2-hydroxycinnamaldehyde inhibits the epithelial-mesenchymal transition in breast cancer cells

12
PRECLINICAL STUDY 2-Hydroxycinnamaldehyde inhibits the epithelial-mesenchymal transition in breast cancer cells Ismail Ahmed Ismail Hye Sook Kang Heon-Jin Lee Hyeyoun Chang Jieun Yun Chang Woo Lee Nam Hee Kim Hyun Sil Kim Jong In Yook Su-Hyung Hong Byoung-Mog Kwon Received: 31 August 2012 / Accepted: 13 December 2012 / Published online: 3 January 2013 Ó Springer Science+Business Media New York 2012 Abstract Since epithelial-mesenchymal transition (EMT) plays a critical role in cancer progression and in maintaining cancer stem cell properties, EMT is emerging as a thera- peutic target for inhibiting the metastatic progression of cancer cells. 2 0 -Hydroxycinnamaldehyde (HCA) and its derivative, 2 0 -benzoyloxycinnamaldehyde, have recently been suggested as promising therapeutic candidates for cancer treatment. The purpose of this study is to investigate the anti-metastatic effect of HCA on breast cancer and the molecular mechanisms by which HCA regulates the transcriptional program during EMT. HCA induces epithe- lial reversion at nanomolar concentrations by suppressing Snail via the nuclear translocalization of GSK-3b, which results in the transcriptional upregulation of E-cadherin. HCA also activates the transcription factor KLF17, which suppresses Id-1, indicating that HCA inhibits EMT by mul- tiple transcriptional programs. Further, HCA treatment sig- nificantly inhibits lung metastasis in a mouse orthotopic breast cancer model. This study demonstrates the anti-met- astatic effect of the non-toxic natural compound HCA through attenuation of EMT in a breast cancer model. Keywords 2 0 -Hydroxycinnamaldehyde Breast cancer cells Epithelial-mesenchymal transition (EMT) Cell invasion Snail KLF17 Introduction Cinnamaldehyde is commonly used as a flavoring and ingredient in food, beverages, medical products, cosmetics, and perfumes. 2 0 -Hydroxycinnamaeldehyde (HCA) is a natural compound that is isolated from the bark of Cinna- momum cassia Blume [1], and 2 0 -benzoyloxycinnamalde- hyde (BCA) is a well-known derivative of HCA. Both HCA and BCA have been reported to have anti-tumor effects on various types of cancer cells, inhibiting proliferation and inducing apoptosis [25]. BCA was approved for clinical tests by the Korean Food and Drug Administration on Jan- uary 31, 2011. Although the cytotoxic or anti-angiogenic effects of these cinnamaldehydes are exerted mainly at lm to mM concentrations [2], the systemic administration of cinnamaldehyde significantly suppressed tumor forma- tion in vivo [5], suggesting that these compounds func- tion to suppress tumorigenesis at physiological levels. I. A. Ismail and H. S. Kang contributed equally to this work. Electronic supplementary material The online version of this article (doi:10.1007/s10549-012-2388-7) contains supplementary material, which is available to authorized users. I. A. Ismail H. S. Kang H.-J. Lee S.-H. Hong (&) Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea e-mail: [email protected] I. A. Ismail Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Assiut 71516, Egypt H. Chang B.-M. Kwon (&) Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology, Daejon 305-806, Republic of Korea e-mail: [email protected] J. Yun C. W. Lee Bio-Evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Republic of Korea N. H. Kim H. S. Kim J. I. Yook Department of Oral Pathology, Oral Cancer Research Institute, College of Dentistry, Yonsei University, Seoul 120-752, Republic of Korea 123 Breast Cancer Res Treat (2013) 137:697–708 DOI 10.1007/s10549-012-2388-7

Upload: independent

Post on 27-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

PRECLINICAL STUDY

2-Hydroxycinnamaldehyde inhibits the epithelial-mesenchymaltransition in breast cancer cells

Ismail Ahmed Ismail • Hye Sook Kang • Heon-Jin Lee • Hyeyoun Chang •

Jieun Yun • Chang Woo Lee • Nam Hee Kim • Hyun Sil Kim • Jong In Yook •

Su-Hyung Hong • Byoung-Mog Kwon

Received: 31 August 2012 / Accepted: 13 December 2012 / Published online: 3 January 2013

� Springer Science+Business Media New York 2012

Abstract Since epithelial-mesenchymal transition (EMT)

plays a critical role in cancer progression and in maintaining

cancer stem cell properties, EMT is emerging as a thera-

peutic target for inhibiting the metastatic progression of

cancer cells. 20-Hydroxycinnamaldehyde (HCA) and its

derivative, 20-benzoyloxycinnamaldehyde, have recently

been suggested as promising therapeutic candidates for

cancer treatment. The purpose of this study is to investigate

the anti-metastatic effect of HCA on breast cancer and

the molecular mechanisms by which HCA regulates the

transcriptional program during EMT. HCA induces epithe-

lial reversion at nanomolar concentrations by suppressing

Snail via the nuclear translocalization of GSK-3b, which

results in the transcriptional upregulation of E-cadherin.

HCA also activates the transcription factor KLF17, which

suppresses Id-1, indicating that HCA inhibits EMT by mul-

tiple transcriptional programs. Further, HCA treatment sig-

nificantly inhibits lung metastasis in a mouse orthotopic

breast cancer model. This study demonstrates the anti-met-

astatic effect of the non-toxic natural compound HCA

through attenuation of EMT in a breast cancer model.

Keywords 20-Hydroxycinnamaldehyde � Breast cancer

cells � Epithelial-mesenchymal transition (EMT) � Cell

invasion � Snail � KLF17

Introduction

Cinnamaldehyde is commonly used as a flavoring and

ingredient in food, beverages, medical products, cosmetics,

and perfumes. 20-Hydroxycinnamaeldehyde (HCA) is a

natural compound that is isolated from the bark of Cinna-

momum cassia Blume [1], and 20-benzoyloxycinnamalde-

hyde (BCA) is a well-known derivative of HCA. Both HCA

and BCA have been reported to have anti-tumor effects on

various types of cancer cells, inhibiting proliferation and

inducing apoptosis [2–5]. BCA was approved for clinical

tests by the Korean Food and Drug Administration on Jan-

uary 31, 2011. Although the cytotoxic or anti-angiogenic

effects of these cinnamaldehydes are exerted mainly at lm

to mM concentrations [2], the systemic administration

of cinnamaldehyde significantly suppressed tumor forma-

tion in vivo [5], suggesting that these compounds func-

tion to suppress tumorigenesis at physiological levels.

I. A. Ismail and H. S. Kang contributed equally to this work.

Electronic supplementary material The online version of thisarticle (doi:10.1007/s10549-012-2388-7) contains supplementarymaterial, which is available to authorized users.

I. A. Ismail � H. S. Kang � H.-J. Lee � S.-H. Hong (&)

Department of Oral Microbiology, School of Dentistry,

Kyungpook National University, Daegu, Republic of Korea

e-mail: [email protected]

I. A. Ismail

Laboratory of Molecular Cell Biology, Department of Zoology,

Faculty of Science, Assiut University, Assiut 71516, Egypt

H. Chang � B.-M. Kwon (&)

Laboratory of Chemical Biology and Genomics, Korea Research

Institute of Bioscience and Biotechnology, University of Science

and Technology, Daejon 305-806, Republic of Korea

e-mail: [email protected]

J. Yun � C. W. Lee

Bio-Evaluation Center, Korea Research Institute of Bioscience

and Biotechnology, Chungbuk, Republic of Korea

N. H. Kim � H. S. Kim � J. I. Yook

Department of Oral Pathology, Oral Cancer Research Institute,

College of Dentistry, Yonsei University, Seoul 120-752,

Republic of Korea

123

Breast Cancer Res Treat (2013) 137:697–708

DOI 10.1007/s10549-012-2388-7

However, the molecular mechanisms underlying the in vivo

effects of these compounds have not been clearly identified.

The pharmacokinetics and metabolism of HCA and BCA

were characterized in male Sprague–Dawley rats as part of a

preclinical evaluation. BCA was converted rapidly to HCA

in the rat serum after either intravenous or oral uptake; HCA

was subsequently converted to o-coumaric acid. The half-

lives of both HCA and BCA were *2 h [6].

Cancer metastases, rather than primary tumors, are

responsible for the most cancer deaths [7–9]. Epithelial-

mesenchymal transition (EMT) is a cellular program in which

polarized epithelial cells undergo complex biological changes

such that the epithelial cells express a mesenchymal pheno-

type, which induces enhanced migratory capacity, invasive-

ness, metastatic potential, and drug resistance [10]. Among

the number of molecular processes involved in EMT,

including the activation of transcription factors, regulation of

cellular adhesion, rearrangement of cytoskeletal proteins, and

changes in the expression of specific microRNAs [11], the

transcriptional programs of EMT are key targets for thera-

peutic intervention with natural compounds as well as for an

understanding of coordinated cellular program of EMT. For

example, members of the Snail superfamily of zinc finger

transcription factors bind directly to the proximal promoter

region of E-cadherin and induce an EMT phenotype in cancer

cells [12]. Interestingly, Snail expression is controlled by

GSK-3b-mediated phosphorylation, which is governed by

canonical Wnt signaling, allowing the coordinated transcrip-

tional regulation of b-catenin and Snail [13]. Importantly, the

p53 tumor suppressor directly regulates the Snail-mediated

EMT program via post-translational and post-transcriptional

mechanisms, suggesting that Snail may be an attractive ther-

apeutic target [14, 15] for the control of cancer metastasis.

The transcription factor Sp1 increases breast cancer cell

invasion and metastasis via the upregulation of urokinase

receptor or matrix metalloproteinase-2 expression [16, 17].

Furthermore, Sp1 binds to and activates Id-1 (inhibitor of

differentiation or DNA binding protein 1) promoter [18]. Id-

1 is overexpressed in highly invasive cancer cells, including

prostate [19], breast [20], cervical [21], and bladder cancers

[22]. Id-1 promotes metastasis in human breast cancer

in vivo via the upregulation of matrix metalloproteinase

MT1-MMP [20]. In human esophageal cancer, Id-1 activates

the PI3 K/AKT signaling pathway [23] or the N-cadherin-

RhoA axis [24], either of which can result in increased cancer

metastasis. Interestingly, KLF17 (Kruppel-like transcription

factor 17) is able to negatively regulate EMT and cell

invasion by directly binding to the Id-1 promoter to inhibit its

transcription in breast cancer cells [25].

Recently, the targeting of cancer stem cells has emerged

as a therapeutic strategy. EMT induced by Snail or TGF-bgenerates stem cell phenotype; the transformed cells exhibit

an increased ability to form mammospheres and resistance to

chemotherapy [26]. Selective inhibitors of cancer stem cells

have been identified through screening for compounds that

selectively kill mesenchymally transformed cells [27].

Although this has been a conceptual advance for cancer

therapeutics, several chemicals that target EMT, such as

salinomycin and etoposide, are of limited potential due to

systemic toxicity in mammals. Thus, natural compounds

with anti-EMT potential but without systemic adverse

effects may provide advanced therapeutic advantages. In this

study, we report that the natural food compound HCA blocks

a number of EMT transcriptional programs in breast cancer

cells, resulting in epithelial reversion. Specifically, we

examined the anti-EMT effect of HCA at non-toxic nano-

molar concentrations to explain the functional relevance of

anti-metastatic potential in vivo.

Materials and methods

Chemicals and reagents

MTT (3-[4, 5-dimethyl-2-thiazolyl]-2,5-diphenyl-2H-tetra-

zolium bromide) was purchased from Sigma (St. Louis, MO,

USA). DMEM, RPMI, FBS, and penicillin/streptomycin

antibiotics were purchased from Gibco (Invitrogen, CA,

USA). Qiazol was purchased from Qiagen (Valencia, CA,

USA), and 29 SYBR Green PCR master mix was purchased

from Takara Biotechnology (Dalian, Japan). The CytoSe-

lectTM 96-well cell invasion assay kit was purchased from

Cell Biolabs (San Diego, CA, USA). Rabbit polyclonal anti-

GSK3-b, anti-Snail, anti-KLF17, anti-Sp1, and rabbit

monoclonal anti-E-cadherin antibodies were purchased from

Abcam (Cambridge, UK). The mouse monoclonal anti-

vimentin antibody and anti-Id-1 antibody were purchased

from Lab Vision (Fremont, CA, USA) and Millipore

(Upstate Chemicon, Temecula, CA, USA), respectively.

Mouse monoclonal anti-HDAC1 and HRP-conjugated

mouse monoclonal IgG anti-b-actin antibody were pur-

chased from Santa Cruz Biotechnology (Santa Cruz, CA,

USA). HRP-conjugated goat anti-rabbit and anti-mouse

secondary antibodies were purchased from Pierce (Rock-

ford, IL, USA). Alexa Fluor goat anti-mouse and anti-rabbit

IgG fluorescent secondary antibodies were purchased from

Molecular Probes (Eugene, Oregon, USA). ECL Western

Blotting Detection Reagent was purchased from Neuronex

(Daegu, South Korea). All other reagents were obtained from

standard commercial sources.

Cell culture

The human breast cancer cell lines MCF-7, T47D, MDA-

MB-231, and MDA-MB-435 were obtained from the

American Type Culture Collection and maintained in

698 Breast Cancer Res Treat (2013) 137:697–708

123

DMEM (MCF-7 and MDA-MB-231) or RPMI (T47D and

MDA-MB-435) media containing 10 % FBS and

1 % penicillin/streptomycin solution. Cells were grown at

37 �C in a humidified atmosphere containing 5 % CO2.

Cell viability assay

Cells were seeded into 96-well plates (at a density of

5,000 cells/well). On the following day, cells were treated

with different concentrations of HCA (0–5 lm) in fresh

medium and incubated for another 24 h. Cell viability was

then assessed using the MTT assay, and the absorbance

was read at 570 nM using an ELISA microplate reader

(Molecular Devices, Downingtown, PA, USA).

Transwell migration assay

The effect of HCA on cell invasion was determined using a

CytoSelectTM 96-well cell invasion assay kit (Cell Biolabs, San

Diego, CA, USA) containing polycarbonate membrane inserts

(8 lm pore size) as indicated in the instruction manual and

previously described methods [28]. The invasion plate was

warmed for 10 min at room temperature. The basement

membrane layer was then rehydrated by adding 100 ll of

warm, serum-free medium to the inner compartment and

incubated for 1 h in a cell culture incubator. A cell suspension

(1 9 106 cells/ml) in serum-free medium (with or without

HCA) was cultured on the basement membrane after the

rehydration media was removed. A 150 ll aliquot of serum-

containing medium was added to the feeder tray. The basement

membrane chamber was inserted carefully to avoid air bubbles,

followed by the addition of 100 ll of cell suspension containing

the indicated doses of HCA. After 24 h of incubation, the

medium in the membrane chamber was transferred to a new

harvesting tray containing 150 ll of detachment solution for

30 min. The cells were dislodged completely from the under-

side of the membrane by gently tilting the membrane several

times. Then, 50 ll of 49 lysis buffer/CyQuant GR dye solution

was added to each sample and incubated for 20 min at room

temperature. Fluorescence measurements were performed in a

fluorescence plate reader at 480/520 nM.

Wound healing assay

The effect of HCA on breast cancer cell migration was

assessed using the wound healing assay as previously

described [29]. Briefly, cells were seeded in 100 mM cul-

ture plates and incubated for 16–24 h. When the cell

confluency reached approximately 80–90 %, a wound was

created manually by scratching the cell layer with a pipette

tip. Then, the cell medium was exchanged with new

medium containing either DMSO or 100 nM HCA. The

images were photographed immediately using phase-

contrast microscopy for the 0 h time point (control). After

24 h of incubation, the wounds were visualized and the

healed wound was compared with the control.

E-cadherin luciferase reporter assay

The E-cadherin reporter gene constructs E-cad (-108)-Luc

and E-cad (-108)-39Ebox. Mut-Luc and the pSV-gal (Pro-

mega) control vector have been described previously [30].

Cells were transfected with 0.75 lg E-cad (-108)-Luc or

E-cad (-108)-3xMut-Luc vector. Reporter gene activities

were measured with a luciferase assay system (Promega) at

48 h after transfection and normalized to the b-galactosidase

activities of co-transfected pSV-gal (0.25 g) measured with

a b-galactosidase enzyme assay system (Promega). Reporter

gene activities were reported as light units relative to those

obtained from mock (pCR3.1, Invitrogen)-transfected cells.

Quantitative real-time PCR

The effect of HCA (100 nM) on the expression of a panel of

genes involved in metastasis and EMT regulation in breast

cancer cells was investigated using real-time quantitative RT-

PCR analysis. In brief, 1 9 105 cells were plated in 60 mM

dishes and grown for 24 h, followed by treatment with the

indicated doses of HCA for 24 h. Total RNA was extracted

with Qiazol (Qiagen, Valencia, CA, USA) according to the

manufacturer’s instructions. Extracted RNA (5 lg) was

reverse transcribed into cDNA using a first-strand cDNA

synthesis kit (Applied Biosystems, Foster City, CA, USA),

and the resulting cDNA was diluted tenfold and kept at

-20 �C until use. The real-time qPCR primers were designed

using the Primer Express 1.5 software (Applied Biosystems)

as follows: E-cadherin forward, 50-CGACCCAACCCAA

GAATCTA-30; E-cadherin reverse, 50-CTCCAAGAATCC

CCAGAATG-30; Snail forward, 50-AGCTCTCTGAGGCCA

AG-GA TCT-30; Snail reverse, 50-TGTGGCTTCGGATGTG

CAT-30; KLF17 forward, 50-CTGCCTGAGCGTGGTATG

AG-30; KLF17 reverse, 50-TCATCCGGGAAGGAGTGA-G

A-30; Sp1 forward, 50-GGACTACCTGGAGTGATGCCT

AA-30; Sp1 reverse, 50-CCCATCAACGGTCTGGAACT-30;Id-1 forward, 50-CTCTACGACATGAACGGCTG -30; Id-1

reverse 50-TGCTCACCTTGCGGTTCTG-30; GAPDH

forward, 50-AGATCATCAGCAATGCCTCCTG-30 and GAP-

DH reverse, 50-ATGGCATGGACTGTGGTCATG-30. The

expressions of these genes were normalized to GAPDH. Real-

time qPCR was carried out using an ABI Prism 7500 sequence

detection system (Applied Biosystems, USA). Each 20 ll PCR

reaction contained 10 ll SYBR Green PCR master mix, 4 ll

diluted cDNA, and 200 nM primer set. All samples were

amplified in triplicate in a 96-well plate using the following

cycling conditions: 2 min at 50 �C, 10 min at 95 �C, and 40

cycles at 95 �C for 15 s followed by 1 min at 60 �C.

Breast Cancer Res Treat (2013) 137:697–708 699

123

Calculations were performed using the Dcycle threshold (DCt)

method, normalizing the average Ct value of each treatment

compared to its endogenous control (GAPDH), and then calcu-

lating the 2-DDCt for each treatment. Statistical analysis was

performed as described previously [31]. These experiments were

each repeated three times.

Western blot analyses

Cells were washed twice with cold PBS, after which

200 ll of PRO-PREP protein extraction solution (Intron,

Daejon, South Korea) was added. The cell lysates were

centrifuged, and protein concentrations were estimated

using the Coomassie protein assay reagent (Thermo Sci-

entific, Rockford, IL, USA). Protein samples (40 lg) were

electrophoresed on 8–15 % SDS-PAGE gels. Proteins

were transferred to nitrocellulose membranes, which were

blocked in 5 % skim milk in TBS (25 mM Tris base and

150 mM NaCl) for 2 h at room temperature, and then

incubated with primary antibody overnight at 4 �C. The

membranes were then incubated with horseradish peroxi-

dase-conjugated secondary antibodies at 1:5,000 dilutions

for 1 h at room temperature and then washed three times in

TBST (TBS and 0.1 % Tween 20). b-actin and HDAC-1

were used as reference proteins for the normalization of

cytosolic and nuclear protein contents, respectively. The

target proteins were detected with ECL detection reagents,

and the relative intensities of the bands were analyzed by

Image-J software.

Immunocytochemical analysis

The effect of HCA on GSK3-b and Snail localization in

breast cancer cells was demonstrated using immunofluo-

rescence staining as described [25]. Briefly, MDA-MB-231

and MDA-MB-435 cells were cultured in 6-well plates

containing cover slides. On the following day, the cells were

treated with 100 nM HCA for 24 h and subsequently washed

three times in PBS. Cells were immediately fixed in 4 %

paraformaldehyde for 1 h and then washed with PBS. Cells

were cleared in a buffer containing 0.1 % Triton X-100 and

0.1 % sodium citrate at pH 6 for 5 min, followed by three

washes in PBS. Cells were blocked by adding Tris-buffered

saline solution (TBS) containing BSA (0.05 M TBS ? 3

drops of albumin serum) for 1 h at room temperature. The

primary rabbit polyclonal GSK3b or Snail antibodies were

added to the TBS solution and incubated at room temperature

for 1 h. After three washes with PBS, the appropriate fluo-

rescent secondary antibodies were added for 40 min, fol-

lowed by three washes in PBS. Cells were then stained with

Hoechst nuclear counterstain for one minute, washed several

times with PBS, and then examined under fluorescence

microscopy (Olympus BX 51, Tokyo, Japan).

Subcellular protein fractionation

The effect of HCA on GSK3-b and Snail localization was

confirmed using subcellular fractionation followed by

western blot analysis, as described previously [32]. Cells

(1 9 106) were cultured in 100 mM plates overnight and

subsequently treated with 100 nM of HCA for 24 h. Cells

were washed in cold PBS and lysed in 300 ll of fraction-

ation buffer (10 mM Tris–HCl, 1 mM EDTA, 0.5 %

NP-40, and protease inhibitor cocktail). After incubation

on ice for 30 min, cells were centrifuged at 6009 g for

10 min. The supernatants and dissolved pellets were used

as cytoplasmic and nuclear fractions, respectively.

In vivo metastasis assay

All animal works were performed in accordance with a

protocol approved by the Institutional Animal Care and Use

Committee. To induce lung metastasis formation, mice were

anaesthetized, and a small incision was made to reveal the

mammary gland; 106 MDA-MB231 cells were injected

directly into the third mammary fat pad. The incision was

closed with wound clips, and the primary tumor was

removed at day 35. Mice were randomized into 3 groups

(n = 5): control (vehicle), HCA (50 mg/kg), and doxoru-

bicin (0.2 mg/kg) as a positive control. HCA was adminis-

tered daily by gavage, whereas doxorubicin was

administered by intraperitoneal injection once per 3 days.

The mouse lungs were harvested at day 35 after the removal

of the primary tumor, and the metastatic nodules in the lungs

were counted. For histological examination, the mouse lungs

were fixed into 10 % buffered formalin and subjected for

paraffin section with routine procedure. Microscopic

metastasis were examined and counted the number from the

H&E stained slide under light microscopy.

Statistical analysis

The differences in mean values among groups were eval-

uated and expressed as the mean ± SD. Averages were

drawn, and the statistical calculations were performed

using a student t test in Microsoft Excel 2007. For the

statistical difference of microscopic metastasis from H&E

sections, Mann–Whitney test in R (ver2.13.2) was used.

Results

HCA and BCA inhibit breast cancer cell migration

without affecting viability

To test the effect of HCA and BCA on EMT in breast

cancer cells, we examined the anti-migratory effect of

700 Breast Cancer Res Treat (2013) 137:697–708

123

these compounds at 100, 500, and 1,000 nM concentrations

by transwell migration assay kit. Under these conditions,

both HCA and BCA showed anti-migratory effects in a

panel of breast cancer cell lines (Fig. 1a) without affecting

cell viability (Fig. 1b). Anti-migratory effects for 100 nM

of HCA was also observed in wound healing assays using

MDA-MB-231 (Fig. 1c) and MDA-MB-435 (Fig. 1d)

breast cancer cells.

HCA suppresses Snail, resulting in increased

E-cadherin transcriptional activity

Since E-cadherin expression is a hallmark of EMT, and

Snail is a well-known EMT inducer that acts as a repressor

of E-cadherin transcription, we investigated the effect of

HCA on E-cadherin transcript expression in MDA-MB-231

and MDA-MB-435 cells. In both cell types, 100 nM HCA

successfully increased E-cadherin transcription (Fig 2a),

revealing that nanomolar concentrations of HCA can

induce epithelial phenotypes in breast cancer cells. To test

whether this effect might be mediated by changes in the

transcriptional repressor that binds to the E-cadherin

proximal promoter, we next examined the E-cadherin

proximal promoter activity in response to HCA treatment.

HCA and BCA increased E-cadherin promoter activity

compared to the E-box mutant promoter (Fig. 2b). In

addition, HCA upregulated mRNA expression of claudin

and occludin which are representative epithelial markers

(Fig. 2c).

On the contrary, the endogenous Snail protein levels in

MDA-MB-231 and MDA-MB-435 cells were suppressed

by HCA (Fig. 2d), while the Snail transcript levels were

not affected significantly (Fig. 2e), suggesting that HCA

suppresses Snail expression via a post-translational mech-

anism. The expression of other mesenchymal markers such

as vimentin and Twist was also decreased by HCA

(Fig. 2d, f, g). Furthermore, cellular morphological change

by HCA was observed after 24 h by phase-contrast mi-

croscophy analysis. As shown in Fig. 2h, HCA treatment

decreased the elongation of cell shape in both MDA-MB-

231 and MDA-MB-435 cells, suggesting the epithelial-like

morphological changes.

Fig. 1 The effect of HCA on breast cancer cell migration and

invasion. Transwell cell invasion assay was performed with HCA and

its derivative BCA (100, 500, or 1,000 nM) in breast cancer cells (a).

To investigate the effect of HCA on cell viability, cells were

incubated with the indicated doses of HCA for 24 h, and then the cell

viability was assessed using MTT assay (b). These results are from

three independent experiments and each bar represents standard

deviation. Statistical significance was calculated using the student’s

t test (*p \ 0.01, **p \ 0.05). A wound healing assay was performed

to evaluate cell migration inhibition by HCA. MDA-MB-231 (c) and

MDA-MB-435 (d) cell monolayers were scratched, allowed to heal in

the presence of 100 nM of HCA for 24 h, and the images were

obtained by phase-contrast microscopy

Breast Cancer Res Treat (2013) 137:697–708 701

123

HCA affects Wnt signaling pathway

Wnt signaling seems to stabilize the nuclear Snail post-

transcriptionally through the shuttling function of the

Axin2-GSK3 complex. Thus, we investigated the effect of

HCA on the cellular translocalization of GSK-3b and Snail.

Indeed, HCA treatment increased nuclear GSK-3b levels

(Fig. 3a) and decreased nuclear Snail levels (Fig. 3b) as

compared with DMSO vehicle control in both MDA-MB-

231 and MDA-MB-435 cells. These data were confirmed

by western blot analysis with cell-fractioned protein sam-

ples (Fig. 3c). These results suggest that nanomolar con-

centration of HCA increases E-cadherin transcriptional

activity and suppresses Snail expression by inhibiting Wnt

signaling.

HCA attenuates EGF-induced EMT in MCF-7 cells

It is well known that EGF is a potent stimulator of EMT in

several cell types [33, 34]. Therefore, we pretreated EGF

(100 ng/ml) or EGF plus HCA for 24 h in MCF-7 cells and

evaluated the effect of HCA on EGF-induced EMT. Con-

trols cells were treated with DMSO vehicle. As shown in

Fig. 4a, b, EGF treatment for 24 h increased Snail and

Twist gene expression remarkably. On the contrary, the

expression of E-cadherin mRNA was decreased (Fig. 4c) at

the same condition. However, EGF plus HCA cotreatment

recovered mRNA or protein expression of these genes,

suggesting that HCA attenuates EMT progression by EGF

in MCF-7 cells. In addition, we evaluated the morpholog-

ical changes of MCF-7 cells at the same experimental

conditions. As shown in Fig. 4d, HCA cotreatment inhib-

ited the elongation and cell scattering by EGF, supporting

EMT markers expression data.

HCA induces KLF17 expression and suppresses SP-1

and ID-1 expression

Id-1 is a key target protein in increasing breast cancer cell

invasion and is upregulated by the transcription factor SP1

[18]. KLF17 acts as a transcriptional repressor for Id-1, and

increased KLF17 activity thus resulting in the downregu-

lation of breast cancer cell invasion or EMT [25]. There-

fore, we compared KLF17, Id-1, and SP1 protein

expression levels in invasive (MDA-MB-231 and MDA-

MB-435) and non-metastatic (MCF-7 and T47D) breast

Fig. 2 The effect of HCA on E-cadherin, Snail, and vimentin

expression. Cells were treated with the indicated doses of HCA for

24 h, and the E-cadherin mRNA level was subsequently investigated

using real-time PCR (a). The E-cadherin repressor activity of wild-

type and NLS mutants in MDA-MB-231 was assessed with the

reporter construct E-cad (-108)-Luc, which contains the wild-type

promoter sequence from nt -108 to ?125 of the endogenous

E-cadherin promoter, or a control construct, E-cad (-108)-3 9 Mut-

Luc, which harbors mutations in all three E-boxes of the E-cadherin

promoter (b). Cells were treated with 100 nM HCA for 24 h, after

which the mRNA expression of Claudine and occludin was analyzed

(c). Snail, and vimentin protein level (d), and mRNA level (e, f) were

determined using western blotting and real-time PCR analyses. Twist

mRNA expression was investigated using real-time PCR under the

same condition (g). Data from three independent experiments were

analyzed, and statistical significance was calculated using the

student’s t test (*p \ 0.01, **p \ 0.05). Cellular morphological

change by HCA was observed after 24 h by phase-contrast micros-

copy (h). Controls cells were treated with DMSO vehicle. Scale bar20 lm

702 Breast Cancer Res Treat (2013) 137:697–708

123

cancer cells. We found that KLF17 expression is signifi-

cantly higher in non-metastatic breast cancer cells, while

Sp1 and Id-1 are expressed at higher levels in metastatic

cells (Suppl. data Fig. 1).

We then investigated the effect of HCA on KLF17,

SP1, and Id-1 expression. KLF17 mRNA (Fig. 5a) and

protein (Fig. 5d) expression were significantly upregu-

lated by HCA treatment in MCF-7, MDA-MB-231, and

MDA-MB-435 breast cancer cells. Furthermore, HCA

treatment significantly decreased SP-1 mRNA (Fig. 5b)

and protein (Fig. 5d) expression levels. Together, these

data show that Id-1 expression is significantly inhibited

by HCA treatment in all three cell lines, as expected

(Fig. 5c, d).

HCA suppressed metastasis of MDA-MB-231 cells

in vivo

To test the effect of HCA on the in vivo metastatic capability

of breast cancer, we next treated mice that had been injected

orthotopically with MDA-MB-231 cells with HCA (50 mg/

kg), doxorubicin (2 mg/kg), or vehicle, administered daily.

After the removal of the primary tumors, the metastatic

potential of the tumors and the effect of HCA were evaluated

by lung autopsy. Indeed, the number of metastatic breast

cancer cell colonies in the lung parenchyma was significantly

reduced by HCA treatment. Metastasis in the HCA treatment

group was comparable to that in the doxorubicin treatment

group (Fig. 6a, b, p \ 0.05), while the body weight was not

Fig. 2 continued

Breast Cancer Res Treat (2013) 137:697–708 703

123

Fig. 3 The effect of HCA on

GSK3-b and Snail localization.

After 24 h of HCA treatment,

the cellular localization of

GSK3-b and Snail was

evaluated by

immunocytochemistry (a, b).

These data were confirmed by

western blot analysis with cell-

fractioned protein samples (c)

704 Breast Cancer Res Treat (2013) 137:697–708

123

Fig. 4 The effect of HCA on

EGF-induced EMT in MCF-7

cells. MCF-7 was treated with

EGF (50 ng/ml) and/or HCA

(100 nM). After 24 h, Snail (a),

Twist (b), and E-cadherin

(c) expression was analyzed.

Cellular morphological change

was observed after 24 h by

phase-contrast microscopy (d).

Controls cells were treated with

DMSO vehicle. These results

are from three independent

experiments and each bar

represents standard deviation.

Scale bar 20 lm

Fig. 5 mRNA (a, b, c) and

protein expression changes

(d) in KLF17, Sp1, and Id-1 in

breast cancer cells. After 24 h

of HCA (100 nM) treatment,

mRNA and protein expression

were evaluated using real-time

PCR and western blot analyses,

respectively. These results are

from three independent

experiments and each bar

represents standard deviation.

Statistical significance was

calculated using the student’s

t test (*p \ 0.01)

Breast Cancer Res Treat (2013) 137:697–708 705

123

affected by HCA (data not shown), demonstrating that HCA

administration effectively suppresses lung metastatic

potential without inducing systemic toxicity in vivo. Further,

HCA treatment was effective to reduce the micrometastasis

under histological examination (Fig. 6c, d, p \ 0.01). High

power examination of metastatic nodules showed similar

histologic findings among the group, suggesting that HCA

treatment prevents metastatic potential of cancer cells from

primary site rather than affect lung parenchyme of the

microenvironment.

Discussion

Breast cancer is the most common malignant disease in

women and is responsible for 23 % of all female cancers

worldwide. The main cause of cancer death is not the

primary tumor but its metastases at distal sites [35]. EMT is

critical for the development of cells with a mesenchymal

phenotype from the epithelial phenotype, which is neces-

sary to induce highly aggressive cancers [36]. Cinnamal-

dehyde and its derivatives inhibit cancer cell proliferation

and have toxic effects in several cancer cells [28, 37–40].

One recent study showed that cinnamon extract reduced

cell migration in human cervical cancer cell via the

downregulation of MMP-2 mRNA [41] or by blocking

VEGFR2 kinase and its downstream signaling [42]. Fur-

thermore, cinnamic acid was reported to reduce the inva-

sive capacity of melanoma cells via downregulation of

MMP-2 [43]. In contrast, it has been reported that cinna-

maldehyde increases Langerhans cell migration via the

upregulation of CXCL12 [44].

We investigated the effect of HCA on breast cancer cell

invasion and metastasis and found that, at subtoxic doses

(100 nM), both HCA and BCA inhibit breast cancer cell

invasion or EMT by upregulating the expression of E-cad-

herin. We investigated the molecular mechanism(s) by

which HCA inhibits breast cancer EMT and cell invasion.

HCA downregulates the Snail protein, which acts as a

transcriptional repressor of E-cadherin, and thus upregulates

E-cadherin promoter activity. HCA-induced Snail protein

downregulation seems to be associated with GSK3-bnuclear localization and Snail nuclear exclusion. This is

consistent with the fact that nuclear GSK3-b phosphorylates

Snail and thereby induces its nuclear export, which is fol-

lowed by Snail protein degradation [45–47].

Fig. 6 The effect of HCA on lung metastasis of breast cancer tissue

transplanted into the MPF of nude mice. Mice that had been injected

orthotopically with MDA-MB-231 cells received daily treatment with

HCA (50 mg/kg) or vehicle for 35 days. Doxorubicin (2 mg/kg) was

administered by intraperitoneal injection once per 3 days for the

same period. After the primary tumors were removed, the number

of metastases was evaluated by lung autopsy (a, b), (**p \ 0.01).

A representative histological finding of metastatic lung nodules

(c) under low magnification (upper panels) and high magnification

(lower panels). Small rectangular squares indicate micrometastatic

tumor nodules in lung parenchyme Scale bar 50 lm. Number of

micrometastsis in mouse lung in each groups (d) revealed HCA also

inhibits metastasis of the breast cancer cells (*p \ 0.05)

706 Breast Cancer Res Treat (2013) 137:697–708

123

Previous studies implicated that EGF can mediate EMT

in breast cancer cells [48] and other carcinomas [49, 50]. In

the present study, we treated EGF and/or HCA, and eval-

uated EMT markers expression and morphological change

to confirm the effect of HCA on the EMT of MCF-7 cell.

EGF treatment induced a mesenchymal phenotypes such

as increased motility and upregulation of mesenchymal

markers. Interestingly, EGF and HCA cotreatment restored

the epithelial phenotype from EGF-induced mesenchymal

cell shape. In addition, HCA cotreatment attenuated

upregulation of mesenchymal markers, supporting the

inhibitory effect of HCA on EMT in breast cancer cells.

Id-1 is one of the key target proteins that increases breast

cancer cell invasion [20], and it is upregulated by the tran-

scription factor SP1 [18]. Meanwhile, KLF17 acts as a

transcriptional repressor for Id-1, and its activity results in

the downregulation of breast cancer cell invasion or EMT

[25]. Therefore, we investigated the effect of HCA on Id-1,

KLF17, and SP1 expression. Interestingly, HCA induces a

significant increase in KLF17 mRNA and protein expression

in breast cancer cells. At the same time, HCA effectively

decreased SP1 and Id-1 transcription and protein levels,

suggesting that HCA inhibits breast cancer invasion via the

inhibition of Id-1 through the upregulation of KLF17 and the

downregulation of SP1. It has been reported that both Id-1

and Snail are overexpressed in several types of cancers,

particularly in highly invasive cells and tissues [18, 51].

Furthermore, Id-1 mRNA and protein expression are

increased in Snail-overexpressing MDCK cells, suggesting

that Id-1 is a downstream target of Snail [18].

In conclusion, we report for the first time that sub-toxic

doses of HCA have anti-metastastic effects on breast can-

cer and inhibit EMT. HCA exerts its EMT inhibitory

activity by downregulating Snail protein activity, which in

turn promotes the upregulation of E-cadherin promoter

activity. Furthermore, HCA inhibits breast cancer invasion

in part via the inhibition of the Sp1/Id-1 signaling pathway

by KLF17 upregulation.

Acknowledgments This work was supported by the Basic Science

Research Program through the National Research Foundation of

Korea Grant funded by the Korean Government (2009-0070462). This

research was supported by the Bio & Medical Technology Devel-

opment Program of the National Research Foundation funded by the

Korean government (2012M3A9C404877).

Disclosures None.

References

1. Kwon BM, Cho YK, Lee SH, Nam JY, Bok SH, Chun SK et al

(1996) 20-Hydroxycinnamaldehyde from stem bark of cinnamo-

mum cassia. Planta Med 62:183–184

2. Han DC, Lee MY, Shin KD, Jeon SB, Kim JM, Son KH et al (2004)

20-benzoyloxycinnamaldehyde induces apoptosis in human carci-

noma via reactive oxygen species. J Biol Chem 279:6911–6920

3. Kwon BM, Lee SH, Choi SU, Park SH, Lee CO, Cho YK et al

(1998) Synthesis and in vitro cytotoxicity of cinnamaldehydes to

human solid tumor cells. Arch Pharm Res 21:147–152

4. Lee CW, Hong DH, Han SB, Park SH, Kim HK, Kwon BM et al

(1999) Inhibition of human tumor growth by 20-hydroxy- and 20-benzoyloxycinnamaldehydes. Planta Med 65:263–266

5. Moon EY, Lee MR, Wang AG, Lee JH, Kim HC, Kim HM et al

(2006) Delayed occurrence of H-ras12V-induced hepatocellular

carcinoma with long-term treatment with cinnamaldehydes. Eur J

Pharmacol 530:270–275

6. Lee K, Kwon BM, Kim K, Ryu J, Oh SJ, Lee KS et al (2009)

Plasma pharmacokinetics and metabolism of the antitumour drug

candidate 20-benzoyloxycinnamaldehyde in rats. Xenobiotica 39:

255–265

7. Steeg PS (2006) Tumor metastasis: mechanistic insights and

clinical challenges. Nat Med 12:895–904

8. Steeg PS (2007) Cancer: micromanagement of metastasis. Nature

449:671–673

9. Eccles SA, Welch DR (2007) Metastasis: recent discoveries and

novel treatment strategies. Lancet 369:1742–1757

10. Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC, Dirks

PB (2009) Tumour-initiating cells: challenges and opportunities

for anticancer drug discovery. Nat Rev Drug Discov 8:806–823

11. Sreekumar R, Sayan BS, Mirnezami AH, Sayan AE (2011)

MicroRNA Control of Invasion and Metastasis Pathways. Front

Genet 2:58

12. Nieto MA (2002) The snail superfamily of zinc-finger tran-

scription factors. Nat Rev Mol Cell Biol 3:155–166

13. Xu C, Kim NG, Gumbiner BM (2009) Regulation of protein stability

by GSK3 mediated phosphorylation. Cell Cycle 8:4032–4039

14. Lim SO, Kim H, Jung G (2010) p53 Inhibits tumor cell invasion

via the degradation of snail protein in hepatocellular carcinoma.

FEBS Lett 584:2231–2236

15. Kim NH, Kim HS, Li XY, Lee I, Choi HS, Kang SE et al (2011)

A p53/miRNA-34 axis regulates Snail1-dependent cancer cell

epithelial-mesenchymal transition. J Cell Biol 195:417–433

16. Zannetti A, Del Vecchio S, Romanelli A, Scala S, Saviano M, Cali

G et al (2005) Inhibition of Sp1 activity by a decoy PNA-DNA

chimera prevents urokinase receptor expression and migration of

breast cancer cells. Biochem Pharmacol 70:1277–1287

17. Hung WC, Chang HC (2009) Indole-3-carbinol inhibits Sp1-

induced matrix metalloproteinase-2 expression to attenuate

migration and invasion of breast cancer cells. J Agric Food Chem

57:76–82

18. Jorda M, Vinyals A, Marazuela A, Cubillo E, Olmeda D, Valero

E et al (2007) Id-1 is induced in MDCK epithelial cells by

activated Erk/MAPK pathway in response to expression of the

Snail and E47 transcription factors. Exp Cell Res 313:2389–2403

19. Zhang X, Ling MT, Wang Q, Lau CK, Leung SC, Lee TK et al

(2007) Identification of a novel inhibitor of differentiation-1 (ID-

1) binding partner, caveolin-1, and its role in epithelial-mesen-

chymal transition and resistance to apoptosis in prostate cancer

cells. J Biol Chem 282:33284–33294

20. Fong S, Itahana Y, Sumida T, Singh J, Coppe JP, Liu Y et al (2003)

Id-1 as a molecular target in therapy for breast cancer cell invasion

and metastasis. Proc Natl Acad Sci USA 100:13543–13548

21. Darnel AD, Wang D, Ghabreau L, Yasmeen A, Sami S, Akil N

et al (2010) Correlation between the presence of high-risk human

papillomaviruses and Id gene expression in Syrian women with

cervical cancer. Clin Microbiol Infect 16:262–266

22. Ding Y, Wang G, Ling MT, Wong YC, Li X, Na Y et al (2006)

Significance of Id-1 up-regulation and its association with EGFR

in bladder cancer cell invasion. Int J Oncol 28:847–854

Breast Cancer Res Treat (2013) 137:697–708 707

123

23. Li B, Tsao SW, Li YY, Wang X, Ling MT, Wong YC et al (2009)

Id-1 promotes tumorigenicity and metastasis of human esopha-

geal cancer cells through activation of PI3K/AKT signaling

pathway. Int J Cancer 125:2576–2585

24. Cheung PY, Yip YL, Tsao SW, Ching YP, Cheung AL (2011) Id-

1 induces cell invasiveness in immortalized epithelial cells by

regulating cadherin switching and Rho GTPases. J Cell Biochem

112:157–168

25. Gumireddy K, Li A, Gimotty PA, Klein-Szanto AJ, Showe LC,

Katsaros D et al (2009) KLF17 is a negative regulator of epi-

thelial-mesenchymal transition and metastasis in breast cancer.

Nat Cell Biol 11:1297–1304

26. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY

et al (2008) The epithelial-mesenchymal transition generates cells

with properties of stem cells. Cell 133:704–715

27. Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg

RA et al (2009) Identification of selective inhibitors of cancer

stem cells by high-throughput screening. Cell 138:645–659

28. Cabello CM, Bair WB 3rd, Lamore SD, Ley S, Bause AS, Azi-

mian S et al (2009) The cinnamon-derived Michael acceptor

cinnamic aldehyde impairs melanoma cell proliferation, inva-

siveness, and tumor growth. Free Radic Biol Med 46:220–231

29. Liang CC, Park AY, Guan JL (2007) In vitro scratch assay: a

convenient and inexpensive method for analysis of cell migration

in vitro. Nat Protoc 2:329–333

30. Yook JI, Li XY, Ota I, Hu C, Kim HS, Kim NH, Cha SY, Ryu JK,

Choi YJ, Kim J, Fearon ER, Weiss SJ (2006) A Wnt-Axin2-

GSK3beta cascade regulates Snail1 activity in breast cancer cells.

Nat Cell Biol 8:1398–1406

31. Livak KJ, Schmittgen TD (2001) Analysis of relative gene

expression data using real-time quantitative PCR and the 2(-Delta

Delta C(T)) method. Methods 25:402–408

32. Fan J, Ren H, Fei E, Jia N, Ying Z, Jiang P et al (2008) Su-

moylation is critical for DJ-1 to repress p53 transcriptional

activity. FEBS Lett 582:1151–1161

33. Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y, Abbruzzese JL,

Hortobagyi GN, Hung MC (2007) Epidermal growth factor

receptor cooperates with signal transducer and activator of tran-

scription 3 to induce epithelial-mesenchymal transition in cancer

cells via up-regulation of TWIST gene expression. Cancer Res

67:9066–9076

34. Vergara D, Valente CM, Tinelli A, Siciliano C, Lorusso V,

Acierno R, Giovinazzo G, Santino A, Storelli C, Maffia M (2011)

Resveratrol inhibits the epidermal growth factor-induced epithe-

lial mesenchymal transition in MCF-7 cells. Cancer Lett 310:1–8

35. Weigelt B, Peterse JL, van ‘t Veer LJ (2005) Breast cancer

metastasis: markers and models. Nat Rev Cancer 5:591–602

36. Arias AM (2001) Epithelial mesenchymal interactions in cancer

and development. Cell 105:425–431

37. Ren Z, Kang YH, Shi ZY, Huang-Fu CS, Hu GQ, Liu B (2010)

Cinnamaldehyde of loxacin-3-ylhydrazone induces apoptosis of

human hepatocarcinoma SMMC-7721 cells. Yao Xue Xue Bao

45:1109–1115

38. Kwon HK, Hwang JS, So JS, Lee CG, Sahoo A, Ryu JH et al

(2010) Cinnamon extract induces tumor cell death through inhi-

bition of NFkappaB and AP1. BMC Cancer 10:392

39. Lee CW, Lee SH, Lee JW, Ban JO, Lee SY, Yoo HS et al (2007)

2-Hydroxycinnamaldehyde inhibits SW620 colon cancer cell

growth through AP-1 inactivation. J Pharmacol Sci 104:19–28

40. Ka H, Park HJ, Jung HJ, Choi JW, Cho KS, Ha J et al (2003)

Cinnamaldehyde induces apoptosis by ROS-mediated mito-

chondrial permeability transition in human promyelocytic leu-

kemia HL-60 cells. Cancer Lett 196:143–152

41. Koppikar SJ, Choudhari AS, Suryavanshi SA, Kumari S,

Chattopadhyay S, Kaul-Ghanekar R (2010) Aqueous cinnamon

extract (ACE-c) from the bark of Cinnamomum cassia causes

apoptosis in human cervical cancer cell line (SiHa) through loss

of mitochondrial membrane potential. BMC Cancer 10:210

42. Lu J, Zhang K, Nam S, Anderson RA, Jove R, Wen W (2010)

Novel angiogenesis inhibitory activity in cinnamon extract blocks

VEGFR2 kinase and downstream signaling. Carcinogenesis

31:481–488

43. Liu L, Hudgins WR, Shack S, Yin MQ, Samid D (1995) Cin-

namic acid: a natural product with potential use in cancer inter-

vention. Int J Cancer 62:345–350

44. Ouwehand K, Santegoets SJ, Bruynzeel DP, Scheper RJ, de

Gruijl TD, Gibbs S (2008) CXCL12 is essential for migration of

activated Langerhans cells from epidermis to dermis. Eur J

Immunol 38:3050–3059

45. Ko H, Kim HS, Kim NH, Lee SH, Kim KH, Hong SH et al (2007)

Nuclear localization signals of the E-cadherin transcriptional

repressor Snail. Cells Tissues Organs 185:66–72

46. Yook JI, Li XY, Ota I, Fearon ER, Weiss SJ (2005) Wnt-

dependent regulation of the E cadherin repressor snail. J Biol

Chem 280:11740–11748

47. Yang Z, Rayala S, Nguyen D, Vadlamudi RK, Chen S, Kumar R

(2005) Pak1 phosphorylation of snail, a master regulator of epi-

thelial-to-mesenchyme transition, modulates snail’s subcellular

localization and functions. Cancer Res 65:3179–3184

48. Hardy KM, Booth BW, Hendrix MJ, Salomon DS, Strizzi L

(2010) ErbB/EGF signaling and EMT in mammary development

and breast cancer. J Mammary Gland Biol Neoplasia 15:191–199

49. Gan Y, Shi C, Inge L, Hibner M, Balducci J, Huang Y (2010)

Differential roles of ERK and Akt pathways in regulation of

EGFR-mediated signaling and motility in prostate cancer cells.

Oncogene 29:4947–4958

50. Cai Z, Zhou Y, Lei T, Chiu JF, He QY (2009) Mammary serine

protease inhibitor inhibits epithelial growth factor-induced epi-

thelial-mesenchymal transition of esophageal carcinoma cells.

Cancer 115:36–48

51. Li J, Jia H, Xie L, Wang X, He H, Lin Y, Hu L (2009) Correlation

of inhibitor of differentiation 1 expression to tumor progression,

poor differentiation and aggressive behaviors in cervical carci-

noma. Gynecol Oncol 114:89–93

708 Breast Cancer Res Treat (2013) 137:697–708

123