Você está na página 1de 11

Journal of Pharmacological Sciences 128 (2015) 97e107

H O S T E D BY Contents lists available at ScienceDirect

Journal of Pharmacological Sciences


journal homepage: www.elsevier.com/locate/jphs

Full paper

ERa down-regulation plays a key role in silibinin-induced autophagy


and apoptosis in human breast cancer MCF-7 cells
Nan Zheng a, Ping Zhang a, Huai Huang a, Weiwei Liu a, Toshihiko Hayashi a, Linghe Zang a,
Ye Zhang a, Lu Liu a, Mingyu Xia a, Shin-ichi Tashiro b, Satoshi Onodera c,
Takashi Ikejima a, *
a
ChinaeJapan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang 110016, China
b
Institute for Clinical and Biomedical Sciences, Kyoto 603-8072, Japan
c
Department of Clinical and Biomedical Sciences, Showa Pharmaceutical University, Tokyo 194-8543, Japan

a r t i c l e i n f o a b s t r a c t

Article history: The estrogen receptor alpha (ERa) has been proven to be one of the most important therapeutic targets
Received 24 November 2014 in breast cancer over the last 30 years. Previous studies pointed out that a natural flavonoid, silibinin,
Received in revised form induced apoptosis in human breast cancer MCF-7 cells. In the present study we report that exposure of
10 March 2015
MCF-7 cells to silibinin led to cell death through the down-regulation of ERa expression. Silibinin-
Accepted 1 May 2015
Available online 12 May 2015
induced apoptosis of MCF-7 cells through up-regulation of caspase 6 due to ERa signalling repression
was further boosted by ERa antagonist. Moreover, up-regulation of autophagy induced by silibinin
accounted for apoptotic exacerbation, being further enhanced by ERa inhibition. Upon ERa activation,
Keywords:
Silibinin
series of downstream signalling pathways can be activated. We found that silibinin reduced the ex-
Estrogen receptor alpha (ERa) pressions of Akt/mTOR and extracellular-signal-related kinase (ERK), which respectively accounted for
MPP dihydrochloride hydrate (MPP) the induction of autophagy and apoptosis. These effects were further augmented by co-treatment with
Apoptosis ERa inhibitor. We conclude that the treatment with silibinin of ERa-positive MCF-7 cells down-regulates
Autophagy the expression of ERa, and subsequently mTOR and ERK signaling pathways, ERa downstream, finally
resulting in induction of autophagy and apoptosis.
© 2015 The Authors. Production and hosting by Elsevier B.V. on behalf of Japanese Pharmacological
Society. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/
licenses/by-nc-nd/4.0/).

1. Introduction thistle Silybum marianum (L.) Gaertn. Silibinin has been extensively
used for its hepatoprotective effectiveness in Asia and Europe (3,4).
In the modern world, breast cancer is one of the most common According to the reports, flavonoids have been shown to be cyto-
malignances in women. It has been known more than a century (1) toxic to cancer cells (5,6). It has been reported that the estrogen
that breast cancer is associated with cell-surface estrogen receptor receptor is required for flavonoid-induced cytotoxicity in breast
(ER). Human ER exists as two subtypes, ERa and ERb, which regu- cancer cell lines (7,8). Silibinin induces a loss of cell viability in MCF-
late the transcription of various target genes upon binding to es- 7 cells. However, underlying mechanism has not been completely
trogen response elements (ERE) present within the regulatory elucidated. It is not known if the estradiol-like effect of silibinin
region of the genes. In most ERa-positive breast cancers, especially contributes to its anti-cancer potency. Therefore, we focused on
in early stages, the expression level of ERa is considerably higher ERa regulation in the cytotoxicity of silibinin in MCF-7 cells that
than that in normal breast epithelium (2). Therefore, targeting the inherently express high levels of ERa.
ERa signalling pathway has already become a focal point in the
development of breast cancer therapy. 2. Materials and methods
Silibinin (Fig. 1A), a natural polyphenolic flavonoid, is a major
bioactive component of silymarin isolated from the plant milk 2.1. Reagents

* Corresponding author. Tel./fax: þ86 24 23844463.


Silibinin with a purity of 99% was purchased from Jurong Best
E-mail address: ikejimat@vip.sina.com (T. Ikejima). Medicine Material (Zhenjiang, Jiangsu, China). The reagent was
Peer review under responsibility of Japanese Pharmacological Society. dissolved in dimethylsulfoxide (DMSO) to make a stock solution.

http://dx.doi.org/10.1016/j.jphs.2015.05.001
1347-8613/© 2015 The Authors. Production and hosting by Elsevier B.V. on behalf of Japanese Pharmacological Society. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
98 N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107

Fig. 1. The cytotoxicity of silibinin in MCF-7 cells is correlated with the down-regulation of estrogen receptor a (ERa). (A) The chemical structure of silibinin. (B) The cells were
treated with various doses of silibinin for 12, 24, 36 or 48 h. Viability was measured by MTT assay. (C) Western blot analysis of ERa levels after treatment with 100, 200 and 300 mM
silibinin for 24 h b-actin was used as an equal loading control. (D) The cells were pre-treated with 10, 20 and 40 mM ICI 182,780 for 1 h prior to silibinin (200 mM) administration for
24 h, and the cell viability was measured by MTT assay, n ¼ 3, mean ± S.E.M, *p < 0.05, compared with silibinin -treated group. (E) The different suppression of silibinin in ERa-high
expressive MCF-7 and ERa-low expressing MDA-MB231 (MB231) human breast cancer cells. Compare between the two strains of human breast cancers at the specific concentration
of silibinin, **p < 0.01. ERa levels were examined by western blot analysis. (F) Cells were cultured for 24 h and then incubated with different concentrations of MPP (a specific ERa
antagonist MPP dihydrochloride) for 1 h, and then silibinin 200 mM was added and cultured for 24 h. The viability was determined by the MTT assay, n ¼ 3, mean ± S.E.M, **p < 0.01,
compared with silibinin-treated group. (G) The cells were transfected with control or ERa-targeting siRNA for 48 h, and then incubated with different concentrations of silibinin for
24 h. The viability was measured by MTT assay, n ¼ 3, mean ± S.E.M, *p < 0.05, **p < 0.01, si-ERa compared with si-con group.
N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107 99

MEM and DMEM complete media were used for dilution and the reader (Thermo Scientific Multiskan MK3, Shanghai, China). Cell
final concentration of DMSO was kept below 0.1% in cell culture, growth inhibitory ratio was calculated using the following
which had no detectable effects on cells. Methylthiazolyldiphenyl- equation:
tetrazolium bromide (MTT), propidiumiodide (PI), RNase A, mon- Cell growth inhibitory ratio (%) ¼ 100  (A490, controlA490, sam-
odansyl cadaverine (MDC), rhodamine 123, estrogen receptor (ER) ple)/(A490, controlA490, blank)
antagonist Fulvestrant (ICI 182,780), estrogen receptor alpha (ERa)-
specific antagonist MPP 1,3-Bis(4-hydroxyphenyl)-4-methyl-5-[4- 2.6. Flow cytometric analysis of dying cells
(2-piperidinylethoxy)phenol]-1H-pyrazole-dihydro- chloride, and
primary antibody against LC3 were purchased from Sigma Chemi- Propidium iodide (PI), a fluorescent dye that specifically binds to
cal (St. Louis, MO, USA). Polyclonal antibody against ERa was pur- cellular DNA, was used to quantify DNA content. Cells subjected to
chased from Proteintech Group (Chicago, IL, USA). Primary the indicated treatments were collected (5  105 cells per sample),
antibodies against p62, caspase-6, caspase-8, Bid, Akt1/2, p-Akt1/2, fixed with 70% (v/v) ethanol at 4  C overnight, rinsed with ice-cold
Bcl-2, Bax, cytochrome c, ERK1, p-ERK 1/2, GRB2, Raf, SOS, inhibitor PBS twice and incubated with 1 mL of PI solution (PI 50 mg/L and
of caspase-activated DNase (ICAD), poly-ADP-ribose polymerase RNase A1g/L) in the dark for 30 min. The cellular DNA content was
(PARP) and b-actin, as well as horseradish peroxidase-conjugated next analyzed using a FACScan flow cytometer. Cells with DNA
secondary antibodies, were purchased from Santa Cruz Biotech- content less than normal diploid (at subG0/G1 phase) are taken as
nology (Santa Cruz, CA, USA). The SuperSignal West Pico Chemi- dead cells.
luminescent Substrate® used in conjunction with the horseradish
peroxidase (HRP) enzyme was purchased from Thermo Scientific 2.7. Determination of mitochondrial membrane potential
(Rockford, IL, USA).
The mitochondrial membrane potential was examined using the
2.2. Cells culture mitochondrial dye rhodamine 123. After incubation with indicated
treatments, MCF-7 cells were collected and stained with 1 mg/mL
Human breast cancer MCF-7, MDA-MB231 cells and human rhodamine 123 at 37  C for 30 min. The fluorescence intensity of
hepatoma G2 (HepG2) were obtained from American Type Culture the cells was analysed by FACScan flow cytometry.
Collection (ATCC) (Manassas, VA, USA). MCF-7 cells were cultured
in Minimum Essential Medium (MEM, Gibco, Grand Island, NY, 2.8. Transfection of siRNA
USA), while MDA-MB231 and HepG2 cells were in Dulbecco's
Modified Eagle medium (DMEM, Gibco, Grand Island, NY, USA). All MCF-7 cells were transfected with negative control siRNA (NC-
media were supplemented with 10% fetal bovine serum (FBS) siRNA) or siRNA targeting ERa (ERa-siRNA) at a final concentration
(Beijing Yuanheng Shengma Research Institution of Biotechnology, of 10 nM using siRNA-Mate (GenePharma, Shanghai, China) ac-
Beijing, China), penicillin (100 U/ml) and streptomycin (100 mg/ cording to the manufacturer's instructions. The sequences of the
ml). Cells were incubated at 37  C with 5% CO2 in a humidified ERa-siRNA duplex were as follows: sense strand, 50 -GAGGGA-
atmosphere. All experiments were performed on logarithmically GAAUGUUGAAACATT-30 ; antisense strand, 50 -UGUUUCAACAUU-
growing cells. CUCCCUCTT-30 . The sequences of NC-siRNA duplex were: sense
strand, 50 -UUCUCCGAACGUGUCACGUTT-30 ; antisense strand, 50 -
2.3. Fluorescent microscopy of MDC staining ACGUGACACGUUCGGAGAATT-30 . The transfected cells were main-
tained for another 48 h before subsequent experiments.
Autofluorescent compound MDC stains acidic lysosomes and
thus it is widely used in studies of autophagy together with the 2.9. Western blot analysis
measurement of other parameters (9). The cells were seeded into
24-well cell culture plates (Corning, NY, USA) at a density of 2  104 After the indicated treatments, both adherent and floating cells
cells per well for 24 h. The cells in culture for another 24 h by the were collected at the predetermined time points and lysed with
treatments with different reagents were examined. The cells RIPA lysis buffer (Beyotime, Haimen, Jiangsu, China) supplemented
treated were rinsed twice with ice-cold PBS, stained with 0.05 mM with PMSF (1 mM) for 30 min. After centrifugation at 12,000  g for
MDC solution in the dark at 37  C for 30 min. They were observed 10 min, the supernatant was collected and the protein concentra-
with a fluorescence microscope (Olympus, Tokyo, Japan). tion was determined with the Bio-Rad protein assay reagent (Bio-
Rad, Hercules, CA, USA). The lysates that were adjusted with the
2.4. Flow cytometry of MDC positive cells total amount of proteins equal were separated on 10e13% SDS-
PAGE. The separated protein bands were transferred onto Milli-
Cells subjected to the indicated treatments were collected pore Immobilon®-P Transfer Membrane (Millipore Corporation,
(5  105 cells per sample), incubated with 0.05 mM MDC solution in Billerica, MA, USA). After blocking with 5% skimmed milk at room
the dark at 37  C for 30 min and analysed using a FACScan flow temperature for 2 h, the membranes were incubated with primary
cytometer (Becton Dickinson, Franklin Lakes, NJ, USA). antibodies at 4  C overnight and then with corresponding HRP
conjugated secondary antibodies at room temperature for 2 h. After
2.5. Cytotoxicity assay that, the blots were visualized using the SuperSignal West Pico
Chemiluminescent Substrate® purchased from Thermo Scientific
MCF-7 cells were seeded into 96-well cell culture plates (Rockford, IL, USA).
(Corning, NY, USA) at a density of 5  103 cells/well and cultured for
24 h. Then the cells were subjected to the indicated treatments for 2.10. Statistical analysis
24 h. The cells were rinsed twice with ice-cold PBS and incubated
with 100 mL of 0.5 mg/mL MTT solution at 37  C for 3 h. Then the All the data and results obtained by at least three independent
supernatant was discarded, and the residual cell layer was dis- experiments were expressed as mean ± S.D. Comparisons between
solved with 150 mL of DMSO. Thereafter, the optical absorbance (A groups were determined using Student's t-test. One-tailed p-values
value) was measured at 490 nm wavelength using a microplate are significant when p < 0.05.
100 N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107

Fig. 2. Treatment with a specific ERa antagonist MPP dihydrochloride (MPP) enhances the apoptosis of MCF-7 cells induced by silibinin. (A) Morphologic changes of apoptotic MCF-
7 cells were observed at 24 h (a, 200  magnification, bar ¼ 20 mm), Con: control, Sili: silibinin. (B) Flow cytometric analysis of apoptotic cell ratios after PI staining (SubG1 cells). (a)
The data are presented as the mean ± S.E.M. of the results for three independent experiments. (b) **p < 0.01 compared with silibinin-treated group. (C) The cells were incubated
with 200 mM silibinin with/without 10 mM MPP for 24 h. The rhodamine-123 fluorescent intensity was analysed by flow cytometry. #p < 0.05 versus control group, *p < 0.05 versus
silibinin group.
N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107 101

Fig. 3. Inhibition of ERa enhances the MCF-7 cell death induced by silibinin through extrinsic and mitochondrial-apoptosis pathways. The cells were incubated with or without
10 mM MPP for 1 h. Then 200 mM silibinin was added to the cells which were cultured for 24 h. Cell lysates were separated by 11% SDS-PAGE, and the levels of (A) caspase 8, bid, (B)
Bax (mitochondria), Bcl-2 (mitochondria), cytochrome c (cytoplasm), (D) caspase 6, PARP, ICAD were detected by Western blot analysis. Band density of the specific protein was
analyzed with Quantity One image software and the results were expressed as relative density to b-actin. (C) The expression of caspase 3 was detected in the MCF-7 cells treated
with silibinin and/or MPP. The expression in HepG2 cells was used as a positive control.
102 N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107

Fig. 4. Inhibition of ERa reinforces silibinin-induced MCF-7 cell autophagy. (A) The cells were incubated with or without 10 mM MPP for 1 h. Following the addition of 200 mM
silibinin, the cells were cultured for 24 h. Changes of fluoresce intensity were observed by fluorescence microscopy with MDC staining (  400 magnification, bar ¼ 20 mm). (B) MCF-
7 cells were cultured in the presence of 0, 100, 200 and 300 mM silibinin for 12 and 24 h. The MDC positive ratio was determined by flow cytometric analysis. (C) The positive ratio of
MDC staining was detected by flow cytometric analysis, n ¼ 3, mean ± S.E.M, **p < 0.01, compared with silibinin-treated group. (D) MCF-7 cells were lysed after the treatment with
100, 200 or 300 mM silibinin for 24 h, and the protein levels of p62 and LC3 were detected by western blot analysis. (E) The protein levels of p62 and LC3 were detected by western
blot analysis. Band density of the specific protein was analysed with Quantity One image software. (n ¼ 3, means ± SD; #p < 0.05 vs control group).
N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107 103

3. Results 3.3. Treatment with MPP enhances the cytotoxicity of silibinin


through both extrinsic and intrinsic-apoptosis pathways
3.1. ERa down-regulation was involved in silibinin-induced
cytotoxicity of MCF-7 cells Western blot assay was carried out to further determine the
features of silibinin and MPP-induced MCF-7 cell death. The
To test the cytotoxicity of silibinin on MCF-7 cells, the cells were treatment with silibinin resulted in that extrinsic apoptosis-related
cultured with 100e300 mM silibinin for 12, 24, 36 and 48 h. Silibinin protein pro-caspase-8 was cleaved into active caspase-8, as well as
was found to decrease the proportion of viable cells in a time and Bid was cleaved. MPP further augmented this effect (Fig. 3A). As for
dose-dependent manner with an IC50 (at 24 h) of 200 mM (Fig. 1B). the intrinsic apoptosis pathway, translocation of the anti-apoptotic
As a flavonoid, silibinin might affect the estrogen receptor signal- protein Bcl-2 to mitochondria was markedly decreased in the
ling pathways (5). Therefore, we investigated the levels of ERa. silibinin-treated group, while the pro-apoptotic translocation of
Western blot analysis showed that treatment with various con- Bax to mitochondria and the release of cytochrome c to cytoplasm
centrations of silibinin for 24 h reduced ERa expression in a were increased. MPP also augmentedthese processes furthermore
concentration-dependent manner (Fig. 1C). Fulvestrant (ICI (Fig. 3B). The results indicate that the intrinsic apoptosis pathway
182,780), an estrogen receptor antagonist which blocks both ERa was also upregulated.
and ERb signalling, was applied with silibinin to examine the effect Caspase 3 is the downstream executor of apoptosis in many
of estrogen receptor signalling pathway on MCF-7 cell growth. The cells. However, MCF-7 cells were reported to be negative in the
results showed that ICI 182,780 augmented silibinin-induced MCF- expression of caspase-3 (13,14). Accordingly, the expressions of
7 cell growth inhibition (Fig. 1D). To further elucidate the role of pro-caspase 3 and activated caspase 3 were not detected in
ERa, we examined the cytotoxicity of silibinin on two different silibinin-treated MCF-7 cells (Fig. 3C). It was reported that silibinin
strains of human breast cancer cells: one is MCF-7 cells with an
inherently high expression of ERa and the other is MDA-MB-231
cell with a rather low expression of ERa. MDA-MB-231cells are
often utilized as a negative control for the examination of ERa
involvement (10). Western blot analysis of the ERa levels of MCF-7
and MDA-MB-231 indicated, in accordance with the previous re-
ports, that the ERa expression in MCF-7 cells was high, but low in
MDA-MB-231 cells (Fig. 1E (a)).
MTT assay showed that the growth inhibitory effect of silibinin
on MCF-7 cells was higher than that on the MDA-MB-231 cells
(Fig. 1E (b)), evidencing the positive correlation between ERa
down-regulation and cytotoxicity. The specific ERa-antagonist,
MPP (11), dose-dependently augmented silibinin-induced MCF-7
cell growth inhibition (Fig. 1F), even to a larger extent than ICI
182,780 (Fig. 1D). Since high dose of MPP (20 mM) alone also
inhibited cell proliferation, we chose a lower dose (10 mM) of MPP
in the following experiments. To further confirm the cytotoxicity of
silibinin in combination with repression of ERa on MCF-7 cells, we
transfected the siRNA targeting ERa (si-ERa) into cells (Fig. 1G (a)).
We found that silencing of ERa markedly enhanced the cytotoxic
effect of silibinin (Fig. 1G (b)). Therefore, silibinin exerted anti-
cancer effects on MCF-7 cells through the inhibition of ERa
pathways.

3.2. Treatment with MPP enhances silibinin-induced apoptosis of


MCF-7 cells

To determine the features of MCF-7 cell growth inhibition, the


morphologic changes were examined. Obvious changes of cell
nuclei and cell number were observed in the cells treated with
200 mM silibinin plus 10 mM MPP for 24 h when compared with the
cells treated with silibinin alone, (Fig. 2A). PI staining was applied
to analysis of the changes of DNA contents: silibinin increased the
ratio of cells at subG1 phase in cell cycle at 24 h, while co-treatment
with MPP increased the ratio furthermore (Fig. 2B). These results
suggested that apoptosis might be involved in this growth inhibi-
tion. It was well documented that many anticancer drugs induced
apoptosis following mitochondrial dysfunctions (12). Therefore, we
examined intactness of mitochondrial membranes by rhodamine
123 staining. Decrease in rodamine-123 fluorescence intensity re-
flects the reduction in mitochondrial transmembrane potential Fig. 5. Autophagy promotes silibinin- and MPP-induced apoptosis. (A) The cells were
(Djm). Compared with silibinin-treated group, co-treatment with treated with 10 mM MPP for 1 h, and then 200 mM silibinin was added and cultured for
24 h in the presence or absence of rapamycin (10 nM) or 3 MA (2 mM), and the cell
MPP resulted in a further reduction in Djm (Fig. 2C), indicating that viability was measured by MTT assay, n ¼ 3, mean ± S.E.M, *p < 0.05, **p < 0.01,
inhibition of ERa markedly enhances the cytotoxicity of silibinin on compared with silibinin and MPP-treated group. (B) The protein levels of caspase 6,
MCF-7 cells through mitochondrial dysfunction. PARP and ICAD were detected by western blot analysis.
104 N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107

Fig. 6. Changes in the expressions of ERa downstream proteins in silibinin- and MPP-treated MCF-7 cells. (A) The cells were incubated with or without 10 mM MPP for 1 h, then
200 mM silibinin was added and cultured for 24 h, the levels of (A) GRB2, SOS, PI3K, AKT, mTOR, (B) MAPK-related proteins Raf, ERK, p-ERK were detected by Western blot analysis.
b-actin was used as an equal loading control. Band density of the specific protein was analyzed with Quantity One image software. (C) The cells were incubated with different
concentrations of PD98059 for 1 h. Following the addition of 200 mM silibinin, and the cells were cultured for 24 h. The viability was measured by MTT assay, n ¼ 3, mean ± S.E.M,
*p < 0.05, compared with silibinin-treated group. (E) The cells were treated with 10 mM MPP for 1 h. Following the addition of 200 mM silibinin, the cells were cultured for 24 h in the
presence or absence of PD98059 (30 mM). The cell viability was measured by MTT assay, n ¼ 3, mean ± S.E.M, *p < 0.05, compared with silibinin and MPP-treated group. (D) The
protein levels of p62 and LC3 and (F) EGFR were detected by western blot analysis.

activated caspase 3 in HepG2 cells (15). Therefore, silibinin-treated silibinin induced the activation of caspase 6. MPP treatment
HepG2 cells were used as a positive control for the detection of augmented the cleavage of pro-caspase-6 to caspase-6 further-
caspase 3 (Fig. 3C). We examined the expression of another com- more. The substrates of caspase-6, ICAD and PARP, were cleaved
mon downstream executor of apoptosis, caspase-6, and found that accordingly (Fig. 3D). In conclusion, MPP pre-treatment enhanced
N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107 105

both intrinsic and extrinsic apoptotic pathways in silibinin-treated As shown in Fig. 6B, treatment with MPP obviously reduced the
MCF-7 cells. p-ERK expression compared with silibinin alone group. To examine
the role of ERK, we pre-treated the cells with ERK inhibitor
3.4. Inhibition of ERa augmented silibinin-induced MCF-7 cell PD98059, and found that the cytotoxicity was obviously enhanced
autophagy by ERK inhibition (Fig. 6C and D). Since p62 degradation and from
LC3-I to LC3-II increased, we further confirmed significantly
Autophagy is a fundamental cellular pathway by which cyto- increased autophagy after PD98059 exposure (Fig. 6E). These
plasmic materials, including soluble macromolecules and organ- findings indicated that apoptotic action of silibinin on MCF-7 cells
elles, are delivered into lysosomes for degradation (15). Autophagy was mediated through ERa downstream RAS/ERK pathway. EGFR
plays an important role in the cellular responses towards stresses signaling also leads to inhibition of ERK1/2. Crosstalk between
such as starvation, hypoxia and DNA damage. The morphologic EGFR and ER signaling has been established and proved to have
changes were observed in the MDC-stained cells by fluorescence significant impact on complex biological processes such as cell
microscopy. Combined treatment with silibinin and MPP caused an growth and cell survival of breast cancer cells. To test the rela-
obvious increase in the number of MDC-labelled autophagolyso- tionship between down-regulation of ERa and EGFR, we examined
somes compared with the silibinin alone group (Fig. 4A). The the changes of expressed EGFR protein levels in MCF-7 cells treated
quantitative analysis of autophagy by MDC flow cytometric analysis with silibinin with/without MPP. We found that inhibition of ERa
showed the consistent results (Fig. 4B and C). Microtubule- with silibinin or/and MPP, the expression levels of EGFR was down-
associated protein light chain 3 (LC3) is now widely used to regulated as shown in (Fig. 6F). Thus, the effect of silibinin on ERK is
monitor autophagy. The levels of LC3 conversion (from cytosolic mediated by ERa that may have cross-talk with EGFR.
LC3-I to membrane-bound lipidated LC3-II) or the amounts of LC3-
II were examined by immunoblotting analysis (16). An alternative 4. Discussion
method for detecting the autophagic flux is the measurement of
p62 degradation, since p62 can bind LC3, thus serving as a selective Breast cancer is one of the most frequently diagnosed cancers
substrate of autophagy (16). Dose-dependent up-regulation of the and the leading cause of death by cancer among females world-
conversion from LC3-I to LC3-II and down-regulation of p62 caused wide. To develop breast cancer therapies, targeting of ERa, which is
by silibinin treatment were observed (Fig. 4D). Combined use of highly expressed in approximately 70% of all breast tumors (21), is
silibinin with MPP enhanced expressions of LC3-II and conversion taken as a targeting molecule.
from LC3-I to LC3-II significantly increased, while p62 amount was ERa is a member of the steroid receptor superfamily that reg-
further reduced (Fig. 4E). ulates processes such as growth and differentiation in various
target cells by affecting transcription. ERa not only plays an
important role in many human tissues and organs, such as mam-
3.5. Autophagy promoted apoptosis in MCF-7 cells by the co- mary gland, genital tract, and central nervous system (22), but also
treatment with silibinin and MPP it has a close correlation with gaining the peak bone mass and
maintaining the bone mass (23). Women who are menopausal or
To investigate the crosstalk of apoptosis and autophagy in sili- postmenopausal usually produce less ERa, leading to bone loss (24).
binin and MPP-treated MCF-7 cells, 3 MA, a specific autophagic However, in the majority of breast cancers, ERa is greatly up-
inhibitor, and rapamycin, an autophagic inducer, were employed. regulated than normal breast cells and its expression is a hall-
As shown in Fig. 5A, silibinin and MPP-induced cell growth inhi- mark of hormone-dependent tumor growth (25). In addition, the
bition was further augmented by rapamycin treatment but partially presence of elevated levels of ERa in benign breast epithelium
reversed by 3 MA. Western blot analysis showed that the activation appears to indicate an increased risk of breast cancer, suggesting a
of caspase-6 and the cleavage of caspase substrates, PARP and ICAD, role for ERa in breast cancer initiation, as well as progression (26).
caused by cotreatment with MPP and silibinin were reversed by Silibinin, which has been used as a liver protectant, induced
3 MA treatment (Fig. 5B). These findings indicated that the auto- apoptosis in human breast cancer MCF-7 cells (27). In this study, we
phagy induced by silibinin and MPP co-treatment intensified the report the evidence for the mechanism of silibinin-induced
apoptotic process in MCF-7 cells. apoptosis in that downregulation of ERa expression in the MCF-7
cells exposed to silibinin underlies. Consistent with previous re-
3.6. Combination of silibinin and MPP treatments repressed the ports, silimarin, which contains silibinin as a major component, has
downstream signals of ERa been used in the clinical treatment of hepatic diseases and phar-
macologic studies have indicated that silymarin is not toxic even at
The pathways through Shc-Grb2-Sos (Src homology-growth high doses. Besides, pretreatment with silibinin 500 mM signifi-
factor receptor binding protein 2-son of sevenless) for activation cantly inhibited UV-induced apoptosis in HaCaT cells after 9 h in-
of MAPK and are involved in the downsteam of activated ERa (17). cubation (28). Therefore we believe that silibinin appears to be
One of them is RAS/ERK pathway, which can induce oncogenic gene toxic for cancer cells, but relatively safe for normal ones. ERa re-
expression programs, being often activated in cancer cells. Another ceptors are known to exist in cytosol and also as membrane bound-
is PI3K/Akt pathway which is related with the mammalian target of ER. However, in MCF-7 cells transfected with GFP-ERa, the fluo-
rapamycin (mTOR). Hou X. and co-workers reported that ERa raised rescence of GFP-ERa was observed only in the nucleus (29).
the phosphorylation levels of PI3K, Akt and mTOR in endometrial Therefore, it seemed that ERa receptors are mainly found in
carcinoma cells (18). We examined the activation of these two ER nucleus.
downstream signaling pathways. The mechanism we propose here is supported by the finding
Western blot analysis showed that PI3K/Akt/mTOR pathway that ERa-specific antagonist, MPP, combined with silibinin
was down-regulated by either silibinin alone or silibinin and MPP enhanced all the effects of silibinin on MCF-7 cells. The enhance-
co-treatment (Fig. 6A). As has been demonstrated in many studies, ment with MPP can be interpreted as inhibition of the activity of
inhibition of mTOR promotes autophagy (19,20). Therefore, silibi- ERa remaining after downregulation with silibinin. Apoptosis is
nin enhanced autophagy by down-regulating ERa downstream characterized by the sequential activation of caspase cascades.
PI3K/Akt/mTOR pathway. Caspase 8 is believed to be one of the initiator caspases that can
106 N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107

activate downstream caspase-3 (30). However, MCF-7 cells do not positive ERb as a negative control. MTT assay showed that the
express caspase-3 (13,14). Instead, upregulation of the caspase-6 growth inhibitory effect of silibinin on MCF-7 cells was higher than
expression was obtained for the MCF-7 cells treated with silibinin that on the MDA-MB-231 cells (Fig. 1E (b)), evidencing the positive
in accordance with the report by Kagawa S et al., showing that Bax- correlation between ERa down-regulation and cytotoxicity. We
induced levels of caspase-6 activation could be used as an evalua- assume that silibinin also dose-dependently induced cytotoxicity in
tion index of apoptosis when the cells did not express caspase-3 MDA-MB231 cells and ERb expression in MDA-MB231 cells was
(31). increased. In order to prove this hypothesis, further experiments
A number of studies showed relationship between ERa are necessary. Thus, ERb could be another key mechanism of
expression and levels of apoptosis related proteins including Bcl-2 silibinin-induced apoptosis and autophagy in many cell lines as
and Bax (32e34). The relationships between ERa and apoptosis well as MCF-7 cell. Therefore, we are going to use ERb as a target
pathways in silibinin-treated cells observed in the present study object for future research.
indicate that death rate of MCF-7 cells induced by silibinin through
the down-regulation of ERa involves both the extrinsic and
intrinsic apoptosis pathways. Conflicts of interest
Estradiol-bound ERa interacts with various signalling molecules
such as PI3K (35) and MAP kinases (36). The PI3K/Akt and MEK/ERK The authors declare that there are no conflicts of interest.
signalling pathways are important downstream pathways of ERa.
The observations in the present study that silibinin reduced the References
phosphorylation levels of both ERK and Akt and that additional
treatment with a specific inhibitor of ERK enhanced the cytotoxic (1) Acconcia F, Totta P, Ogawa S, Cardillo I, Inoue S, Leone S, et al. Survival versus
effects on MCF-7 cells treated with silibinin and MPP are rationally apoptotic 17beta-estradiol effect: role of ER alpha and ER beta activated non-
genomic signaling. J Cell Physiol. 2005;203:193e201.
interpreted, since they are downstream signaling pathways of ERa. (2) Saji S, Okumura N, Eguchi H, Nakashima S, Suzuki A, Toi M, et al. MDM2
Our results are in accordance with the results of Melyssa and co- enhances the function of estrogen receptor alpha in human breast cancer
workers, who reported that MCF-7 breast carcinoma cells pro- cells. Biochem Biophys Res Commun. 2001;281:259e265.
(3) Dehmlow C, Erhard J, de Groot H. Inhibition of Kupffer cell functions as an
moted survival through activation of PI3K/Akt crosstalk pathway explanation for the hepatoprotective properties of silibinin. Hepatology.
(37). We therefore conclude that silibinin shows cytotoxic effect 1996;23:749e754.
against MCF-7 cells through the down-regulation of ERa signalling (4) Gandara L, Sandes E, Di Venosa G, Prack Mc Cormick B, Rodriguez L,
Mamone L, et al. The natural flavonoid silybin improves the response to
pathway.
Photodynamic Therapy of bladder cancer cells. J Photochem Photobiol B.
Physiological levels of autophagy are essential for normal 2014;133:55e64.
cellular homeostasis. The absence of autophagy increases cell death (5) Sak K. Cytotoxicity of dietary flavonoids on different human cancer types.
during metabolic stress and on treatment with cytotoxic chemo- Pharmacogn Rev. 2014;8:122e146.
(6) Millimouno FM, Dong J, Yang L, Li J, Li X. Targeting apoptosis pathways in
therapeutic agents. By contrast, excessive levels of autophagy cancer and perspectives with natural compounds from mother nature. Cancer
promotes cell death (38). It is postulated that autophagic cell death Prev Res (Phila). 2014;7(11):1081e1107.
induced by some anticancer agents underlines their potential as a (7) Yadegarynia S, Pham A, Ng A, Nguyen D, Lialiutska T, Bortolazzo A, et al.
Profiling flavonoid cytotoxicity in human breast cancer cell lines: determi-
new cancer therapy modality (39). MPP in combination with sili- nation of structure-function relationships. Nat Prod Commun. 2014;9:
binin activates autophagy which enhances apoptosis in MCF-7 cells. 597e606.
The results are consistent with the notions in several studies in that (8) Martinez-Perez C, Ward C, Cook G, Mullen P, McPhail D, Harrison DJ, et al.
Novel flavonoids as anti-cancer agents: mechanisms of action and promise for
autophagy is triggered in some cancers in response to various their potential application in breast cancer. Biochem Soc Trans. 2014;42:
anticancer agents, including As2O3, tamoxifen and temozolomide 1017e1023.
(40). (9) Mizushima N. Methods for monitoring autophagy. Int J Biochem Cell Biol.
2004;36:2491e2502.
In conclusion, the present study demonstrates that down-
(10) Wang X, Kilgore MW. Signal cross-talk between estrogen receptor alpha and
regulation of ERa expression by the treatment with silibinin of beta and the peroxisome proliferator-activated receptor gamma1 in MDA-MB-
MCF-7 cells plays a key role in leading the cells to follow ERa-MEK/ 231 and MCF-7 breast cancer cells. Mol Cell Endocrinol. 2002;194:123e133.
ERK and ERa-Akt/mTOR pathways to apoptosis. ERa inhibition with (11) Sun J, Huang YR, Harrington WR, Sheng S, Katzenellenbogen JA,
Katzenellenbogen BS. Antagonists selective for estrogen receptor a. Endocri-
ERa-specific antagonist, MPP, augments autophagy and apoptosis nology. 2002;143:941e947.
by extrinsic-apoptosis and mitochondrial-apoptosis pathways. (12) Chen G, Wang F, Trachootham D, Huang P. Preferential killing of cancer cells
Autophagy plays a pro-apoptotic role with the detailed mechanism with mitochondrial dysfunction by natural compounds. Mitochondrion.
2010;10:614e625.
remaining to be cleared. €nicke RU. MCF-7 breast carcinoma cells do not express caspase-3. Breast
(13) Ja
In the present study, we highlight just ERa signalling for the Cancer Res Treat. 2009;117:219e221.
silibinin effect. However, it is not known whether the antitumor €nicke RU, Sprengart ML, Wati MR, Porter AG. Caspase-3 is required for DNA
(14) Ja
fragmentation and morphological changes associated with apoptosis. J Biol
activity of ERb is involved. Estrogen receptors ERa and ERb share Chem. 1998;273:9357e9360.
considerable sequence homology yet exert opposite effects on (15) Zhang S, Yang Y, Liang Z, Duan W, Yang J, Yan J, et al. Silybin-mediated in-
breast cancer cell proliferation. Bin et al. illustrated that modulation hibition of Notch signaling exerts antitumor activity in human hepatocellular
carcinoma cells. PLoS One. 2013;8:e83699.
of ERb-specific antitumor activity can be considered as a molecular (16) Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy.
strategy for cancer therapy (41). It is suggested that the mecha- 2007;3:542.
nisms of silibinin's toxic effect on MCF-7 cells may also be associ- (17) Song RX-D, McPherson RA, Adam L, Bao Y, Shupnik M, Kumar R, et al. Linkage
of rapid estrogen action to MAPK activation by ERa-Shc association and Shc
ated with ERb. Up-regulation of ERb is another mechanism of toxic pathway activation. Mol Endocrinol. 2002;16:116e127.
effect on cancer cells. Studies also have shown that ERb exerts (18) Hou X, Zhao M, Wang T, Zhang G. Upregulation of estrogen receptor mediates
antiproliferative effects in ERa expressing MCF7 cells, probably by migration, invasion and proliferation of endometrial carcinoma cells by
initiating degradation of ERa. Meanwhile, ERb-selective agonist regulating the PI3K/AKT/mTOR pathway. Oncol Rep. 2014;31:1175e1182.
(19) Noack M, Richter-Landsberg C. Activation of autophagy by rapamycin does
DPN inhibited cell growth and induced apoptosis. In addition, there not protect oligodendrocytes against protein aggregate formation and cell
are reports which show that the proliferating MCF-7 cells express death induced by proteasomal inhibition. J Mol Neurosci. 2014;55:99e108.
both ERa and ERb. In order to examine whether the cytotoxic effect (20) Bove J, Martinez-Vicente M, Vila M. Fighting neurodegeneration with rapa-
mycin: mechanistic insights. Nat Rev Neurosci. 2011;12:437e452.
of silibinin is related with ERa, we performed MTT experiments (21) Moerkens M, Zhang Y, Wester L, van de Water B, Meerman JH. Epidermal
using MDA-MB231 cells that have low expression of ERa and growth factor receptor signalling in human breast cancer cells operates
N. Zheng et al. / Journal of Pharmacological Sciences 128 (2015) 97e107 107

parallel to estrogen receptor alpha signalling and results in tamoxifen (31) Kagawa S, Gu J, Honda T, McDonnell TJ, Swisher SG, Roth JA, et al. Deficiency
insensitive proliferation. BMC Cancer. 2014;14:283. of caspase-3 in MCF7 cells blocks Bax-mediated nuclear fragmentation but
(22) Kininis M, Chen BS, Diehl AG, Isaacs GD, Zhang T, Siepel AC, et al. Genomic not cell death. Clin Cancer Res. 2001;7:1474e1480.
analyses of transcription factor binding, histone acetylation, and gene (32) Wang TT, Phang JM. Effects of estrogen on apoptotic pathways in human
expression reveal mechanistically distinct classes of estrogen-regulated pro- breast cancer cell line MCF-7. Cancer Res. 1995;55:2487e2489.
moters. Mol Cell Biol. 2007;27:5090e5104. (33) Jiang Z, Guo J, Shen J, Jin M, Xie S, Wang L. The role of estrogen receptor alpha
(23) Nakamura T, Imai Y, Matsumoto T, Sato S, Takeuchi K, Igarashi K, et al. Es- in mediating chemoresistance in breast cancer cells. J Exp Clin Cancer Res.
trogen prevents bone loss via estrogen receptor alpha and induction of Fas 2012;31:42.
ligand in osteoclasts. Cell.. 2007;130:811e823. (34) Tabuchi Y, Matsuoka J, Gunduz M, Imada T, Ono R, Ito M, et al. Resistance to
(24) Styrkarsdottir U, Halldorsson BV, Gretarsdottir S, Gudbjartsson DF, paclitaxel therapy is related with Bcl-2 expression through an estrogen re-
Walters GB, Ingvarsson T, et al. Multiple genetic loci for bone mineral density ceptor mediated pathway in breast cancer. Int J Oncol. 2009;34:313e319.
and fractures. N Engl J Med. 2008;358:2355e2365. (35) Sun M, Paciga JE, Feldman RI, Z Yuan, Coppola D, Lu YY, et al. Phosphatidy-
(25) Williams C, Lin CY. Oestrogen receptors in breast cancer: basic mechanisms linositol-3-OH kinase (PI3K)/AKT2, activated in breast cancer, regulates and is
and clinical implications. Ecancermedicalscience. 2013;7:370. induced by estrogen receptor a (ERa) via interaction between ERa and PI3K.
(26) Ali S, Coombes RC. Estrogen receptor alpha in human breast cancer: occur- Cancer Res. 2001;61:5985e5991.
rence and significance. J Mammary Gland Biol Neoplasia. 2000;5:271e281. (36) Chakrabarti S, Morton JS, Davidge ST. Mechanisms of estrogen effects on the
(27) Wang HJ, Tashiro S, Onodera S, Ikejima T. Inhibition of insulin-like growth endothelium: an overview. Can J Cardiol. 2014;30:705e712.
factor 1 receptor signaling enhanced silibinin-induced activation of death (37) Bratton MR, Duong BN, Elliott S, Weldon CB, Beckman BS, McLachlan JA, et al.
receptor and mitochondrial apoptotic pathways in human breast cancer MCF- Regulation of ERalpha-mediated transcription of Bcl-2 by PI3K-AKT crosstalk:
7 cells. J Pharmacol Sci. 2008;107:260e269. implications for breast cancer cell survival. Int J Oncol. 2010;37:541e550.
(28) Li LH, Wu LJ, Tashiro S, Onodera S, Uchiumi F, Ikejima T. Silibinin prevents UV- (38) Levine B. Cell biology: autophagy and cancer. Nature. 2007;446:745e747.
induced HaCaT cell apoptosis partly through inhibition of caspase-8 pathway. (39) Gozuacik D, Kimchi A. Autophagy as a cell death and tumor suppressor
Biol Pharm Bull. 2006;29(6):1096e1101. mechanism. Oncogene. 2004;23:2891e2906.
(29) Takahashi T, Ohmichi M, Kawagoe J, Ohshima C, Doshida M, Ohta T, et al. (40) Choi S, Lim MH, Kim KM, Jeon BH, Song WO, Kim TW. Cordycepin-induced
Growth factors change nuclear distribution of estrogen receptor-alpha via apoptosis and autophagy in breast cancer cells are independent of the es-
mitogen-activated protein kinase or phosphatidylinositol 3-kinase cascade in trogen receptor. Toxicol Appl Pharmacol. 2011;257:165e173.
a human breast cancer cell line. Endocrinology. 2005;146(9):4082e4089. (41) Yuan B, Cheng L, Chiang HC, Xu X, Han Y, Su H, et al. A phosphotyrosine switch
(30) Chen M, Wang J. Initiator caspases in apoptosis signaling pathways. Apoptosis. determines the antitumor activity of ERbeta. J Clin Invest. 2014;124:
2002;7:313e319. 3378e3390.

Você também pode gostar