Arch Toxicol DOI 10.1007/s00204-014-1301-z Genotoxicity and Carcinogenicity Bisphenol A modulates colorectal cancer protein profile and promotes the metastasis via induction of epithelial to mesenchymal transitions Zhuo‑Jia Chen · Xiang‑Ling Yang · Hao Liu · Wei Wei · Kun‑Shui Zhang · Hong‑Bin Huang · John P. Giesy · Huan‑Liang Liu · Jun Du · Hong‑Sheng Wang Received: 16 January 2014 / Accepted: 17 June 2014 © Springer-Verlag Berlin Heidelberg 2014 Abstract More and more evidences indicate that endocrine disruptor chemicals such as bisphenol A (BPA) can act as carcinogens and enhance susceptibility to tumorigenesis. Although the gut is in direct contact with orally ingested BPA, effects of BPA on occurrence and development of colorectal cancer remain an unexplored endpoint. Colorectal cancer SW480 cells treated with nanomolar (10−8 M) or greater (10−5 M) concentrations of BPA were compared with responses of a control group. Proteomic study revealed that more than 56 proteins were modulated following exposure to BPA, which are relevant to structure, motility and proliferation of cells, production of ATP, oxidative stress, and protein metabolism. Further studies revealed that BPA increased migration and invasion and triggered transformations from epithelial to mesenchymal transitions (EMTs) of colorectal cancer cells, which was characterized by acquiring mesenchymal spindle-like Electronic supplementary material The online version of this article (doi:10.1007/s00204-014-1301-z) contains supplementary material, which is available to authorized users. Z.-J. Chen (*) · H.-B. Huang Department of Pharmacy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China e-mail: chenzhuojia86@163.com X.-L. Yang · H.-L. Liu Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Institute of Human Virology, Key Laboratory of Tropical Disease Control (Ministry of Education), Sun Yat-sen University, Guangzhou 510655, China H. Liu Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China morphology and increasing the expression of N-cadherin with a concomitant decrease of E-cadherin. Accordingly, BPA treatment increased the expression of transcription factor Snail. Furthermore, signal AKT/GSK-3β-mediated stabilization of Snail is involved during BPA-induced EMT of colon cancer cells. Our study first demonstrated that the xenoestrogen BPA at nanomolar and greater concentrations modulates the protein profiles and promotes the metastasis of colorectal cancer cells via induction of EMT. Keywords Colorectal cancer · BPA · EMT · Proteomic · Tumorigenesis · Migration Abbreviations BPABisphenol A CRCColorectal cancer DMSODimethyl sulfoxide E2Estradiol E-CadE-cadherin EDCsEndocrine disruptor chemicals W. Wei · J. Du (*) · H.-S. Wang (*) Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, No. 132 Waihuandong Road, University Town, Guangzhou 510006, China e-mail: dujun@mail.sysu.edu.cn H.‑S. Wang e-mail: whongsh@mail.sysu.edu.cn; hongshengwang@foxmail.com K.-S. Zhang Department of Pharmacy, Sun Yat‑sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China J. P. Giesy Department of Veterinary Biomedical Sciences, Toxicological Center, University of Saskatchewan, Saskatoon, SK, Canada 13 EMTEpithelial to mesenchymal transitions ERREstrogen-related receptor ERα/βEstrogen receptor α/β FBSFetal bovine serum FNFibronectin GPERG-protein-coupled estrogen receptor hsp27Heat-shock protein 27 N-CadN-cadherin VimVimentin ZO-1Zona occludin-1 Introduction The xenoestrogen bisphenol A (BPA), a food contaminant with endocrine disrupting activity, is widely used to manufacture consumer products including drinking water bottles and reusable food containers (Brotons et al. 1995). BPA can leach from the polymers into food and water under normal conditions, furthermore, exposure to elevated temperatures (boiling, heating) greatly increases its rate of migration (Le et al. 2008). Due to an increase in products based on epoxy resins and polycarbonate plastics, exposure of human beings to BPA via food consumption and environmental intakes has increased in recent years (Kang et al. 2006). Numerous studies have suggested that human exposure to BPA is widespread, which is evidenced by its presence in urine, blood, fetal tissues, and amniotic fluid (Geens et al. 2012; Vandenberg et al. 2007a). Colorectal cancer (CRC), also called colon cancer or large bowel cancer, is the third most common form of cancer and the second leading cause of death due to cancer in the Western world (Siegel et al. 2013). Previous studies indicated that more than 80 % of CRC cases and deaths are attributable to diet and environmental factors (Jasperson et al. 2010). In recent years, several lines of clinical and experimental evidences have reported that estrogens, xenoestrogens, and their receptors are involved in development and regulation of CRC (Chen and Iverson 2012). Classical estrogen receptor α/β (ERα/β), G-protein-coupled estrogen receptor (GPER) and estrogen-related receptor (ERR) have been detected in various colon cancer cell lines (Kennelly et al. 2008). Considering all these estrogen receptors can mediate estrogenic signaling of endocrine disruptor chemicals (EDCs), the roles of BPA on tumorigenesis and progression of CRC should be illustrated. There is a global concern for adverse endocrine disruptive effect and disturbing of normal sex hormone balance caused by BPA. Recent studies revealed that chronic exposure to reference doses of BPA can decrease sperm production and fertility (Salian et al. 2011), stimulate the development of mammary gland (Vandenberg et al. 2007b), increase body weight (Vom Saal et al. 2012), and 13 Arch Toxicol promote the tumorigenesis and development. To date, these studies examining effects of BPA on tumorigenesis have focused on the reproductive tract and mammary glands. Rats exposed prenatally to greater doses of BPA-induced in situ development of carcinomas (Durando et al. 2007). Exposure to BPA during nursing followed by exposure at 50 days of age to dimethylbenzantracene resulted in more of tumors per rat and a shorter latency period compared with animals not exposed to BPA (Jenkins et al. 2009). Although gut is in direct contact with BPA consumed orally (Braniste et al. 2010; Dekant and Voelkel 2008), there is very limited data concerning the effects of BPA on progression and development of CRC. Therefore, the present study was designed to investigate effects of BPA on progression of colon cancer. Profiles of proteins altered by exposure to either nanomolar or micromolar concentrations of BPA on SW480 cells were determined by proteomic analysis. According to the identified proteins, expression of which were altered by exposure to BPA on migration of colon cancer were further confirmed and investigated. To our knowledge, this is the first study to identify changes in expression of proteins and illustrate effects of BPA on CRC metastasis. Materials and methods Reagents All reagents used in 2-DE were bought from Bio-Rad (Hercules, CA, USA). All chemicals were of reagent grade or better and purchased from Sigma Chemical Co. (St. Louis, MO, USA) unless otherwise noted. BPA was dissolved in dimethyl sulfoxide (DMSO) to prepare a 10 mM stock solution, and stored at −20 °C. Primary antibodies against fascin, Krt8, TIF4A, p-ERK1/2 (Thr202/204), ERK1/2, Bcl2, E-cadherin (E-Cad), Zona occludin-1(ZO-1), N-cadherin (N-Cad), Vimentin (Vim), Snail, Slug, ZEB, Twist, p-AKT (Ser473), AKT, p-GSK-3β (Ser9), GSK-3β, p-p38-MAPK (Thr180/Tyr182), p38-MAPK, and β-actin were purchased from Cell Signaling Technology (Beverly, MA, USA). Primary antibody to fibronectin (FN) was obtained from Bioworld Technology, Inc (Minneapolis, MN, USA). Horseradish peroxidase-conjugated secondary antibody was from Santa Cruz Biotechnology (Santa Cruz, CA, USA). All compounds were solubilized in DMSO. Medium containing 0.5 % DMSO was used as the control. Cell line and cell culture The human CRC cell lines SW480 (passage 8–15) and LoVo (passage 15–18) were cultured in RPMI medium 1640 supplemented with 10 % fetal bovine serum (FBS). The Arch Toxicol HCT-116 cells (passage 12–15) were grown in DMEM supplemented with 10 % FBS. Cells were incubated at 37 °C in a 5 % CO2 atmosphere. Both the plastic items used for the experiments and the water used to prepare the reagents were pretreated by enhanced sonochemical degradation to reduce any potential background BPA (Sheng and Zhu 2011). Analysis of cell proliferation and apoptosis Cell proliferation and apoptosis were detected by use of previously described procedures (Wang et al. 2012). Briefly, SW480 cells were inoculated in 96-well plates for 24 h before exposure to BPA. Viability of cells after treated with increasing concentrations of BPA for the indicated times were evaluated by use of the CCK-8 kit (Dojindo Molecular Technologies, Gaithers burg, MD, USA) according to the manufacturer’s instructions. For cell apoptosis, both the suspension and adherent cells treated with BPA for 48 h were collected into flow cytometry tubes and centrifuged at 2,000 rpm for 5 min to obtain cell pellets. Cells were washed twice with PBS, stained with Annexin V-FITC for 15 min and propidium iodide for 5 min, and then analyzed by flow cytometry using FL1 (Em: 525 nm) and FL3 (Em: 670 nm). time periods. Cells were washed three times with PBS, fixed with 4 % paraformaldehyde for 20 min and permeabilized with 0.3 % Triton X-100 for 10 min. After blocking with goat serum for 2 h at room temperature, cells were incubated with antibodies against E-Cad (1:100 dilutions) at 4 °C overnight. Slides were washed three times with PBS and incubated with Alexa Fluor 488-conjugated secondary antibodies (1:1,000 dilutions) for 1 h at room temperature. Nuclei were stained with DAPI for 10 min. Samples were examined with Confocal Laser Scanning Microscopy (Zeiss) to analyze expression of E-Cad. Statistical analysis All values were reported as mean ± SD of three independent experiments unless otherwise specified. Data were analyzed by two-tailed unpaired Student’s t test between two groups and by one-way ANOVA followed by Bonferroni test for multiple comparison involved. Homogeneity of variance was confirmed by use of Levine’s test. The statistical analyses were performed using SPSS 17.0 for Windows. A p value of <0.05 was considered to be statistically significant. Results Proteomic analysis of proteins in SW480 cells Cytotoxic effects of BPA on SW480 cells SW480 cells were exposed to vehicle (0.5 % DMSO), 10−8 or 10−5 M BPA and incubated for 48 h. Two-dimensional gel electrophoresis (2-DE), protein visualization, image analysis was performed essentially as described in the Supplementary Materials (SM). The detailed procedures for ingel tryptic digestion of proteins and MALDI–TOF–MS/MS analysis were also described in the SM. Wound healing and transwell migration/invasion assay For the in vitro wound healing assay, confluent monolayers of SW480 cells were scratched, and the migration distance of the cells into the scratched area was measured in 10 randomly chosen fields. Migration and invasion assays were performed in Boyden chambers according to the previous study (Wang et al. 2013) and described in the SM. Western blotting analysis Western blotting was performed as previously described (Jiang et al. 2013) and described in the SM. To determine cytotoxicity of BPA, SW480 cells were treated with concentrations of BPA ranging from 10−8 to 10−4 M for 48 or 72 h, and then cell viability was assessed by CCK-8 kit assay. Our results revealed that concentrations of BPA >10−5 M significantly inhibited proliferation of SW480 cells (Fig. 1a). There was no statistically significant effect on apoptotic/necrotic populations at both 10−8 and 10−5 M BPA as compared to vehicle treated cells of both cell lines after 48 h (Fig. 1b). Concentrations of BPA in blood, fetuses, urine, saliva, and various tissues are in the range of 0.5–10 ng/mL or ng/g, which is equivalent to 2 × 10−9–4.4 × 10−8 M (Vandenberg et al. 2010). Micromolar BPA has been reported to stimulate human cancer cell migration (Derouiche et al. 2013) and proangiogenic of human primary endothelial cells (Andersson and Brittebo 2012). Therefore, 10−8 and 10−5 M were chosen to represent the oral equivalent and high concentrations of BPA for further proteomic study (Ge et al. 2014). Immunofluorescence The analysis and validation of BPA‑induced proteome alterations in SW480 cells SW480 cells were cultured on chamber slides, serum starved for 12 h, then exposed to BPA for the indicated The differentially expressed proteins in SW480 cells treated with BPA were analyzed by MALDI–TOF–MS/ 13 Arch Toxicol Fig. 1 Effects of BPA on proliferation and apoptosis of SW480 cells. a Cells were treated with various concentrations (10−8–10−4 M) of BPA for 48 or 72 h. Viability of cells was assessed by CCK8 kit assay. b Cells were exposed to BPA for 48 h. Collected cells were stained with annexin V-FITC and PI, and then analyzed by flow cytometry. Data are presented as mean ± SD of three independent experiments. *p < 0.05 compared with control MS. Representative two-dimensional gel images of control, 10−8 and 10−5 M BPA-treated cells are shown in Fig. S1. The 2-DE maps were compared with PDQuest software to identify protein spots that varied among treatments. After exposure to BPA, significantly (p < 0.05) differentially expressed protein spots observed in all replicate gels were scored. The fold difference is represented by the ratio of the intensity value of the BPA-treated group to the value of the control group (Table S1). Totally, 68 differently expressed proteins were found as indicated by the spots marked with arrows (Fig. S1). All these spots were excised from gels for protein identification. And then 56 differentially expressed proteins were identified by MALDI–TOF–MS/MS analysis (Table S1). The Western blotting analysis was performed to further verify the up- or down-regulation of identified proteins (Fig. S2). Validation confirmed up-regulation of Fascin (spot 13) and TIF4A (spot 29) by BPA in SW480 (p53 wt, APC wt, and MSI−), LoVo (p53 mutant, APC wt, and MSI+) and HCT-116 (p53 mutant, APC mutant, and MSI−) cells. Furthermore, down-regulation of Krt8 (spot 64) was also confirmed in SW480 cells treated with BPA. The results not only validated the results of proteomic 13 study but also indicated that protein profiles changed by BPA on SW480 cells were representative for other colorectal cancer cells. Functional categories of identified proteins The PANTHER classification system was used to classify the 56 identified proteins. Those identified proteins were classified into seven categories according to their functions during biological processes (Fig. 2a). Our results indicated that cell structure and motility-related proteins, cell proliferation and apoptosis-related proteins, energy metabolism, and oxidative stress proteins accounted for the major proportions among the detected proteins. Protein–protein interaction network among the identified proteins was predicted based on the STRING system. Notably, five major clusters of interacting proteins were determined by STRING (Fig. 2b), including cell structure and motility, cell proliferation and cell cycle, energy metabolism, oxidative stress, protein folding and metabolism. It suggested that BPA significantly changed expression of proteins related to tumorigenesis and metastasis in colorectal cancer cells. Arch Toxicol Fig. 2 Functional classification and distribution of all identified proteins. a Seven protein groups were categorized based on the putative biological functions of identified proteins and the percentages of each protein group were indicated. b The protein–protein interaction network of the identified proteins. The network containing 56 identified proteins was mapped using STRING system based on evidence with different types BPA promotes the metastasis of colorectal cancer cells The proteomic study revealed that proteins of cell motility and structure were markedly changed after treatment with oral equivalent or greater concentrations of BPA. Therefore, metastasis of SW480 cell was further investigated by use of wound healing and transwell migration/invasion assays. Treatment with either 10−8 or 10−5 M BPA for 48 or 72 h significantly increased wound closure as compared to the control group (p < 0.05) (Fig. 3a, b). Furthermore, exposure to BPA resulted in a significantly more migration (Fig. 3c) and invasion (Fig. 3d) cells. Compared with the untreated cells, the number of migrated and invaded cells was approximately 45 % (migration) and 72 % (invasion) greater after treatment with 10−8 M BPA for 72 h, and about 88 % (migration) and 125 % (invasion) greater after treatment with 10−5 M BPA for 72 h. Collectively, our results indicated that both the oral equivalent and greater concentrations of BPA promotes metastasis of SW480 cells. BPA triggers epithelial to mesenchymal transition (EMT) of SW480 cells Previous studies indicated that increased migration and invasion abilities of tumor cells are consistent with events at EMT, during which, the epithelial makers such as E-cad and ZO-1 are down-regulated, whereas mesenchymal markers such as N-cad and FN are up-regulated (Wang et al. 2013). The EMT of SW480 cells was observed after stimulation with 10−8 and 10−5 M BPA for 96 h. Cells resulted in a significant change in morphology, from cobblestone morphology to mesenchymal spindle-like and fusiform features (Fig. 4a). Western blotting analysis revealed that this morphological change was associated with down-regulation of 13 Arch Toxicol Fig. 3 BPA-triggered migration and invasion of SW480 cells. a Representative images of the monolayer wound healing assay utilizing the SW480 cell line immediately after scratching 48 and 72 h. Cells were treated with 0.5 % DMSO (control), 10−8 and 10−5 M BPA, b quantitative analysis of wound healing assay for SW480 cells treated with BPA; SW480 cells were allowed to migrate (c) and invasive (d) transwell chambers for 48 and 72 h in the presence or absence of BPA (10−8 or 10−5 M). Migrated and invasion cells were fixed, stained, and photographed. The % number of migrated and invasion cells. Data represent the average of three independent experiments. *p < 0.05 compared with control, **p < 0.01 compared with control expression of epithelial characteristics E-Cad, ZO-1 and the up-regulation of expression of mesenchymal characteristics FN, N-cad and Vim (Fig. 4b). Furthermore, BPA treatment for 96 h significantly decreased the expression of E-Cad while increased the expression of Vim in HCT116 cells (Fig. 4c). These results confirmed that BPA can induce EMT of colorectal cancer cells. Since transcription factors Snail, ZEB1, Twist, and Slug play essential roles in regulating EMT (Cano et al. 2007), their expressions were investigated to determine whether they were involved in BPA-induced EMT of colorectal cancer cells. Compared to untreated cells, 10−8 M BPA significantly increased protein levels of Snail and Twist, but not Slug or ZEB, while 10−5 M BPA significantly increased protein levels of Snail, Twist, and ZEB, but not Slug (Fig. 4c). We further performed knockdown assays to investigate the role of Snail in BPA-induced EMT. SW480 cells were transfected with nontargeting control siRNA or siSnail for 24 h, and then treated with 10−5 M BPA for another 96 h. The expression of Vim, E-Cad, and Snail were detected by Western blotting. Results showed that silencing of Snail by siRNA significantly attenuated BPA-induced up-regulation of Vim and down-regulation of E-Cad (Fig. 4d). Collectively, these observations demonstrated that Snail acts as a key regulator in BPA-induced EMT of SW480 cells. 13 Akt/GSK‑3β mediates BPA‑induced EMT of colorectal cancer cells Recent studies revealed that the occurrence of EMT requires AKT/GSK-3β-mediated stabilization of Snail in colorectal cancer cells (Wang et al. 2013). Therefore, phosphorylation and protein levels of Akt, GSK-3β, ERK1/2, and p38-MAPK were detected by Western blotting in SW480 cells exposed to BPA for 96 h. Phosphorylation of Akt and GSK-3β were significantly increased after stimulated by BPA, particularly in cells exposed to 10−5 M BPA for 96 h. However, there was lesser phosphorylation of p-ERK1/2 and no significant change in phosphorylation of p38-MAPK in SW480 cells treated with BPA (Fig. 5a). These results suggested that AKT/GSK-3β may be involved in BPA-induced EMT of CRC. Arch Toxicol Fig. 4 BPA treatment induced EMT of SW480 cells. a SW480 cells were treated with 10−8 or 10−5 M BPA for 96 h. Cell morphological changes associated with EMT are shown in the phase-contrast image. b The expression of epithelial makers (E-cad and ZO-1) and mesenchymal markers (N-cad, Vim and FN) in SW480 cells treated with BPA for 96 h. c The expression of EMT-related transcription factors Snail, Slug, ZEB, and Twist in SW480 cells treated with BPA for 96 h. d SW480 cells transfected with siSnail or si-NC were stimulated with or without 10−5 M BPA for 96 h, and the expression of Vim, E-Cad, and Snail were detected by Western blotting. β-Actin servers as the loading control for Western blotting analysis To verify whether AKT/GSK-3β is involved in BPAinduced EMT of colorectal cancer cells, SW480 and HCT116 cells were treated with the inhibitor of PI3 K/Akt, LY294002 (20 μM), prior to exposure to BPA (10−5 M), expression of p-AKT, p-GSK-3β, Snail, and markers of EMT were determined by Western blotting. The results showed that levels of p-AKT, p-GSK-3β, Snail, and mesenchymal marker Vim were increased while E-Cad was decreased after exposure to BPA for 96 h. However, these effects were reversed upon treating with LY294002 alone or in combination with BPA in both SW480 (Fig. 5b) and HCT-116 (Fig. 5c) cells. Furthermore, the inhibitor of PI3 K/Akt abolished the BPA-induced EMT-like morphological changes and down-regulation of E-Cad in SW480 cells (Fig. 5d). Collectively, these observations suggested that colorectal cancer cells had undergone an EMT after treated by BPA and that AKT/GSK-3β-mediated stabilization of Snail mediated this process. Discussion Proteomic modifications of SW480 cells in response to oral and high concentrations of BPA The results of the proteomic analysis revealed that treatment with BPA significantly modulates expression of proteins involved in various functional activities such as cytoskeletal dynamic flexibility, cell cycle and proliferation, ATP production, antioxidant mechanisms and protein metabolism. Because of the limitations of in identifying proteins, proteomics data must be validated by use of independent methods. Due to the large number of “hits” observed in this study, validation of all proteomic results could not be accomplished. Therefore, three proteins were selected from different functional protein networks, Fascin, Krt8, and TIF4A for Western blotting analysis. The results confirmed the significant greater expression of Fascin and TIF4A and lesser amount of Krt8 in SW480 cells after exposure to BPA (Fig. S2). Furthermore, the results also showed that BPA can increase the protein expression of Fascin and TIF4A in both LoVo (p53 mutant, APC wt, and MSI+) and HCT-116 (p53 mutant, APC mutant, and MSI−) cells, suggesting that proteomic observations are representative in various colorectal cancer cells. Further studies showed that BPA can modulate the proliferation and promote metastasis of SW480 cells. Thus, together Western blotting and functional studies confirmed validity of the proteomic results. The five most represented functional activities for the modulated proteins are cell structure and motility (Krt 8 and fascin), cell cycle and proliferation (translation initiation factor), protein folding and metabolism (heat-shock cognate 71 kDa protein and elongation factor 1), oxidative stress (complex I and protein DJ-1), and energy metabolism (glucose-6-phosphate dehydrogenase and glutamate dehydrogenase 1) (Table S1). These functions are known to be essential in steroid signaling which involves fast 13 Arch Toxicol Fig. 5 Akt/GSK-3β-mediated BPA-induced EMT of colorectal cancer cells. a SW480 cells were treated with 10−8 or 10−5 M BPA for 96 h, and the phosphorylation and protein levels of Akt, GSK-3β, ERK1/2, and p38-MAPK were detected by Western blotting. SW480 cells (b) and HCT-116 cells (c) were pretreated with or without LY294002 (20 μM) for 1 h, followed by stimulation with or without BPA (10−5 M) for 96 h. The expression of Snail, E-Cad and the activation of AKT and GSK-3β were examined by Western blotting. d SW480 cells were pretreated with or without LY294002 (20 μM) for 1 h, followed by stimulation with or without BPA (10−5 M) for 96 h. Changes in morphology and expression of E-Cad were analyzed by a phase-contrast microscopy and immunofluorescence staining, respectively. Nuclei were visualized with DAPI staining nongenomic activities (including the transport and metabolism of signaling molecules) and genomic mechanisms mediated by receptors of environmental estrogens. Considering the functions of these proteins in colorectal cancer, the directions in which their expression modulated by BPA were confirmed by the following results that BPA can promote the metastasis of colorectal cancer cells. presented here revealed, for the first time, that BPA can promote the metastasis of colorectal cancer cells. EMT is considered to be the first step of invasion of adjacent tissues by tumors and metastasis. It occurs at the invasive front of colon carcinoma concomitant with a selective loss of basement membrane (Sheehan et al. 2008). Results of the present study revealed, for the first time, that oral equivalent or greater concentrations of BPA can induce EMT in SW480 cells, which resulted in down-regulation of epithelial characteristics, such as E-cad, ZO-1, up-regulation of mesenchymal characteristics FN, N-cad and Vim, and increased expression of transcription factors Snail and Twist (Fig. 4). Up-regulation of the mesenchymal biomarker Vim has also been found at in mammary glands of rat offspring exposed in utero to BPA at day 50 (Betancourt et al. 2010). Estradiol (E2) has been reported to enhance the EMT of lung adenocarcinoma through ERβ and regulate the expression of E-cad in breast cancer via ERα (Ye et al. 2010). These studies suggested that as an xenoestrogen, BPA can promote the metastasis of colorectal cancer via trigger the EMT of cancer cells. Furthermore, our study found that Snail mediated the BPA, particularly for 10−5 M BPA, induced EMT of colorectal cancer cells. Snail, a zinc finger transcription factor first identified in Drosophila, can bind to the E-box site BPA triggers metastasis and EMT of colorectal cancer via Akt/GSK‑3β/Snail Proteomic study revealed that many proteins related to cell structure and motility were significantly modulated by BPA. Exposure to both oral equivalent and greater concentrations of BPA can significantly promote migration and invasion ability of SW480 cells (Fig. 3). There is very limited published data about effects of EDCs on the metastasis of colorectal cancer. It has been reported that BPA can induce migration of breast cancer—associated fibroblasts (CAFs) and conditioned medium from BPA-treated CAFs can induce migration of SKBR3 cells (Pupo et al. 2012). In the cell migration and invasion assay, exposure to BPA or E2 resulted in greater motility or invasiveness in neuroblastoma SK–N–SH cells (Zhu et al. 2010). Our results 13 Arch Toxicol in the promoter of E-cad and trigger the EMT of many types of cancer (Kudo-Saito et al. 2009). Snail can directly inhibit transcription of the epithelial markers such as Krt-8 (Christiansen and Rajasekaran 2006), which is confirmed by the results of proteomic study (Krt 50.9 and 84.4 % of control, respectively). Furthermore, silencing of Snail by siRNA attenuated BPA-induced EMT of SW480 cells, which was not observed in control siRNA-transfected cells (Fig. 4). These results demonstrated that Snail is essential for BPA-induced EMT in CRC. In the present study, our results found that BPA can up regulate the expression of Snail via Akt/GSK-3β signal pathway. Phosphorylation of Akt has been reported to be associated with a loss of cell adhesion, decrease in cell– matrix adhesion, loss of apico-basolateral cell polarization and induction of cell motility (Bellacosa et al. 2005). Phosphorylation of Akt was increased and phenotypic changes associated with EMT has been suggested in mammary glands of 50-day-old rats exposed prenatally to BPA (Betancourt et al. 2010). The results of previous study revealed that stabilization of Snail mediated by AKT/GSK3β mediates EMT of colorectal cancer induced by TNFα (Wang et al. 2013). In the present study, BPA-induced EMT of both SW480 and HCT-116 cells, down-regulation of E-Cad, up-regulation of p-AKT, p-GSK-3β, and Snail, was blocked by treatment with the PI3K inhibitor, suggesting that PI3K/AKT signaling pathway plays an essential role for BPA-induced EMT of CRC cells. The functional proteomic study also identified other functional signaling molecules and pathways that may be also involved in the promotion effects of metastasis of SW480 cells. Annexin A1 and A2 are well-studied receptors for plasminogen, as it converts plasminogen to plasmin after binding. Accumulating evidences suggested that Annexin A1 and A2 and their receptor axis can significantly promote the tumor metastasis (Lokman et al. 2011). Results of the present study revealed that 10−8 and 10−5 M BPA resulted in significantly greater expression of proteins Annexin A1 and A2 (Table S1). This is the first evidence that Annexin can be up-regulated by BPA. Expression of the actin-bundling protein fascin was greater in cells exposed to BPA than that of control, which can result in actin reorganization and synaptic remodeling and therefore promoting the colorectal cancer invasion and migration (Vignjevic et al. 2007). BPA promotes tumorigenesis and progression of colorectal cancer The proteomic study revealed that levels of proteins related to cell cycle and proliferation were significantly modulated by BPA. Our previous studies revealed that micromolar BPA inhibited proliferation of Sertoli cells while nanomolar BPA stimulated proliferation of cells (Ge et al. 2014). The present study found that micromolar BPA inhibited cells proliferation while nanomolar BPA had limited effects on SW480 cells. This might be due to nanomolar and micromolar BPA had opposite effects on proteins related to cell proliferation of SW480 cells. For example, the small heat-shock protein 27 (hsp27) was increased by 10−8 M BPA (182 %) while decreased by 10−5 M BPA (93.2 %). Hsp27 has been documented to inhibit both the Fas- and mitochondria-mediated apoptosis pathways and over expressed in tumors (Mehlen et al. 1996). It suggested that effects of oral equivalent or greater concentrations of BPA might be different on proliferation of CRC cells, which was confirmed by cytotoxic tests. Other proteins related to cell proliferations were also significantly altered by treatment with BPA. Ran (Ras-related nuclear protein) is a small GTP-binding protein belonging to the Ras super family (Di Fiore et al. 2004). Over-expression of Ran caused by 10−5 M BPA might result in disruption of transport of crucial cell-cycle regulators or effectors and thus lesser proliferation of cells (Clarke and Zhang 2008). 14–3–3 is a family of highly conserved proteins that are crucial in regulating multiple cellular processes, including maintenance of cell-cycle checkpoints and repair of DNA, the onset of cell differentiation and senescence, and the coordination of cell adhesion and motility (Betancourt et al. 2011). Prenatal BPA exposure increased expression of 14–3–3 eta and increased susceptibility for mammary cancer (Betancourt et al. 2011). Up-regulation of 14–3–3 epsilon has also been found in rat offspring on postnatal day 50 following prenatal BPA exposure (Betancourt et al. 2010). In the present study, both oral equivalent and greater concentrations of BPA significantly elevated expression of 14–3–3, suggesting that BPA can promote the tumorigenesis of colorectal cancer. Collectively, the effects of BPA on proliferation of CRC cells and progression of colorectal cancer should be further investigated. BPA alters energy metabolism and increases antioxidant capacity Many proteins altered by exposure to BPA belong to the “Metabolism” group (Table S1). Regulation of cellular energetic pathways by BPA was evidenced by greater expression of enzymes of the glycolysis (glucose-6-phosphate dehydrogenase and glutamate dehydrogenase 1) and TCA cycle (aldehyde dehydrogenase family 1 member A3, malate dehydrogenase, and fumarate hydratase). On the other hand, proteins within the electron transport chain and complex I were increased in expression (complex I, ATP synthase subunit alpha, Human Mitochondrial Acetoacetyl-Coa Thiolase cytochrome b-c1 complex subunit). The result is consistent with increased glycolytic metabolic activity and suggests that energy consumption of colorectal cancer cells was covered from sources of glycolysis (Deberardinis et al. 2008). Metabolic activity 13 regulates the production of free radicals. Enhanced oxidative phosphorylation, and thus increased ATP synthesis via the respiratory chain, requires more oxygen and generates more free radicals, which can damage cells (Vander Heiden et al. 2009). In the present study, BPA, particularly 10−8 M BPA, also increased the protein levels of peroxiredoxin (PRDX-3), which naturalized the majority of the mitochondrial peroxides (Cox et al. 2010). It indicated that BPA is not only an oxidant hormone, but also induces synthesis of mitochondrial antioxidant proteins (Barros and Gustafsson 2011). Confirming our protein expression data, BPA did induce generation of ROS in liver of rat and resulted in greater expression of peroxiredoxin in mammary glands of offspring in the rat (Betancourt et al. 2010). BPA regulates protein folding, metabolism, and modification Heat-shock proteins (HSPs) play an important role in physiological processes, such as division, apoptosis, and differentiation of cells by interacting with multiple key components of signaling pathways that regulate growth and development. HSP70 has been previously identified as a potential target of BPA (Papaconstantinou et al. 2001). The proteomic study reported upon here also revealed that BPA significantly down-regulated HSP70 in SW480 cells. Results of previous studies indicated that HSP70 is a good marker for proliferation of colorectal cancer cells (Wang 2011), it was consistent with the fact that 10−5 M BPA inhibited proliferation of SW480 cells and down-regulated expression of HSP70. Greater expression of HSP27 protein has previously been reported in colon cancer cells and associated with poor prognosis (Thanner et al. 2005). The fact that exposure to the oral equivalent concentration of BPA resulted in significantly greater expression of HSP27 protein might indicate that low dose of BPA can also promote tumorigenesis and development of colorectal cancer. Acknowledgments This research was supported by the National Natural Science Foundation of China (Grant Nos. 31101071 and 81302317), the Fundamental Research Funds for the Central Universities (Sun Yat-sen University) (No. 12ykpy09), the Opening Project of Guangdong Provincial Key Laboratory of New Drug Design and Evaluation (No. 2011A060901014-007), the National Basic Research Program of China (973 Program, No. 2011CB9358003), the Science and Technology Planning Project of Guangdong Province, China (No. 2012B031500005), and the Seed Collaborative Research Fund from the State Key Laboratory in Marine Pollution (SCRF0003). References Andersson H, Brittebo E (2012) Proangiogenic effects of environmentally relevant levels of bisphenol A in human primary endothelial cells. Arch Toxicol 86(3):465–474 13 Arch Toxicol Barros RP, Gustafsson JA (2011) Estrogen receptors and the metabolic network. Cell Metab 14(3):289–299 Bellacosa A, Kumar CC, Di Cristofano A, Testa JR (2005) Activation of AKT kinases in cancer: implications for therapeutic targeting. Adv Cancer Res 94:29–86 Betancourt AM, Mobley JA, Russo J, Lamartiniere CA (2010) Proteomic analysis in mammary glands of rat offspring exposed in utero to bisphenol A. J Proteomic 73(6):1241–1253 Betancourt AM, Eltoum IA, Desmond RA, Russo J, Lamartiniere CA (2011) In utero exposure to bisphenol A shifts the window of susceptibility for mammary carcinogenesis in the rat. Environ Health Perspect 118(11):1614–1619 Braniste V, Jouault A, Gaultier E, Polizzi A, Buisson-Brenac C, Leveque M, Martin PG, Theodorou V, Fioramonti J, Houdeau E (2010) Impact of oral bisphenol A at reference doses on intestinal barrier function and sex differences after perinatal exposure in rats. Proc Natl Acad Sci 107(1):448–453 Brotons JA, Olea-Serrano MF, Villalobos M, Pedraza V, Olea N (1995) Xenoestrogens released from lacquer coatings in food cans. Environ Health Perspect 103(6):608–612 Cano A, Peinado H, Olmeda D (2007) Snail, ZEB and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7(6):415–428 Chen J, Iverson D (2012) Estrogen in obesity-associated colon cancer: friend or foe? Protecting postmenopausal women but promoting late-stage colon cancer. Cancer Cause Control 23(11):1767–1773 Christiansen JJ, Rajasekaran AK (2006) Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res 66(17):8319–8326 Clarke PR, Zhang C (2008) Spatial and temporal coordination of mitosis by Ran GTPase. Nat Rev Mol Cell Biol 9(6):464–477 Cox AG, Winterbourn CC, Hampton MB (2010) Mitochondrial peroxiredoxin involvement in antioxidant defence and redox signalling. Biochem J 425(2):313–325 Deberardinis RJ, Sayed N, Ditsworth D, Thompson CB (2008) Brick by brick: metabolism and tumor cell growth. Curr Opin Genet Dev 18(1):54–61 Dekant W, Voelkel W (2008) Human exposure to bisphenol A by biomonitoring: methods, results and assessment of environmental exposures. Toxicol Appl Pharmacol 228(1):114–134 Derouiche S, Warnier M, Mariot P, Gosset P, Mauroy B, Bonnal JL, Slomianny C, Delcourt P, Prevarskaya N, Roudbaraki M (2013) Bisphenol A stimulates human prostate cancer cell migration remodelling of calcium signalling. Springerplus 2(1):54 Di Fiore B, Ciciarello M, Lavia P (2004) Mitotic functions of the Ran GTPase network: the importance of being in the right place at the right time. Cell Cycle 3(3):305–313 Durando M, Kass L, Piva J, Sonnenschein C, Soto AM, Luque EH, Munoz-de-Toro M (2007) Prenatal bisphenol A exposure induces preneoplastic lesions in the mammary gland in Wistar rats. Environ Health Perspect 115(1):80–86 Ge LC, Chen ZJ, Liu HY, Zhang KS, Liu H, Huang HB, Zhang G, Wong CK, Giesy JP, Du J, Wang HS (2014) Involvement of activating ERK1/2 through G protein coupled receptor 30 and estrogen receptor alpha/beta in low doses of bisphenol A promoting growth of Sertoli TM4 cells. Toxicol Lett 226(1):81–89 Geens T, Aerts D, Berthot C, Bourguignon JP, Goeyens L, Lecomte P, Maghuin-Rogister G, Pironnet AM, Pussemier L, Scippo ML, Van Loco J, Covaci A (2012) A review of dietary and non-dietary exposure to bisphenol-A. Food Chem Toxicol 50(10):3725–3740 Jasperson KW, Tuohy TM, Neklason DW, Burt RW (2010) Hereditary and familial colon cancer. Gastroenterology 138(6):2044–2058 Jenkins S, Raghuraman N, Eltoum I, Carpenter M, Russo J, Lamartiniere CA (2009) Oral exposure to bisphenol a increases dimethylbenzanthracene-induced mammary cancer in rats. Environ Health Perspect 117(6):910–915 Arch Toxicol Jiang GM, Wang HS, Zhang F, Zhang KS, Liu ZC, Fang R, Wang H, Cai SH, Du J (2013) Histone deacetylase inhibitor induction of epithelial–mesenchymal transitions via up-regulation of Snail facilitates cancer progression 1833. BBA-Mol Cell Res 3:663–671 Kang JH, Kondo F, Katayama Y (2006) Human exposure to bisphenol A. Toxicology 226(2–3):79–89 Kennelly R, Kavanagh DO, Hogan AM, Winter DC (2008) Oestrogen and the colon: potential mechanisms for cancer prevention. Lancet Oncol 9(4):385–391 Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y (2009) Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 15(3):195–206 Le HH, Carlson EM, Chua JP, Belcher SM (2008) Bisphenol A is released from polycarbonate drinking bottles and mimics the neurotoxic actions of estrogen in developing cerebellar neurons. Toxicol Lett 176(2):149–156 Lokman NA, Ween MP, Oehler MK, Ricciardelli C (2011) The role of annexin A2 in tumorigenesis and cancer progression. Cancer Microenviron 4(2):199–208 Mehlen P, Schulze-Osthoff K, Arrigo AP (1996) Small stress proteins as novel regulators of apoptosis. Heat shock protein 27 blocks Fas/APO-1- and staurosporine-induced cell death. J Biol Chem 271(28):16510–16514 Papaconstantinou AD, Fisher BR, Umbreit TH, Goering PL, Lappas NT, Brown KM (2001) Effects of beta-estradiol and bisphenol A on heat shock protein levels and localization in the mouse uterus are antagonized by the antiestrogen ICI 182,780. Toxicol Sci 63(2):173–180 Pupo M, Pisano A, Lappano R, Santolla MF, De Francesco EM, Abonante S, Rosano C, Maggiolini M (2012) Bisphenol A induces gene expression changes and proliferative effects through GPER in breast cancer cells and cancer-associated fibroblasts. Environ Health Perspect 120(8):1177–1182 Salian S, Doshi T, Vanage G (2011) Perinatal exposure of rats to bisphenol A affects fertility of male offspring—an overview. Reprod Toxicol 31(3):359–362 Sheehan KM, Gulmann C, Eichler GS, Weinstein JN, Barrett HL, Kay EW, Conroy RM, Liotta LA, Petricoin EF 3rd (2008) Signal pathway profiling of epithelial and stromal compartments of colonic carcinoma reveals epithelial–mesenchymal transition. Oncogene 27(3):323–331 Sheng ZG, Zhu BZ (2011) Low concentrations of bisphenol A induce mouse spermatogonial cell proliferation by G protein-coupled receptor 30 and estrogen receptor-alpha. Environ Health Perspect 119(12):1775–1780 Siegel R, Naishadham D, Jemal A (2013) Cancer statistics, 2012. CA Cancer J Clin 62(1):10–29 Thanner F, Sutterlin MW, Kapp M, Rieger L, Morr AK, Kristen P, Dietl J, Gassel AM, Muller T (2005) Heat shock protein 27 is associated with decreased survival in node-negative breast cancer patients. Anticancer Res 25(3A):1649–1653 Vandenberg LN, Hauser R, Marcus M, Olea N, Welshons WV (2007a) Human exposure to bisphenol A (BPA). Reprod Toxicol 24(2):139–177 Vandenberg LN, Maffini MV, Wadia PR, Sonnenschein C, Rubin BS, Soto AM (2007b) Exposure to environmentally relevant doses of the xenoestrogen bisphenol-A alters development of the fetal mouse mammary gland. Endocrinology 148(1):116–127 Vandenberg LN, Chahoud I, Heindel JJ, Padmanabhan V, Paumgartten FJR, Schoenfelder G (2010) Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A. Environ Health Perspect 118(8):1055–1070 Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324(5930):1029–1033 Vignjevic D, Schoumacher M, Gavert N, Janssen KP, Jih G, Lae M, Louvard D, Ben-Ze’ev A, Robine S (2007) Fascin, a novel target of beta-Catenin-TCF signaling, is expressed at the invasive front of human colon cancer. Cancer Res 67(14):6844–6853 Vom Saal FS, Nagel SC, Coe BL, Angle BM, Taylor JA (2012) The estrogenic endocrine disrupting chemical bisphenol A (BPA) and obesity. Mol Cell Endocrinol 354(1–2):74–84 Wang RE (2011) Targeting heat shock proteins 70/90 and proteasome for cancer therapy. Curr Med Chem 18(27):4250–4264 Wang HS, Chen ZJ, Zhang G, Ou XL, Yang XL, Wong CKC, Giesy JP, Du J, Chen SY (2012) A novel micro-linear vector for in vitro and in vivo gene delivery and its application for EBV positive tumors. PLoS ONE 7(10):e47159 Wang H, Wang HS, Zhou BH, Li CL, Zhang F, Wang XF, Zhang G, Bu XZ, Cai SH, Du J (2013) Epithelial-mesenchymal transition (EMT) induced by TNF-alpha requires AKT/GSK-3betamediated stabilization of snail in colorectal cancer. PLoS ONE 8(2):e56664 Ye Y, Xiao Y, Wang W, Yearsley K, Gao J, Shetuni B, Barsky S (2010) ERα signaling through slug regulates E-cadherin and EMT. Oncogene 29(10):1451–1462 Zhu H, Zheng J, Xiao X, Zheng S, Dong K, Liu J, Wang Y (2010) Environmental endocrine disruptors promote invasion and metastasis of SK–N–SH human neuroblastoma cells. Oncol Rep 23(1):129–139 13