylation of EGFR as well as extracellular signal-regu-
lated kinase (ERK), serine/threonine kinase Akt activation,
and GPR30-mediated gene expression.
Materials and methods
Reagents and antibodies. Baicalein (purity >98%) was
provided by Professor Zhiyu Li (China Pharmaceutical
University, Jiangsu, China). It was dissolved in dimethyl
sulfoxide (DMSO) as a stock solution at 0.1 M and stored at
-20˚C. E2 and G1 from Sigma-Aldrich (St. Louis, MO, USA)
were dissolved in DMSO as a stock solution at 10-1 M and
stored at 4˚C. Matrigel was obtained from BD Biosciences
(Bedford, MA, USA). Bovine serum albumin (BSA) was
purchased from Beijing Solarbio Science and Technology
Co., Ltd. (Beijing, China). 3-(4,5-Dimethylthiazol-2-
yl)-2,5-diphenyltetrazolium bromide (MTT) and cytosine
β-D-arabinofuranoside hydrochloride were purchased from
Sigma-Aldrich. The GPR30 (N-15)-R antibody (sc-48525-R,
polyclonal antibody, rabbit anti-human, 1:800) was purchased
from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA).
Primary antibodies against p-ERK (T202/Y204) (AP0484,
polyclonal antibody, rabbit anti-human, 1:1,000), extracel-
lular regulated protein kinase 1/2 (ERK1/2) (L352) (BS1112,
polyclonal antibody, rabbit anti-human, 1:1,000), and GAPDH
(1A6) (MB001, monoclonal antibody, mouse anti-human,
1:1,000) were purchased from Bio-World (Dublin, OH, USA).
Primary antibodies for p-Akt (Ser473) (193H12) (#4058,
monoclonal antibody, rabbit anti-human, 1:1,000), Akt (11E7)
(#4685, monoclonal antibody, rabbit anti-human, 1:1,000),
p-EGFR (Tyr1173) (53A5) (#4407, monoclonal antibody,
rabbit anti-human, 1:1,000) and EGFR (D38B1) (#4267, mono-
clonal antibody, rabbit anti-human, 1:1,000) were purchased
from Cell Signaling Technology (Beverly, MA, USA). The
anti-mouse or anti-rabbit IgG horseradish peroxidase-conju-
gated secondary antibodies (1:3,000) were purchased from
Santa Cruz Biotechnology, Inc.
Cell lines and culture. Human breast cancer cell lines MCF-7
and SK-BR-3 were obtained from the Kunming Cell Bank of
the Chinese Academy of Sciences (Yunnan, China). MCF-7
cells were maintained in Dulbecco's modied Eagle's medium
(DMEM) with 10% FBS, and SK-BR-3 cells were maintained
in RPMI-1640 (all from Hyclone, Logan, UT, USA) with 10%
FBS (Gibco-BRL, Carlsbad, CA, USA), both supplemented
with 100 U/ml penicillin and 100 µg/ml streptomycin (both
from Sigma-Aldrich). These cells were incubated at 37˚C in
humidied air with 5% CO2.
Cell treatment. Before the indicated treatments, cells were
cultured in phenol red (PR)-free medium without serum for
24 h. Cells were then treated with or without 10 nM of E2
(or 1 µM G1) and different concentrations of baicalein (5, 10,
and 15 µM) in PR-free medium without serum for a speci-
ed period of time. Control cells were incubated in DMSO
(0. 01%, v / v).
MTT assay. The 96-well microculture plates were individu-
ally seeded with 100 µl of the respective cell suspension and
100 µl of treatment medium. After incubation at 37˚C for
24 h, 20 µl of MTT (5 mg/ml) was added, and the plates were
incubated for an additional 4 h at 37˚C. The medium was then
removed, and 100 µl DMSO was added and mixed thoroughly.
Spectrometric absorbance values at 570 nm were measured
using a microplate reader (Bio-Rad, Lincoln, NE, USA). The
results are expressed as inhibitory rates with respect to the
controls, calculated using the following formula:
Inhibitory rate (%) = [(Acontrol - Atreated)/Acontrol] x100.
Wound-healing assay. Cells were seeded into 12-well plates
at 90% conuency. Monolayers were wounded with a micro-
pipette tip and rinsed with phosphate-buffered saline (PBS)
3 times to remove any oating cells. Then the cells were
incubated in the absence or presence of E2 (10 nM) with or
without various concentrations of baicalein (5, 10, and 15 µM)
for 24 h. Cytosine β-D-arabinofuranoside hydrochloride
(10 µM) was added 1 h before the test treatment to inhibit cell
proliferation (17). Images were captured at 0 and 24 h after
wound application. The level of cell migration into the wound
was quantied as the migration rate compared against the cells
at 0 h for each group.
ONCOLOGY REPORTS 3
Cell adhesion assay. The cell adhesion assay was performed
as described (18). Briefly, 96-well plates were coated with
Matrigel (50 µg/well) at 37˚C for 1 h and blocked with 0.5%
BSA (v/v) at 4˚C for 4 h. After treatment with E2 (10 nM) or
E2 (10 nM) plus baicalein (5, 10, and 15 µM) for 24 h, the cells
were harvested and suspended in serum-free medium. Cells
at a density of 2x105 cells/ml (100 µl) were added into the
plates and then incubated at 37˚C for 1 h. After incubating for
adhesion, the medium was carefully suctioned out. The wells
were then washed with PBS to remove non-adherent cells. The
colorimetric MTT assay was employed to analyze the number
of adherent cells. Spectrometric absorbance was measured at
570 nm by using a microplate reader (Bio-Rad). The results
are expressed as adhesive cells with respect to the control as
a percentage.
Invasion assay. Invasive ability of the cells was measured by
assay using a Transwell chamber (Millipore, Billerica, MA,
USA) containing membranes with an 8-µm pore size and,
coated with Matrigel as previously described (19). The cells
were exposed to E2 (10 nM) with or without different concen-
trations of baicalein (5, 10, and 15 µM) for 24 h. The cells
were then trypsinized and suspended at a nal concentration
of 5x105 cells/ml in serum-free medium. The cell suspension
was added into each 10-mm upper chamber, and medium with
10% FBS was added into the bottom chamber as a chemoat-
tractant. After incubating for 24 h, the upper surfaces of the
membranes were swabbed to remove non-invaded cells, and
the cells that attached to the lower surface were fixed in
100% methanol, stained with hematoxylin and eosin (H&E)
(Beyotime Institute of Biotechnology, Jiangsu, China), and
counted under a microscope. Five randomly chosen elds were
analyzed for each group.
Western blot analysis. Cells were collected after E2 (10 nM)
or G1 (1 µM) with or without baicalein (5, 10, and 15 µM)
treatment for 1 h, and lysed in lysis buffer (Beyotime Institute
of Biotechnology). The lysates were claried by centrifugation
at 4˚C for 15 min at 13,000 x g. The concentration of protein
in the supernatants was measured using bicinchoninic acid
(BCA) assay kit (Beyotime Institute of Biotechnology) with a
Varioskan Multimode microplate spectrophotometer (Thermo
Waltham, MA, USA). Total proteins (30 µg/lane) were sepa-
rated on a 12 or 8% SDS-PAGE gel, and transferred to a PVDF
membrane (Millipore, Bedford, MA, USA). The membrane
was blocked with 5% non-fat milk and subsequently incubated
with the indicated antibodies. Blots were visualized by using
an enhanced chemiluminescence kit (Millipore, Billerica,
MA, USA). Digital images of the blots were produced by a
Syngene Gel Imaging System and quantied with GeneSnap
(Syngene, Frederick, MD, USA) software.
Real-time reverse transcription-polymerase chain reaction
(RT-PC R). Total RNA was extracted after a 1-h treatment with
10 nM E2 or G1 (1 µM) in the presence or absence of baical-
cein (5, 10 and 15 µM) by using an RNA extraction kit (code
no. 9767; Takara, Dalian, China) according to the manufac-
turer's instructions. Complementary DNA (cDNA) was
synthesized using a Strand Complementary DNA Synthesis kit
(code no. 6210A; Takara). Real time PCR was performed on an
iCycle iQ (Bio-Rad) using 20 ng of cDNA template and 10 µM
primers. Cysteine-rich 61 (CYR61) primers (forward, 5'-ACT
Figure 1. Effects of baicalein on cell viability. (A) Chemical structure of 17β-estradiol (E2) and baicalein. (B) Inhibitory effects of baicalein on the cell viability
of MCF-7 and SK-BR-3 cells at 24 h as detected by MTT assay. Cells were treated with various concentrations of baicalein for 24 h. The result is represented
by the inhibitory rate. (C) Cells were incubated with the indicated doses of baicalein with or without E2 (10 nM) after 24 h, and cellular viability was assessed
in (a) MCF-7 and (b) SK-BR-3 cells. Values are mean ± SEM. #P<0.05 compared with control group, *P<0.05 compared with the E2-treated group.
SHANG et al: BAICALEIN INHI BITS ESTROGEN-INDUCED MIGRATION, ADHESION AN D INVASION
4
TCA TGG TCC CAG TGC TC-3' and reverse, 5'-AAA TCC
GGG TTT CTT TCA CA-3') (20), connective tissue growth
factor (CTGF) primers (forward, 5'-ACC TGT GGG ATG
GGC ATC T-3' and reverse, 5'-CAG GCG GCT CTG CTT
CTC TA-3') (21) and β-actin primers (forward, 5'-AGT TGC
GTT ACA CCC TTT C-3' and reverse, 5'-CCT TCA CCG
TTC CAG TTT-3') (22) were synthesized by Sangon Biotech
(Shanghai, China). The mRNA levels were measured using
SYBR Premix Ex Taq™ (code no. RR037A; Takara). The
reactions were performed for 30 sec at 95˚C, followed by
40 cycles at 95˚C for 5 sec and 60˚C for 30 sec. The melting
curve was analyzed at 55 to 95˚C to detect a single gene-
specic peak and to verify the absence of primer dimer peaks.
The CYR61 or CTGF mRNA level was quantied in relation to
β-actin mRNA level by using the 2-ΔΔCt method.
Statistical analysis. Statistically signicant differences were
calculated by one-way ANOVA followed by the Bonferroni
post-hoc test for multiple-group comparisons. A P<0.05 was
considered to indicate a signicant difference, and data are
expressed as mean ± standard error of the mean (SEM). The
data shown in this test were obtained from 3 independent
experiments.
Results
Cytotoxic effect of baicalein on MCF-7 and SK-BR-3 cells.
To investigate the effect of baicalein on cell viability, MCF-7
and SK-BR-3 cells were treated with various concentrations of
baicalein for 24 h, and viability was detected by MTT assay.
The IC50 values of baicalein in the MCF-7 and SK-BR-3 cells
after 24 h were 66.3±5.9 and 103.5±9.4 µM, respectively
(Fig. 1B). According to the results, baicalein at 5, 10 and 15 µM
had no obvious cytotoxic effect on the MCF-7 or SK-BR-3
cells in the presence or absence of E2 (Fig. 1C), and were used
in following study.
Figure 2. Effects of baicalein on the E2-induced migration of MCF-7 and SK-BR-3 breast cancer cells. (A) Conuent monolayers were scratched and incubated
in the absence or presence of E2 (10 nM) with or without various concentrations of baicalein (5, 10, and 15 µM) for 24 h. The images were captured at 0
and 24 h after wounding (magnication, x100). These gures are representative of 3 separate experiments. (B) The inhibition of E2-stimulated migration by
baicalein. Values are mea n ± SEM. *P<0.05 compared with the E2-treated group. E2, 17β-estradiol.
ONCOLOGY REPORTS 5
Baicalein inhibits E2-induced cell migration. After treatment
with E2, MCF-7 and SK-BR-3 cells were found to have signi-
cantly enhanced migration into the wound, while cells treated
with baicalein had less migratory ability (Fig. 2A). E2 stimula-
tion for 24 h signicantly increased the migration rate from
13±2 to 52±3% in the MCF-7 cells, and from 10±4 to 40±5%
in the SK-BR-3 cells, respectively. After treatment with 15 µM
baicalein for 24 h, the migration rate decreased to 17±2% in
the MCF-7 cells and 13±4% in the SK-BR-3 cells, respectively
(Fig. 2B). These results demonstrated that baicalein suppressed
E2-induced cell migration.
Baicalein suppresses E2-promoted cell adhesion to Matrigel.
Adhesion of tumor cells to extracellular matrix (ECM) is
an essential step for tumor invasion and metastasis. We next
examined the effect of baicalein on E2-promoted cell adhesion
Figure 4. Effect of baicalein on E2-stimulated cell invasion in MCF-7 and SK-BR-3 cells. (A) Cells were treated with or without E2 (10 nM) and di fferent
concentrations of baicalein (5, 10 and 15 µM) for 24 h. Invasive cells that passed through the membrane were evaluated using H&E staining (magnication,
x200). The images a re representative of 3 separate experiments. (B) The results are expressed as invasive cells with respect to the cont rol (as 100%). Values
are mean ± SEM. *P<0.05 indicates signicant difference from the E2-treated group. E2, 17β-estr adiol.
Figure 3. Effects of baicalein on E2-promoted cell adhesion to Matrigel in (A) MCF-7 and (B) SK-BR-3 cells. Cells were treated with or without E2 (10 nM)
or E2 (10 nM) plus baicalein (5, 10 and 15 µM) for 24 h. The results are expressed as adhesive cells with respect to the control (as 100%). *P<0.05 i ndicates
signicant difference from the E2-treated group. Values are mean ± SEM. E2, 17β-estr adiol.
SHANG et al: BAICALEIN INHI BITS ESTROGEN-INDUCED MIGRATION, ADHESION AN D INVASION
6
to Matrigel, a reconstituted basement membrane (an impor-
tant component of ECM). E2 greatly increased cell-Matrigel
adhesion by 195.0±27% in the MCF-7 and by 161±8% in
the SK-BR-3 cells, when compared with the control (Fig. 3).
Treatment with 5, 10, and 15 µM baicalein reduced the adhe-
sive cells to 151±16, 126±10, and 119±13% in the MCF-7 cells
(Fig. 3A); and 152±5, 103±15, and 89±9% in the SK-BR-3
cells (Fig. 3B), respectively. The data showed that baicalein
effectively suppressed E2-promoted cell adhesion to Matrigel.
Baicalein reduces E2-stimulated cell invasion. T he
Matrigel-coated Transwell chamber system was used to inves-
tigate the effects of baicalein on E2-enhanced cell invasion.
After incubation with E2 for 24 h, the invaded cells across
the Matrigel-coated membrane were increased in both cell
lines (Fig. 4A). In the MCF-7 cell line, the invaded cells were
increased to 176±18% in the E2 treatment group compared
with the control, but decreased to 132±6 and 106±5% in
the 10 and 15 µM baicalein treatment groups, respectively
(Fig. 4B). Similar results were observed in the SK-BR-3 cells.
After SK-BR-3 cells were treated with baicalein, the invaded
cells in the E2-treated group decreased from 240±34 to
231±28%, 165±10% and 114±4% in the 5, 10 and 15 µM treat-
ment groups, respectively (Fig. 4B). Baicalein was evidently
able to suppress E2-enhanced cell invasion.
Baicalein prevents E2-induced GPR30 signal transduction.
The transmembrane receptor, GPR30, is able to mediate
estrogen responsiveness in ER-positive or ER-negative breast
cancer cells. Here, we aimed to determine whether or not
baicalein has inhibitory effects on GPR30 expression or its
signal transduction. We observed that GPR30 expression
remained unchanged after exposure to E2 with or without
baicalein in both the MCF-7 and SK-BR-3 cells (Fig. 5A).
Previous studies have indicated that GPR30 activation triggers
EGFR transactivation by tyrosine phosphorylation in breast
cancer cells (23,24), thus we sought to evaluate the inuence
of baicalein in EGFR phosphorylation upon E2 stimulation.
When cells were treated with baicalein in the presence of E2,
there was a signicant decrease in EGFR phosphorylation
(Tyr 1173) at 10 and 15 µM (Fig. 5B). GPR30 triggers EGFR
transactivation leading to the phosphorylation of ERK1/2 (24)
Figure 5. Effect of baicalein on E2-stimulated activation of the GPR30 signaling pathway. The cells were harvested after E2 (10 nM) with or without baicalein
(5, 10 and 15 µM) treatment for 1 h. (A) Western blot assay was used to examine the expression of GPR30. GAPDH was used as a loading control. (B) The total
and phosphorylated protein levels of EGFR, ERK1/2 and Akt. *P<0.05, signicantly different from the E2-treated group. (C) The total and phosphorylated
protein levels of EGFR, ER K1/2 and Akt in the cells treated with G1 (1 µM) in the presence or absence of baicalein (15 µM) treatment for 1 h. *P<0. 05 vs.
the G1-treated group. Densitometric analysis of the relative ratios of GPR30/GAPDH, phosphor ylated EGFR (Try 1173)/total EGF R, phosphorylated ERK1/2
(Thr 202/Tyr 204)/total ER K1/2 and phosphorylated Akt (Ser473)/total Akt. Values are mean ± SEM. E2, 17β-estradiol; GPR30, G protein-coupled estrogen
receptor; ER K, extracellular signal-regulated kinase; EGFR, epidermal growth factor receptor.
ONCOLOGY REPORTS 7
and activation of the PI3K/Akt transduction pathway (25).
Thus, it was essential to analyze the phosphorylation status
of ERK1/2 and Akt. Consequently, E2-induced phosphory-
lation of ERK1/2 at Thr 202/Tyr 204 and Akt at Ser 373
were repressed in the presence of baicalein at 10 and 15 µM
(Fig. 5B). In addition, GPR30 agonist G1 was used to further
conrm whether baicalein interferes with GPR30 signal trans-
duction. Indeed, treatment with baicalein at 15 µM prevented
G1-induced EGFR, ERK and Akt phosphorylation (Fig. 5C).
Baicalein suppresses E2-induced GPR30-mediated gene
expression. Previous reports have shown that CYR61 a nd CTGF
are the target genes which mediate the stimulatory effects
triggered by GPR30 activation (26). The potential of baicalein
to regulate GPR30-mediated expression of both target genes
was detected by real-time PCR. E2 upregulated CYR61 and
CTGF mRNA levels in the 2 cell lines. The downregulation
of the 2 genes in the MCF-7 and SK-BR-3 cells (Fig. 6A) was
observed after exposure to baicalein, when compared with
the E2-treated groups. In addition, the upregulation of CYR 61
and CTGF mRNA levels induced by G1 in the MCF-7 and
SK-BR-3 cells was also inhibited in the presence of baicalein
at 15 µM (Fig. 6B).
Discussion
Estrogens are essential for the growth and development of
mammary glands (27). However, prolonged exposure to
endogenous or exogenous estrogens is a major risk factor
for the development of breast cancer (28). Estrogens have
been confirmed to play a key role in breast cancer metas-
tasis (29,30). Tumor metastasis consists of a series of highly
relevant biological processes including disruption of cell-cell
and augmentation of cell-ECM adhesion capacity, induction of
cellular motility, invasiveness to the basement membrane and
ECM constituents, angiogenesis, and formation of a metastatic
lesion (31). In the present study, baicalein treatment inhibited
cell migration in response to E2 stimulation. Furthermore, it
suppressed E2-enhanced cell adhesion to Matrigel. Moreover,
it signicantly decreased E2-promoted invasive ability across
a Transwell membrane coated with Matrigel. These results
imply that baicalein may possess anti-estrogenic ability to
suppress E2-induced migration, adhesion and invasion of
MCF-7 and SK-BR-3 cells in vitro.
GPR30 is independent of the ER status in breast cancer
cells and tissue samples, and its action differs from the clas-
sical nuclear ERs, ERα and ERβ (7). Recent studies have
demonstrated that GPR30 mediates E2-enhanced cell migra-
tion and invasion in triple-negative or ERα-negative breast
cancer cells (32,33). Furthermore, we recently found that
GPR30 mediates E2-induced calpain activation, which is
correlated with the metastatic phenotype of breast cancer, in
both ERα-positive and -negative cells (34). We also showed
that tamoxifen, an ER antagonist but a GPR30 agonist, could
not suppress E2-promoted migration but enhanced migra-
tion in breast cancer cells and induced truncation of cyclin E
Figure 6. Effect of baicalein on E2-induced GPR30 target gene transcription. The CY R 61 and CTGF mRNA expression levels were detected by real-time
PCR. (A) The cells were treated with 10 nM E2 in the presence or absence of baicalcei n (5, 10 and 15 µM) for 1 h. Relative mRNA levels of (a) CY R61 and
(b) CTGF. *P<0.05 vs. E2-treated group. (B) The cells were treated with 1 µM G1 with or without 15 µM baicalcein for 1 h. Relative mRNA levels of (a) CYR61
and (b) CTGF. *P<0.05 vs. the G1-treated group. Results obtained from experiments performed in triplicate were norma lized to the β-actin mRNA level and
are shown as fold changes compared to the control cells. Values are mean ± SEM. E2, 17β-estradiol; GPR30, G protein-coupled estrogen receptor; CY R61,
cysteine-rich 61; CTGF, connective tissue growth factor.
SHANG et al: BAICALEIN INHI BITS ESTROGEN-INDUCED MIGRATION, ADHESION AN D INVASION
8
via GPR30 (35). It appears that GPR30, regardless of ERα
expression, plays an important role in E2-enhanced migration
and invasion of breast cancer cells. Our results showed that
baicalein suppressed E2-promoted migration and invasion
in GPR30-positive/ERα-negative SK-BR-3 cells and also
in GPR30/ERα-positive MCF-7 cells. Although baicalein
had little effect on GRP30 expression, it markedly inhibited
the E2-induced GPR30 signal activation, by decreasing
EGFR phosphorylation and activation of ERK1/2 and Akt.
Meanwhile, baicalein suppressed G1-induced upregulation of
the phosphorylation of EGFR, ERK1/2 and Akt. The results
indicate that the inhibitive effect of baicalein on the response
of MCF-7 and SK-BR-3 cells to estrogen may derive partly
from interfering with GPR30 signal transduction.
CYR61 and CTGF, also known as CCN1 and CCN2, belong
to the CCN protein family, which exerts their biological func-
tions by binding and activating cell surface integrins to stimulate
cell growth, adhesion, matrix production and migration, and by
regulating angiogenesis and tumorigenesis (36). CYR61 and
CTGF expression is signicantly associated with tumor size and
lymph node metastasis, suggesting that they play a role in the
progression of breast cancer (20). CYR61 is a survival and pro-
angiogenic factor and induces metastasis and drug resistance
in breast cancer cells (37). CTGF enhances migratory/invasive
processes in human breast cancer by activating the integrin-
αvβ3-ERK1/2-dependent upregulation of S100 calcium-binding
protein A4 (S100A4) (38). The two genes related to breast cancer
metastasis are the target genes of GPR30. Here, we found that
baicalein signicantly in hibited E2- or G1-induced upregulation
of CTGF and CYR61 gene expression. These results suggest
that baicalein may prevent E2-induced migration, invasion
and adhesion of MCF-7 and SK-BR-3 cells also by suppressing
GPR30-dependent gene transcription.
Flavonoids possess a chemical structure similar to estrogen,
and some have been considered to exert an anti-estrogenic
effect and reduce the risk of breast cancer. Genistein, which is
the main ingredient responsible for the cancer chemopreven-
tive activity of soy-derived foods, has garnered much research.
However, it displays a biphasic effect on the proliferation and
death of MCF-7 cells and displays estrogenic activity to trans-
activate ERα at low concentrations. Although baicalein is an
isomer of genistein, it does not have a biphasic effect on ER,
which suggests that it could be a better chemopreventive agent
than genistein (14). Moreover, genistein can stimulate GPR30
to mediate c-fos upregulation in breast cancer cells (39)
and induce the proliferation of thyroid cancer cells (40). In
the present study, we found that baicalein suppressed E2 or
G1-induced GPR30 signal activation indicating a different
activity from genistein. The precise difference between the
two compounds with similar chemical structure remains to
be resolved in future studies. The relationship between the
structure of avonoids and the inhibition of E2 events warrants
further study. To fully investigate the mechanism, we need to
explain the detailed molecular mechanisms with which baica-
lein inuences GPR30 and downstream signaling, as well as
crosstalks with other receptors. In addition, whether baicalein
has dual effects on both ERα and GPR30 still needs further
investigation.
In conclusion, baicalein attenuates E2-induced migration,
adhesion and invasion of MCF-7 and SK-BR-3 breast cancer
cells through interference with GPR30 pathway activation.
This indicates that baicalein may be a promising agent for the
treatment of GPR30-positive breast cancer metastasis.
Acknowledgements
The present study was supported by the Natural Science
Foundation of Guizhou Province of China [grant no. QKH-
J(2014)2007], the National Natural Science Foundation of China
(grant nos. 81302804 and 31360252), Startup Foundation for
Doctors of Guiyang Medical University [grant no. (2013)09] and
the Foundation for Training Programs of Innovation and
Entrepreneurship for Undergraduates of Guiyang Medical
University (grant no. 201410660038).
References
1. Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D:
Global cancer statistics. CA Cancer J Clin 61: 69-90, 2011.
2. Libson S and Lippman M: A review of clinical aspects of breast
cancer. Int Rev Psychiatry 26: 4-15, 2014.
3. Yager JD and Davidson NE: Estrogen carcinogenesis in breast
cancer. N Engl J Med 354: 270-282, 2006.
4. Planey SL, Kumar R and Arnott JA: Estrogen receptors (ERα
versus ERβ): friends or foes in human biology. J Recept Signal
Transduct Res 34: 1-5, 2014.
5. Herynk MH and Fuqua SA: Estrogen receptors in resistance to
hormone therapy. Adv Exp Med Biol 608: 130-143, 2007.
6. Prossnitz ER and Maggiolini M: Mechanisms of estrogen
signaling and gene expression via GPR30. Mol Cell
Endocrinol 308: 32-38, 2009.
7. Maggiolini M and Picard D: The unfolding stories of GPR30,
a new membrane-bound estrogen receptor. J Endocrinol 204:
105-114, 2010.
8. Filardo EJ, Graeber CT, Quinn JA, Resnick MB, Giri D,
DeLellis RA, Steinhoff MM and Sabo E: 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, 2006.
9. Berger CE, Qian Y, Liu G, Chen H and Chen X: p53, a target
of estrogen receptor (ER) α, modulates DNA damage-induced
growth suppression in ER-positive breast cancer cells. J Biol
Chem 287: 30117-30127, 2012.
10. Liu Q, Li JG, Zheng XY, Jin F and Dong HT: Expression of
CD133, PAX2, ESA, and GPR30 in invasive ductal breast
carcinomas. Chin Med J (Engl) 122: 2763-2769, 2009.
11. Takemura H, Sakakibara H, Yamazaki S and Shimoi K: Breast
cancer and avonoids - a role in prevention. Curr Pharm Des 19:
6125-6132, 2 013.
12. Rosenberg Zand RS, Jenkins DJ and Diamandis EP: Flavonoids
and steroid hormone-dependent cancers. J Chromatogr B Analyt
Technol Biomed Life Sci 777: 219-232, 2002.
13. Srinivas NR: Baicalin, an emerging multi-therapeutic agent:
pharmacodynamics, pharmacokinetics, and considerations from
drug development perspectives. Xenobiotica 40: 357-367, 2010.
14. Po LS, Chen ZY, Tsang DS and Leung LK: Baicalein and
genistein display differential actions on estrogen receptor (ER)
transactivation and apoptosis in MCF-7 cells. Cancer Lett 187:
33-40, 2002.
15. Shenouda NS, Zhou C, Browning JD, Ansell PJ, Sakla MS,
Lubahn DB and Macdona ld RS: Phytoestrogens in common herbs
regulate prostate cancer cell growth in vitro. Nutr Cancer 49:
200-208, 2004.
16. Lin CW, Yang LY, Shen SC and Chen YC: IGF-I plus E2 induces
proliferation via activation of ROS-dependent ERKs and JNKs
in human breast carcinoma cells. J Cell Physiol 212: 666-674,
20 0 7.
17. Li Y, Wang JP, Santen RJ, Kim TH, Park H, Fan P and Yue W:
Estrogen stimulation of cell migration involves multiple signaling
pathway interactions. Endocrinology 151: 5146-5156, 2010.
18. Qi Q, Lu N, Wang XT, Gu HY, Yang Y, Liu W, Li C, You QD and
Guo QL: Anti-invasive effect of gambogic acid in MDA-MB-231
human breast carcinoma cells. Biochem Cell Biol 86: 386-395,
2008.
ONCOLOGY REPORTS 9
19. Chen P, Lu N, Ling Y, Chen Y, Hui H, Lu Z, Song X, Li Z, You Q
and Guo Q: Inhibitory effects of wogonin on the invasion of
human breast carcinoma cells by downregulating the expression
and activity of matrix metalloproteinase-9. Toxicology 282:
122-128, 2011.
20. Xie D, Nakachi K, Wang H, Elashoff R and Koefer HP: Elevated
levels of connective tissue growth factor, WISP-1, and CYR61 in
primary breast cancers associated with more advanced features.
Cancer Res 61: 8917-8923, 2001.
21. Pupo M, Pisano A, Lappano R, Santolla MF, De Francesco EM,
Abonante S, Rosano C and Maggiolini M: Bisphenol A induces
gene expression changes and proliferative effects through GPER
in breast cancer cells and cancer-associated broblasts. Environ
Health Perspect 120: 1177-1182, 2012.
22. Ferreira E and Cronjé MJ: Selection of suitable reference genes
for quantitative real-time PCR in apoptosis-induced MCF-7
breast cancer cells. Mol Biotechnol 50: 121-128, 2012.
23. Filardo EJ, Quinn JA and Sabo E: Association of the membrane
estrogen receptor, GPR30, with breast tumor metastasis
and transactivation of the epidermal growth factor receptor.
Steroids 73: 870-873, 2008.
24. Filardo EJ, Quinn JA, Bland KI and Frackelton AR Jr:
Estrogen-induced activation of Erk-1 and Erk-2 requires the
G protein-coupled receptor homolog, GPR30, and occurs via
trans-activation of the epidermal growth factor receptor through
release of HB-EGF. Mol Endocrinol 14: 1649-1660, 2000.
25. Ge C, Yu M and Zhang C: G protein-coupled receptor 30
mediates estrogen-induced proliferation of primordial germ cells
via EGFR/Akt/β-catenin signaling pathway. Endocrinology 153:
3504 -3516, 2012.
26. Pandey DP, Lappano R, Albanito L, Madeo A, Maggiolini M
and Picard D: Estrogenic GPR30 signalling induces proliferation
and migration of breast cancer cells through CTGF. EMBO J 28:
523-532, 20 09.
27. Stingl J: Estrogen and progesterone in normal mammary gland
development and in cancer. Horm Cancer 2: 85-90, 2011.
28. Haldosén LA, Zhao C and Dahlman-Wright K: Estrogen receptor
beta in breast cancer. Mol Cell Endocrinol 382: 665-672, 2014.
29. Ganapathy V, Banach-Petrosky W, Xie W, Kareddula A,
Nienhuis H, Miles G and Reiss M: Lum inal breast cancer metast asis
is dependent on estrogen signaling. Clin Exp Metastasis 29:
493-509, 2012.
30. Zheng S, Huang J, Zhou K, Zhang C, Xiang Q, Tan Z, Wang T
and Fu X: 17β-estradiol enhances breast cancer cell motility and
invasion via extra-nuclear activation of actin-binding protein
ezrin. PLoS One 6: e22439, 2011.
31. Perlikos F, Harrington KJ and Syrigos KN: Key molecular mech-
anisms in lung cancer invasion and metastasis: a comprehensive
review. Crit Rev Oncol Hematol 87: 1-11, 2013.
32. Yu T, Liu M, Luo H, Wu C, Tang X, Tang S, Hu P, Yan Y,
Wang Z and Tu G: GPER mediates enhanced cell viability and
motility via non-genomic signaling induced by 17β-estradiol
in triple-negative breast cancer cells. J Steroid Biochem Mol
Biol 143: 392-403, 2014.
33. Jiang QF, Wu TT, Yang JY, Dong CR, Wang N, Liu XH and
Liu ZM: 17β-estradiol promotes the invasion and migration of
nuclear estrogen receptor-negative breast cancer cells through
cross-talk between GPER1 and CXCR1. J Steroid Biochem Mol
Biol 138: 314-324, 2013.
34. Chen Y, Li Z, He Y, Shang D, Pan J, Wang H, Chen H, Zhu Z,
Wan L and Wang X: Estrogen and pure antiestrogen fulvestrant
(ICI 182 780) augment cell-matrigel adhesion of MCF-7 breast
cancer cells through a novel G protein coupled estrogen receptor
(GPR30)-to-calpain signaling axis. Toxicol Appl Pharmacol 275:
176 -181, 2014.
35. Li Y, Chen Y, Zhu ZX, Liu XH, Yang L, Wan L, Lei TW and
Wang XD: 4-Hydroxytamoxifen-stimulated processing of
cyclin E is mediated via G protein-coupled receptor 30 (GPR30)
and accompanied by enhanced migration in MCF-7 breast cancer
cells. Toxicology 309: 61- 65, 2013.
36. Jun JI and Lau LF: Taking aim at the extracellular matrix:
CCN proteins as emerging therapeutic targets. Nat Rev Drug
Discov 10: 945-963, 2011.
37. Espinoza I, Liu H, Busby R and Lupu R: CCN1, a candidate
target for zoledronic acid treatment in breast cancer. Mol Cancer
Ther 10: 732-741, 2011.
38. Chen PS, Wang MY, Wu SN, et al: CTGF enhances the motility of
breast cancer cel ls via an integr in-alphavbeta 3-ER K1/2-dependent
S100A4-upregulated pathway. J Cell Sci 120: 2053-2065, 2007.
39. Maggiolini M, Vivacqua A, Fasanella G, Recchia AG, Sisci D,
Pezzi V, Montanaro D, Musti AM, Picard D and Andò S: 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, 2004.
40. Vivacqua A, Bonofiglio D, Albanito L, Madeo A, Rago V,
Carpino A, Musti AM, Picard D, Andò S and Maggiolini M:
17beta-estradiol, genistein, and 4-hydroxytamoxifen induce the
proliferation of thyroid cancer cells through t he g protein-coupled
receptor GPR30. Mol Pharmacol 70: 1414-1423, 2006.
... These results suggest that the antiproliferative activity of flavonoid is independent of the presence of estrogen receptors on the cell surface, which excludes the possibility of competition between baicalein and estradiol for binding via the estrogen receptor. Besides, baicalein can inactivate the G protein-coupled estrogen receptor (GPR30) signaling pathway and decrease the phosphorylation of AKT and ERK 1/2, the tyrosine phosphorylation of AKT and ERK 1/2, and the tyrosine phosphorylation of epidermal growth factor receptor (EGFR) in breast cancer cells, leading to the suppression of the migration and invasion of cancer [52]. The mentioned properties In the first stage of our study, we analyzed the cytotoxicity of baicalin and its aglycone baicalein towards the estrogen-dependent breast cancer cell line MCF-7 and the human umbilical vein endothelial cell line HUVEC-ST. ...
... These results suggest that the antiproliferative activity of flavonoid is independent of the presence of estrogen receptors on the cell surface, which excludes the possibility of competition between baicalein and estradiol for binding via the estrogen receptor. Besides, baicalein can inactivate the G protein-coupled estrogen receptor (GPR30) signaling pathway and decrease the phosphorylation of AKT and ERK 1/2, the tyrosine phosphorylation of AKT and ERK 1/2, and the tyrosine phosphorylation of epidermal growth factor receptor (EGFR) in breast cancer cells, leading to the suppression of the migration and invasion of cancer [52]. The mentioned properties make baicalein helpful in supporting the treatment of receptor-negative cancers characterized by a worse prognosis and poor response to therapy. ...
Article
Full-text available
Breast cancer is a major health concern and the leading cause of death among women worldwide. Standard treatment often involves surgery, radiotherapy, and chemotherapy, but these come with side effects and limitations. Researchers are exploring natural compounds like baicalin and baicalein, derived from the Scutellaria baicalensis plant, as potential complementary therapies. This study investigated the effects of baicalin and baicalein on the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel, commonly used chemotherapeutic drugs for breast cancer. The analysis included breast cancer cells (MCF-7) and human endothelial cells (HUVEC-ST), to assess potential effects on healthy tissues. We have found that baicalin and baicalein demonstrated cytotoxicity towards both cell lines, with more potent effects observed in baicalein. Both flavonoids, baicalin (167 µmol/L) and baicalein (95 µmol/L), synergistically enhanced the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel in breast cancer cells. In comparison, their effects on endothelial cells were mixed and depended on concentration and time. The results suggest that baicalin and baicalein might be promising complementary agents to improve the efficacy of doxorubicin and docetaxel anticancer activity. However, further research is needed to validate their safety and efficacy in clinical trials.
... Baicalein (BAI) is the principal avonoid isolated from S.baicalensis, which possesses numerous bene cial properties and is always utilized for treating multiple ailments [9]. Recent research has revealed that BAI exerts its anticancer activity by facilitating apoptosis, blocking cell growth, activating autophagy and regulating some molecular pathways, such as EGFR/ERK/NF-κβ pathway [10] or EGFR/ERK/Akt pathway [11]. Moreover, scholars overlapped BAI targets with oral squamous cell carcinoma-associated genes, EGFR belongs to the top 10 genes with high centrality measures [12]. ...
... These results provided compelling evidence that EGFR is essential for LGR4 to facilitate the progression of GBM. BAI has been reported to have the potential to treat HCC or BC by negatively regulating EGFR/ERK/NF-κβ pathway [10] or EGFR/ERK/Akt pathway [11]. Consistently, in vitro experiments revealed that after treatment with BAI, the expressions of LGR4 and EGFR were decreased and BAI interacted with LGR4 directly. ...
Preprint
Full-text available
Patients with glioblastoma (GBM) have poor prognoses and limited therapeutic options. LGR4 was reported to overexpressed in GBM and involved in tumorigenesis of many cancers, Baicalein (BAI) is a kind of flavonoid that exhibited anti-tumor effects in various tumors. However, the function and association of BAI and LGR4 in GBM are still unclear. In thisi study, firstly, GEPIA and HPA databas was used to perform expression and survival analysis of LGR4 in GBM patients. Then, the significance of LGR4-EGFR in GBM cells (HS683 and KNS89) and GBM animal models was explored by RNA interference and subcutaneous transplantation. Additionally, GBM cells were treated with BAI to explore the role and mechanism of BAI involved in GBM. The results showed that LGR4 was highly expressed in GBM and related to bad prognosis. LGR4 knockdown obviously repressed the proliferation and EGFR expression but induced apoptosis in GBM cells, however, the situations were reserved by EGFR overexpression and CBL knockdown. In contrast, both in vitro and in vivo experiments revealed LGR4 overexpression facilitated GBM cellular biological activities and promoted tumor development, but the effects were rescued by BAI and EGFR inhibitor. In addition, si-LGR4 accelerated EGFR protein degradation while oe-LGR4 exhibit opposite effect. Without affecting normal cellular viability, BAI inhibited malignant behaviour, interacted with LGR4 and blocked the LGR4-EGFR pathway in both GBM cells. Taken together, our data suggested that BAI could inhibit GBM cell proliferation and induce apoptosis via downregulation of the LGR4-EGFR pathway, and the LGR4-EGFR pathway may be an underlying target for GBM therapy of BAI.
... Choi et al. reported that baicalein from Scutellaria baicalensis significantly inhibited cell migration and invasion by inhibiting the activities of MMP-2 and MMP-9 (via the AKT1 pathway) [23], which showed great future prospects for the treatment of breast cancer. Shang et al. found that baicalein could suppress 17-β-estradiol-induced migration, adhesion, and invasion of breast cancer cells via the G-protein-coupled receptor 30 signaling pathway [24]. Ma et al. demonstrated that baicalein could inhibit epithelial-mesenchymal transition (EMT) by downregulating SATB1 and the Wnt/β-catenin pathway and further inhibited the metastasis of MDA-MB-231 breast cancer cells in vivo [25]. ...
... Sci. 2023, 24, 3594 ...
Article
Full-text available
Scutellaria baicalensis is often used to treat breast cancer, but the molecular mechanism behind the action is unclear. In this study, network pharmacology, molecular docking, and molecular dynamics simulation are combined to reveal the most active compound in Scutellaria baicalensis and to explore the interaction between the compound molecule and the target protein in the treatment of breast cancer. In total, 25 active compounds and 91 targets were screened out, mainly enriched in lipids in atherosclerosis, the AGE–RAGE signal pathway of diabetes complications, human cytomegalovirus infection, Kaposi-sarcoma-associated herpesvirus infection, the IL-17 signaling pathway, small-cell lung cancer, measles, proteoglycans in cancer, human immunodeficiency virus 1 infection, and hepatitis B. Molecular docking shows that the two most active compounds, i.e., stigmasterol and coptisine, could bind well to the target AKT1. According to the MD simulations, the coptisine–AKT1 complex shows higher conformational stability and lower interaction energy than the stigmasterol–AKT1 complex. On the one hand, our study demonstrates that Scutellaria baicalensis has the characteristics of multicomponent and multitarget synergistic effects in the treatment of breast cancer. On the other hand, we suggest that the best effective compound is coptisine targeting AKT1, which can provide a theoretical basis for the further study of the drug-like active compounds and offer molecular mechanisms behind their roles in the treatment of breast cancer.
... Baicalein neither directly binds to GPR 30 receptor nor does it affects the expression of GPR 30 receptor, rather it suppresses the E2 induced phosphorylation of SRC, EGFR, ERK & AKT, the mechanism responsible for GPR 30 signaling activation. It also down regulate the expression of CYR 61, CTGF, target gene of GPR30 [44]. ...
Article
Full-text available
Breast cancer is the most prominent type of cancer among females and treatment through chemotherapy generates serious side effects too along with the cure. Hence, the use of natural compounds as promising therapeutic agents is emerging as an effective alternate due to their low toxicity and bio-friendly nature. One such bioactive compound is baicalein, a trihydroxy flavone, obtained from Scutellaria baicalensis, Scutellaria lateriflora, Oroxylum indicum, and thyme plant. The extract of the different parts of the plant is traditionally used in ayurvedic preparations because of its antioxidant, anti-inflammatory, antimicrobial, antidiarrheal, and hepatoprotective properties. The anticancer potential of baicalein against breast cancer comprises the mechanism of cell cycle arrest, apoptosis generation, induction of cell invasion, and migration. This review describes the general characteristics of baicalein, the detailed molecular mechanism of action against breast cancer and the different delivery approach developed so far to increase its solubility and bioavailability. Overall, baicalein has the properties to develop as a new therapeutic agent for breast cancer treatment. Graphical Abstract
Article
G protein coupled receptors (GPCRs) are among the largest family of cell surface receptors found in the human genome. They govern a wide range of physiological responses in both health and diseases, making them one of the potential targeted surface receptors for pharmaceuticals. Flavonoids can modulate GPCRs activity by acting as allosteric ligands. They can either enhance or reduce the GPCR's effect. Emerging research shows that individual flavonoids or mixtures of flavonoids from plant extracts can have relevant pharmacological effects against a number of diseases, particularly by influencing GPCRs. In the present review, we are considering to give a comprehensive overview of flavonoids and related compounds that exhibit GPCRs activity and to further explore which beneficial structural features. Molecular docking was used to strengthen experimental evidence and describe flavonoid-GPCRs interactions at molecular level
Article
Full-text available
The high reoperation rate in pelvic organ prolapse (POP) surgery is probably due to impaired wound healing, and therefore, novel strategies that promote tissue regeneration are urgently needed. Estrogen therapy shows beneficial effects for vaginal wound healing: it reduces the inflammatory response and improves vaginal tissue strength and quality. Earlier, scaffolds that release the most potent estrogen, β‐estradiol (E2) have been described. This work describes the results of conjugating E2 covalently to the synthetic, but highly mimetic, polyisocyanide hydrogel (PIC‐E2). As adipose‐derived stem cells (ASCs) possess regenerative capabilities, the combination of PIC‐E2 and ASCs may have a synergistic effect on pelvic floor tissue regeneration. The results show that the PIC‐E2 bioactivity is not hampered upon covalent E2 conjugation and upregulates various extracellular matrix (ECM) genes in ASCs. Moreover, PIC‐E2 exerts similar effects regarding ECM metabolism compared to other E2‐releasing biomaterials, but at much lower doses, which is economically and therapeutically favorable. Based on these in vitro findings, the combination of the PIC‐E2 hydrogel and ASCs may have the potential to stimulate tissue regeneration in vivo, and therefore, feasibly improve the surgical outcomes for POP in the future.
Article
The upregulation of the HER2 oncogene is associated with a variety of human cancers and is associated with poor prognosis. Baicalein is reported to have anti-tumor activity, but the molecular mechanism of this effect in HER2-positive cancer cells has not been studied. In this study, our data showed that baicalein can inhibit the proliferation and transformation potential of ovarian cancer cells overexpressing HER2. Baicalein treatment caused a dose-dependent inhibition of HER2 gene expression at the transcriptional level. Baicalein acted on ovarian cancer cells overexpressing HER2 to downregulate the PI3K/Akt signaling pathway downstream of HER2 and inhibit the expression or activity of downstream targets, such as VEGF and cyclin D1 and MMP2. Oral administration of baicalein supplemented with a pharmaceutical excipient significantly inhibited the growth of HER2-overexpressing ovarian SKOV-3 cancer xenografts in mice. These results suggest that downregulation of HER2 gene expression by baicalein at the transcriptional level contributes to inhibit the in vitro and in vivo proliferation and HER2-mediated malignant transformation of HER2-overexpressing ovarian cancer cells.
Article
The actions of estrogens and related estrogenic molecules are complex and multifaceted in both sexes. A wide array of natural, synthetic, and therapeutic molecules target pathways that produce and respond to estrogens. Multiple receptors promulgate these responses, including the classical estrogen receptors of the nuclear hormone receptor family (estrogen receptors α and β), which function largely as ligand-activated transcription factors, and the 7-transmembrane G protein–coupled estrogen receptor, GPER, which activates a diverse array of signaling pathways. The pharmacology and functional roles of GPER in physiology and disease reveal important roles in responses to both natural and synthetic estrogenic compounds in numerous physiological systems. These functions have implications in the treatment of myriad disease states, including cancer, cardiovascular diseases, and metabolic disorders. This review focuses on the complex pharmacology of GPER and summarizes major physiological functions of GPER and the therapeutic implications and ongoing applications of GPER-targeted compounds.
Article
Breast cancer is one of the most common malignancies in women, and exhibits high metastasis, recurrence and fatality rates. Novel therapies for breast cancer are constantly emerging, such as targeted therapy, oncolytic virotherapy, and immunotherapy. Despite their potential, these new therapies are still in their infancy, and chemotherapy remains the standard treatment for breast cancer. Therefore, it is of great significance to develop safe and efficient treatment drugs or adjuvants for breast cancer treatment. Traditional Chinese medicine (TCM) has a long clinical history in China, in which Scutellaria baicalensis Georgi exhibits favorable antibreast cancer activities. We therefore conducted a systematic review of the available literature to better understand the molecular mechanisms of S. baicalensis in breast cancer treatment. S. baicalensis and its active components (baicalein, baicalin, wogonin, wogonoside, oroxylin A and scutellarin) exhibited promising antibreast cancer activity through proliferation inhibition, apoptosis induction, invasion and metastasis blockading, and drug-resistance and non-coding RNA regulation. Additionally, senescence, autophagy, angiogenesis, and glycolysis mechanisms were observed to play a role in their antibreast cancer activity. Furthermore, multiple signaling pathways contributed to the antitumor effects of S. baicalensi, such as the NF-[Formula: see text]B, Wnt/[Formula: see text]-catenin, SATB1, Bcl2 family proteins, Caspase, PI3K/Akt, mTOR, ERK, p38-MAPK, TGF-[Formula: see text]/Smad, and Hippo/YAP pathways. This review provides valuable insights into the role of S. baicalensis as a breast cancer treatment and acts as a foundation for further investigations in this field.
Article
Flavonoids are a major group of phytoestrogens associated with physiological effects, and ecological and social impacts. Although the estrogenic activity of flavonoids was reported by researchers in the fields of medical, environmental and food studies, their molecular mechanisms of action have not been comprehensively reviewed. The estrogenic activity of the respective classes of flavonoids, anthocyanidins/anthocyanins, 2-arylbenzofurans/3-arylcoumarins/α-methyldeoxybenzoins, aurones/chalcones/dihydrochalcones, coumaronochromones, coumestans, flavans/flavan-3-ols/flavan-4-ols, flavanones/dihydroflavonols, flavones/flavonols, homoisoflavonoids, isoflavans, isoflavanones, isoflavenes, isoflavones, neoflavonoids, oligoflavonoids, pterocarpans/pterocarpenes, and rotenone/rotenoids, was summarized through a comprehensive literature search, and their structure-activity relationship, biological activities, signaling pathways, and applications were discussed. Although the respective classes of flavonoids contained at least one chemical mimicking estrogen, the mechanisms varied, such as those with estrogenic, anti-estrogenic, non-estrogenic, and biphasic activities, and additional activities through crosstalk/bypassing, which exert biological activities through cell signaling pathways. Such mechanistic variations of estrogen action are not limited to flavonoids and are observed among other broad categories of chemicals, thus this group of chemicals can be termed as the “estrogenome”. This review article focuses on the connection of estrogen action mainly between the outer and the inner environments, which represent variations of chemicals and biological activities/signaling pathways, respectively, and form the basis to understand their applications. The applications of chemicals will markedly progress due to emerging technologies, such as artificial intelligence for precision medicine, which is also true of the study of the estrogenome including estrogenic flavonoids.
Article
Full-text available
Estrogen is essential for growth and development of the mammary glands and has been associated with the promotion and growth of breast cancer and in line with this, most human breast cancers are initially estrogen-dependent and undergo regression when deprived of their supporting hormone. Estrogen exerts many of its effects via two nuclear estrogen receptors (ERs), ERα and ERβ. The discovery of a second ER, ERβ, demanded a full re-evaluation of estrogen action in all target tissues and different estrogen associated diseases, including human breast cancer. However, despite over 15 years of research, the exact role, if any, of ERβ in human breast cancer remains elusive. The main challenges now are to develop highly selective anti-ERβ antibodies that are applied to large well characterized human breast cancer samples to validate their diagnostic potential and to explore ERβ-selective agonists in animal models of breast cancer to validate their therapeutic potential.
Article
Full-text available
Abstract Most of the biological effects of estrogens are mediated via the estrogen receptors (ERs) at the level of gene regulation. Recently, new information regarding the role of ERs in physiology, pathology and the mechanisms through which estrogens bring about these functions has emerged. The physiological effects of estrogen are manifested through two ER isoforms - ERα and ERβ - which display distinct regions of sequence homology. The crystal structures of these receptors bound to their specific ligands (e.g. agonists or antagonists) have revealed much about how ligand binding alters receptor structure/conformation and the interaction with coactivators or corepressors as well as how it determines the cellular response to a ligand. ERs are involved in the variety of physiological and pathological activities and different cells and tissues have shown divergent responses to these two receptor isoforms. The discovery of sub-isoforms of ER alpha and beta has further complicated our understanding of how the interaction between ERs and its ligands contribute to the development of disease. Nevertheless, continuing efforts in the study of ERs have helped us to more clearly define their role in disease and to develop novel, ER-targeted therapeutics.
Article
Steroid-hormone dependent cancers, including those of the breast, prostate and colon, are leading causes of morbidity and mortality in western countries. In rural Asian areas, these diseases are relatively uncommon. Dietary factors, including low consumption of fruit, vegetables and soy in the west have been shown in various epidemiologic studies as reasons for these differences. This review discusses flavonoids, one component of these plant foods that is being investigated for their role in chemoprevention. Epidemiological, in vitro, animal and human studies shall be explored to look at mechanisms involved, including steroid hormone activity, effects on cell growth, antioxidant activities, inhibition of chemical carcinogenesis and influences on modulators of cancer risk. Although the in vitro and animal models point to several pathways by which flavonoids may reduce incidence of these cancers, the clinical data are still relatively lacking. More research is needed to determine how best to use foods containing these compounds to reduce steroid hormone-dependent cancer risk.
Article
Triple-negative breast cancer (TNBC) is an aggressive breast cancer with a generally poor prognosis. Due to lack of specific targets for its treatment, an efficient therapy is needed. G protein-coupled estrogen receptor (GPER), a novel estrogen receptor, has been reported to be expressed in TNBC tissues. In this study, we investigated the effects of blocking non-genomic signaling mediated by the estrogen/GPER pathway on cell viability and motility in the TNBC cells. GPER was strongly expressed in the TNBC cell lines MDA-MB-468 and MDA-MB-436, and the estrogen-mediated non-genomic ERK signaling activated by GPER was involved in cell viability and motility of TNBC cells. Treatment with 17β-estradiol (E2), the GPER-specific agonist G-1 and tamoxifen (TAM) led to rapid activation of p-ERK1/2, but not p-Akt. Moreover, estrogen/GPER/ERK signaling was involved in increasing cell growth, survival, and migration/invasion by upregulating expression of cyclinA, cyclinD1, Bcl-2, and c-fos associated with the cell cycle, proliferation, and apoptosis. Immunohistochemical analysis of TNBC specimens showed a significantly different staining of p-ERK1/2 between GPER-positive tissues (58/66, 87.9%) and GPER-negative tissues (13/30, 43.3%). The positivity of GPER and p-ERK1/2 displayed a strong association with large tumor size and poor clinical stage, indicating that GPER/ERK signaling might also contribute to tumor progression in TNBC patients which corresponded with in vitro experimental data. Our findings suggest that inhibition of estrogen/GPER/ERK signaling represents a novel targeted therapy in TNBC.
Article
Abstract Breast cancer is the most frequently diagnosed cancer in women and ranks second among causes for cancer related death in women. The ability to identify and diagnose breast cancer has improved markedly. Treatment decisions which were based in the past predominantly on the anatomic extent of the disease are shifting to the underlying biological mechanisms. Gene array technology has led to the recognition that breast cancer is a heterogeneous disease composed of different biological subtypes, and genetic profiling enables response to chemotherapy to be predicted. Breast conservation became an established standard of care and the oncoplastic approach enables wide excisions without compromising the natural shape of the breast. Sentinel lymph node biopsy has replaced axillary dissection as the standard procedure to stage the axilla and spared many patients the excess morbidity of axillary dissection. Targeted therapy to the oestrogen receptor plays a major role in systemic therapy; pathways responsible for endocrine resistance have been targeted as well. Biological therapy has been developed to target HER2 receptor and combination of antibody drug conjugates linked cytotoxic therapy to HER2 antibodies. Meaningful improvements in survival resulted from the new effective systemic agents and patients with metastasis are likely to have a longer survival.
Article
G protein-coupled estrogen receptor 1 (GPER1) is widely expressed in human breast cancers correlating with increased tumor size and malignancy. Although estrogen signaling via GPER1 was extensively studied in recent years, the underlying molecular mechanism of GPER1-associated metastasis of breast cancer still remains unclear. In this study, the main aims were focused on the potential role of GPER1 in regulating migration and invasion of nuclear estrogen receptor (ER)-negative breast cancer cells upon 17β-estradiol (E2) stimulation and the involved signaling pathway. Key events in estrogen signaling were chosen for our studies, such as the activation of ERK and AKT, nuclear translocation of NF-κB and secretion of Interleukin-8 (IL-8). The migration and invasion activities upon E2 stimulation were also examined in ER-negative SKBR3 and BT-20 breast cancer cells. Compared with ER-positive MCF-7 breast cancer cells, both SKBR3 and BT-20 cells had very similar expression of GPER1, but relatively high expression of CXC receptor-1 (CXCR1), which is considered as an active regulator for cancer metastasis upon binding IL-8. Results showed that E2 facilitated the activation of ERK, AKT and NF-κB, which could be significantly attenuated by GPER1 blockage or knock-down in both SKBR3 and BT-20 cells. Moreover, increased secretion of IL-8 induced by E2 was also inhibited either by specific inhibitors for GPER1, ERK, AKT, and NF-κB, or by knock-down for GPER1. Furthermore, E2 could activate the migration and invasion of both SKBR3 and BT-20 cells, which in turn could also be inhibited by blocking GPER1, ERK, AKT, NF-κB, and CXCR1, respectively, or knock-down for GPER1 and CXCR1. In conclusion, we demonstrated that estrogen signaling via GPER1 associated with the metastasis of breast cancer, which might be through GPER1/ERK&AKT/NF-κB/IL-8/CXCR1 cascade. The cross-talk between GPER1 and CXCR1 could be another potential target for the therapy of metastatic breast cancer.
Article
Over-expression of cleaved cyclin E in breast tumors is closely associated with tumor progression and resistance to antiestrogens. 17β-estradiol (E2) has been recently shown to induce cyclin E processing in breast cancer cells. Tamoxifen has been used in patients with estrogen-sensitive breast cancer, yet resistance to antiestrogens and reccurrence will appear in some of the patients after its continued use. We therefore addressed possible effects of tamoxifen on the generation of cleaved cyclin E and its signal mechanism(s) in estrogen-responsive MCF-7 breast cancer cells that express both G protein-coupled protein (GPR) 30 and estrogen receptor α (ERα). 4-hydroxytamoxifen (OHT, tamoxifen's active form) failed to prevent E2-induced proteolysis of cyclin E and migration, but rather triggered cyclin E cleavage coincident with augmented migration. OHT-induced cyclin E truncation also occurred in SK-BR-3 cells that express GPR30 and lack ERα, but not in MDA-MB-231 cells that express neither GPR30 nor ERα. G1, a specific GPR 30 agonist, caused dramatic proteolysis of cyclin E and enhanced migration. Furthermore, OHT-stimulated cleavage of cyclin E and migration were tremendously attenuated by G15, a GPR30 antagonist, or siRNA against GPR30. In addition, inhibitors for EGFR or ERK1/2 remarkably suppressed OHT-induced truncation of cyclin E, suggesting involvement of EGFR signaling. Collectively, our data indicate that OHT contributes to the production of proteolyzed cyclin E via GPR30 with augmented migration in MCF-7 cells.
Article
Endogenous estrogens, such as 17β-estradiol (E2), are implicated in the development of breast cancer. The putative mechanisms by which estrogens exert the carcinogenic effects have been recognized to involve the redox cycling of estrogen metabolites and subsequent estrogen-DNA adduct formation as well as the estrogen receptor-dependent pathway of estrogen-induced cell growth. The former pathway is regulated by phase I enzymes, mainly cytochrome P450 (CYP) 1A1, 1A2, and 1B1. Among them, CYP1B1 predominantly catalyzes the C4-position of E2 and forms carcinogenic 4-hydroxy-E2 (4-OHE2), whereas CYP1A1 and CYP1A2 convert E2 to noncarcinogenic 2-hydroxy-E2. Formed 4-OHE2 is further oxidized to semiquinones and quinones, which form DNA adducts, leading to mutagenic lesions. Consequently, CYP1B1 is highly expressed, and 4-OHE2 is predominantly detected in estrogen target neoplastic tissues. Moreover, invasion and metastasis are also involved in the development of breast cancer. Epidemiological studies suggest an inverse association between a higher intake of flavonoids and breast cancer risk. Flavonoids, which are widely distributed in the plant kingdom, have been recently reported as candidate compounds that can exert chemopreventive effects in estrogen-dependent or independent breast cancer. In this review, we provide a comprehensive overview of breast cancer and chemoprevention by flavonoids, mainly focusing on ER-mediated hormonal regulation, redox cycling of estrogen metabolites, and selective inhibition of CYP1B1.