Biosci. Rep. (2016) / 36 / art:e00389 / doi 10.1042/BSR20160053

Silencing of SOX12 by shRNA suppresses migration, invasion and proliferation of breast cancer cells Hanzhi Ding*, Hong Quan*, Weiguo Yan* and Jing Han*1 *Department of Breast Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China

Synopsis Sex determining region Y-box protein 12 (SOX12) is essential for embryonic development and cell-fate determination. The role of SOX12 in tumorigenesis of breast cancer is not well-understood. Here, we found that SOX12 mRNA expression was up-regulated in human breast cancer tissues. To clarify the roles of SOX12 in breast cancer, we used lentiviral shRNAs to suppress its expression in two breast cancer cells with relatively higher expression of SOX12 (BT474 and MCF-7). Our findings strongly suggested that SOX12 was critical for cell migration and invasion of breast cancer cells. We found that silencing of SOX12 significantly decreased the mRNA and protein levels of MMP9 and Twist, while notably increased E-cadherin. Moreover, SOX12 knockdown significantly inhibited the proliferation of breast cancer cells in vitro and the growth of xenograft tumours in vivo. Flow cytometry analysis revealed that breast cancer cells with SOX12 knockdown showed cell cycle arrest and decreased mRNA and protein levels of proliferating cell nuclear antigen (PCNA), CDK2 and Cyclin D1. Taken together, SOX12 plays an important role in growth inhibition through cell-cycle arrest, as well as migration and invasion of breast cancer cells. Key words: breast cancer, cell cycle arrest, invasion, migration, proliferation, SOX12. Cite this article as: Bioscience Reports (2016) 36, e00389, doi:10.1042/BSR20160053

INTRODUCTION Breast cancer is one of the most common and important malignant tumours affecting women. Despite the advances in screening, prevention, surgical resection and adjuvant therapies, breast cancer still remains the leading cause of death in women worldwide, accounting 23 % of all cancer deaths [1]. Metastasis of breast cancer to other organs is the major cause of breast cancer deaths [2,3]. Consequently, there is a disquieting need to identify novel diagnosing marker and effective therapeutic targets for breast cancer. Sex determining region Y-box protein 12 (SOX12) is a member of SOX genes, which encode a family of transcription factors belonging to the high mobility group (HMG) superfamily. SOX family proteins are important for embryonic developmental processes [4]. Disruption of SOX genes has been involved in various human diseases including cancer [5,6]. SOX family members may act as oncogenes or tumour suppressor genes. It is reported

that SOX2 [7,8], SOX5 [9–11], SOX9 [12–14] and SOX18 [15] act as oncogenes in multiple types of human cancers through promoting cell proliferation, migration and invasion. On the contrast, SOX1 [16], SOX6 [17] and SOX7 [18,19] serves as a tumour suppressor gene to suppress cell proliferation and metastasis. A previous study has reported the critical role of SOX12 in embryonic development and cell-fate determination [20]. Recently, Huang et al. [21] demonstrated that SOX12 can promote invasion and metastasis of hepatocellular carcinoma (HCC). However, the expression and function of SOX12 in breast cancer remains unknown. In this work, we presented the first evidence that SOX12 was overexpressed in breast cancer. We also investigated the potential role of SOX12 in breast cancer by knocking down its expression in two breast cancer cell lines. SOX12 knockdown inhibited migration and invasion of breast cancer cells. SOX12 knockdown also suppressed cell proliferation in vitro and growth of xenograft breast tumours in vivo. Together, our data strongly suggest the important role of SOX12 in the tumorigenesis and development of breast cancer.

.................................................................. ............................................................. ................................................................. .............................................................. ..............................................

Abbreviations: DMEM, Dulbecco’s minimum essential medium; EMT, epithelial–mesenchymal transition; HCC, hepatocellular carcinoma; HE, haematoxylin and eosin; OD, optical density; PCNA, proliferating cell nuclear antigen; PI, propidium iodide; SOX12, sex determining region Y-box protein 12; WT, wild-type. 1 To whom correspondence should be addressed (email [email protected]).

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

1

H. Ding and others

MATERIALS AND METHODS Patient samples From June 2009 to July 2010, 35 patients with breast cancer (6 in AJCC stage I, 20 in stage II and 9 in stage III) admitted to Department of Breast Surgery, Shanghai East Hospital (Shanghai, China) with written informed consent were selected in this study. The patients had not received chemotherapy or radiation before surgery. The median age of the participants was 58 years (ranging from 35 to 78 years). The diagnosis of each case was independently confirmed by two pathologists. Human breast cancer and paired normal adjacent tissues (>2 cm from cancer tissue) were resected, snap frozen and then stored at -80 ◦ C until further use. This study was approved by the ethics committee of Shanghai East Hospital (Shanghai, China).

Cell culture HEK-293T and human breast cancer cell lines, MDA-MB-231, MCF-7, SK-BR-3, BT474, ZR-75-1 and ZR-75-30 were obtained from A.T.C.C. All cells were maintained in Dulbecco’s minimum essential medium (DMEM) (Life Technologies) supplemented with 10 % FBS (Hyclone), 100 units/ml penicillin and 100 μg/ml streptomycin at 37 ◦ C in a 5 % CO2 atmosphere.

GATGAAA, shSOX12-1) and 4616–4638 (CAGGTGCTTATTCCTAATA, shSOX12-2) of human SOX12 and a nonspecific scramble shRNA sequence (CCTAAGGTTAAGTCGCCCTCG, shNC) were constructed. The lentivirus particles were produced in HEK-293T cells transfected with psPAX2, PMD2G (Addgene) and pLKO-shRNA-containing plasmids. BT474 cells were infected with the lentivirus and stable cells were established by puromycin selection (Sigma).

Western blotting and quantification Cells were lysed in RIPA Lysis buffer [50 mM Tris/HCl, pH 8.0, 250 mM NaCl, 1 % NP40, 0.5 % (w/v) sodium deoxycholate, 0.1 % sodium dodecylsulfate] supplemented with protease inhibitors (Roche). Whole-cell extracts containing 40 μg protein were electrophoresed on sodium dodecyl sulfate polyacrylamide (SDS-PAGE) gels, then transferred to polyvinylidene difluoride membranes (Millipore) and blocked in TBST-T with 5 % skim milk. Anti-E-cadherin, proliferating cell nuclear antigen (PCNA), CDK2, Cyclin D1 and anti-GAPDH (Cell Signaling Technology) were used at 1:1000 dilution with overnight incubation at 4 ◦ C in TBS-T with 5 % BSA. Anti-SOX12, Twist and MMP9 antibodies (Abcam) were used at 1:1000 dilution. After incubation with a horseradish peroxidase (HRP)-linked secondary antibody (Beyotime), blots were detected with Clarity Western ECL kit (Bio-Rad Laboratories). Band intensities were quantified by using ImageJ software (http://rsb.info.nih.gov/ij/).

RNA preparation and real-time PCR Total RNA was prepared from the tissue samples or breast cancer cell lines by using TRIzol reagent (Invitrogen). First-strand cDNA was synthesized from total RNA with the M-MLV reverse transcriptase (Promega) following the manufacturer’s manual. Quantitative real-time PCR was performed using the SYBR Green mix (Thermo Fisher Scientific) on the ABI Prism 7300 sequence detection system (Applied Biosystems). Primer sequences were 5’-AGCACCCGTGTGACTCTTTCC-3’ and 5’-AGCAGAACCAAGCCCTGTCTC-3’ for SOX12; 5’-AAGG GCGTCGTGGTTCCAACTC-3’ and 5’-AGCATTGCCGTCC TGGGTGTAG-3’ for MMP9; 5’-AGTCCGCAGTCTTACGAG -3’ and 5’-GCTTGCCATCTTGGAGTC-3’ for Twist1; 5’-GAG AACGCATTGCCACATACAC-3’ and 5’-AAGAGCACCTTCC ATGACAGAC-3’ for E-cadherin; 5’-GGTGTTGGAGGCACT CAAGG-3’ and 5’-CAGGGTGAGCTGCACCAAAG-3’ for PCNA; 5’-AACAAGTTGACGGGAGAG-3’ and 5’-AAGAGG AATGCCAGTGAG-3’ for CDK2; 5’-GCTGCGAAGTGGAA ACCATC-3’ and 5’-CCTCCTTCTGCACACATTTGAA-3’ for Cyclin D1; 5’-CACCCACTCCTCCACCTTTG-3’ and 5’CCACCACCCTGTTGCTGTAG-3’ for GAPDH. The relative expression of the genes was calculated using the comparative cycle threshold (CT ) method (2−CT ) [22].

shRNA constructs and lentiviral infections For SOX12 knockdown, pLKO-1 plasmid containing shRNAtargeting targeting position 3728–3750 (GCTGCTTCACAG-

Transwell assay Effect of SOX12 knockdown on cell motility and invasion were quantified by Transwell chamber (Costar). For cell invasion assay, the filters were pre-coated with Matrigel. Briefly, BT474 or MCF7 cells were infected with shSOX12-1, shSOX12-2 or shNC virus and cultured at 37 ◦ C for 24 h. After starving in FBS-free DMEM medium overnight, cells in in serum-free medium were added into 24-well inserts (2 × 104 cells per well). DMEM medium containing 10 % FBS was added to the lower well of the chambers as chemoattractant. After 24 h, the cells on the upper surface of the filter were completely removed and migrated cells were fixed with 4 % paraformaldehyde and stained with 0.5 % crystal violet. Cells traversing the membrane were counted in 10 random fields under a microscope.

Cell proliferation assays Effect of SOX12 knockdown on cell proliferation was measured by using CCK-8 (Dojindo Laboratories). Briefly, BT474 or MCF7 cells were plated on to 96-well plates at density of 3×103 cells per well. After cultured overnight, cells were infected with the shSOX12-1, shSOX12-2 virus or control virus (shNC). At 0 h, 24 h, 48 h and 72 h after viral infection, CCK-8 solution was added to every well and incubated at 37 ◦ C for 1 h. Absorbance (A) values were determined at wavelength 450 nm (A450 ) on a microplate reader (Bio-Rad Laboratories).

..........................................................................................................................................................................................................................................................................................................................................................................

2

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

Role of SOX12 in breast cancer

Figure 1

SOX12 is overexpressed in breast cancer. Real-time PCR analysis of SOX12 mRNA levels in 35 pairs of breast cancer and normal tissues. Positive log 2 (Tumour/Normal) on the y-axis indicated the elevated expression of SOX12 in tumour tissue whereas negative log 2 indicated the reduced expression of SOX12 in tumour tissue.

Tumour xenografts Four to five-week-old female BALB/c nude mice were purchased from Shanghai Laboratory Animal Company and maintained under standard conditions in line with the NIH Guide for the Care and Use of Laboratory Animals. BT474 cells stably infected with shNC or shSOX12-1 were collected, resuspended in PBS and injected subcutaneously into the right flank of each mouse (2 × 104 per mouse). Tumour xenografts were measured with calipers every 3 days, and tumour volume was determined using the following formula: (length × width2 ) × 0.5. At the end of the experiment, after anesthetized with sodium pentobarbital, the mice were killed by cervical dislocation and tumour xenografts were recovered. A portion of tissues were fixed in formalin, processed to obtain sections of 5 μm and stained with using haematoxylin and eosin (HE) or PCNA (Abcam) immunohistochemically. The other portion of tissues was employed for protein extraction and Western blotting analysis to examine SOX12 expression. All experiments were approved by the Committee on the Ethics of Animal Experiments of Shanghai East Hospital (Shanghai, China).

FACS analysis BT474 or MCF-7 cells were infected with shSOX12-1, shSOX122 or shNC virus and cell cycle distribution was monitored 48 h after viral infection. The cells were collected, washed with PBS and fixed in ice-cold ethanol at -20 ◦ C overnight. The cells were washed with PBS and resuspended in PBS buffer containing RNase A (1 mg/ml, Sigma) at 37 ◦ C for 30 min. The cells were then stained with propidium iodide (PI, 60 μg/ml, Sigma) and analysed for DNA content with a flow cytometer (Accuri C6, BD).

RESULTS SOX12 is overexpressed in breast cancer compared with normal breast tissue To assess SOX12 expression in breast cancer, real-time RT-PCR was used to evaluate SOX12 expression level in 35 breast cancer tissues and paired normal tissues. As shown in Figure 1, SOX12 expression was significantly increased in 94 % (33/35) of breast cancer tissues when compared with normal tissues.

Knockdown of SOX12 in breast cancer cells To investigate the role of SOX12 in breast cancer, the mRNA levels of SOX12 were evaluated in six human breast cancer cell lines, including MDA-MB-231, MCF-7, SK-BR-3, BT474, ZR-75-1 and ZR-75-30, by performing real-time PCR analysis (Figure 2A). The expression levels of SOX12 were much higher in MCF-7 and BT474 cells than in other four cell lines. The higher expression level of SOX12 was confirmed at the protein level by Western blot analysis in both MCF-7 and BT474 cells (Figure 2B), which were selected for the following experiments. To study the role of SOX12 on breast tumorigenesis, SOX12shRNA lentivirus (shSOX12-1 and shSOX12-2) and non-specific scramble shRNA lentivirus (shNC) were produced and infected BT474 and MCF-7 cells, which have high endogenous SOX12 levels. At 48 h after lentiviral infection, mRNA and protein levels of SOX12 were measured. As shown in Figures 2(C) and 2(D), shSOX12 viral infection significantly decreased SOX12 expression at both mRNA and protein levels compared with those of wild-type (WT) and shNC lentivirus infected cells.

Statistics All statistics were calculated using GraphPad Prism, version 6.0 (GraphPad). All in vitro experiments assays were performed in triplicate, and mean values and S.D. were reported. Differences between groups were determined by Student’s t test. P less than 0.05 was considered statistically significant.

SOX12 knockdown inhibits migration and invasion of breast cancer cells Huang et al. [21] have demonstrated the promoted effects of SOX12 on the invasion and metastasis of HCC. To investigate the effects of SOX12 on the invasion and migration capacities

..........................................................................................................................................................................................................................................................................................................................................................................

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

3

H. Ding and others

Figure 2

Knockdown of SOX12 in breast cancer cells. (A) The mRNA expression levels of SOX12 in six human breast cancer cells were determined by real-time PCR and normalized with GAPDH. (B) The protein levels of SOX12 were determined in human breast cancer cells by Western blot. GAPDH was used as the loading control. (C) Knockdown of SOX12 in BT474 and MCF-7 cells detected by real-time PCR. (D) Knockdown of SOX12 in BT474 and MCF-7 cells detected by Western blot. Upper panels: representative images; lower panels: quantifications of Western blot. WT: wild-type cells; shNC: cells infected with scrambled shRNA virus; shSOX12-1: cells infected with shSOX12-1 virus; shSOX12-2: cells infected with shSOX12 virus. ** P < 0.01 and *** P < 0.001 compared with shNC.

of breast cancer cells, Transwell assays were performed. As shown in Figures 3(A) and 3(B), infection with shSOX12-1 and shSOX12-2 virus in both BT474 and MCF-7 cells notably reduced the abilities of invasion and migration compared with WT and shNC cells, whereas no significant difference was observed in the invasive capability between WT and NC cells. In addition, protein levels of epithelial–mesenchymal transition (EMT)-related proteins, which contribute to metastasis of tumour cells, were evaluated by Western blot. Infection with shSOX12-1 and shSOX12-2 virus significantly decreased the protein levels of MMP9 and Twist, while notably increased the main factor of EMT, E-cadherin (Figures 3C and 3D). These

results demonstrated that SOX12 promoted the abilities of invasion and migration of breast cancer cells.

Knockdown of SOX12 suppresses the proliferation of breast cancer cells in vitro and in vivo In addition to promoting metastasis, we investigate the effect of SOX12 on cell proliferation in breast cancer cells by CCK-8 assay. Knockdown of endogenous SOX12 significantly inhibited the growth of BT474 cells (Figure 4A) and MCF-7 (Figure 4B) at 24 h, 48 h and 72 h after viral infection. Consistent with the results obtained from in vitro assays, knockdown of endogenous

..........................................................................................................................................................................................................................................................................................................................................................................

4

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

Role of SOX12 in breast cancer

Figure 3

SOX12 knockdown inhibited the migration and invasion ability of breast cancer cells. (A) SOX12 knockdown inhibited the migration ability of BT474 and MCF-7 cells as determined by Transwell assays in chambers without Matrigel. Left panels: representative images; right panels: quantifications of average migrated cell number. (B) SOX12 knockdown repressed the invasion ability of BT474 and MCF-7 cells as determined by Transwell assays in chambers coated with Matrigel. Left panels: representative images; right panels: quantifications of average migrated cell number. (C and D) The levels of MMP9, Twist and E-cadherin were examined with real-time PCR and Western blot. GAPDH was used as internal control. Left panels: real-time PCR results; middle panels: representative images of Western blot; right panels: quantifications of Western blot. WT: wild-type cells; shNC: cells infected with scrambled shRNA virus; shSOX12-1: cells infected with shSOX12-1 virus; shSOX12-2: cells infected with shSOX12 virus. *P < 0.05, ** P < 0.01 and *** P < 0.001 compared with shNC.

..........................................................................................................................................................................................................................................................................................................................................................................

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

5

H. Ding and others

Figure 4

Knockdown of SOX12 suppressed the proliferation of breast cancer cells in vitro and in vivo. (A and B) Knockdown of endogenous SOX12 inhibited the growth of BT474 and MCF-7 cells. (C–F) SOX12 knockdown reduced the growth rate of xenograft breast tumours. BALB/c nude mice were inoculated with BT474 cells stably infected with shNC or shSOX12 virus (n = 6). The tumour size was monitored every 3 days and growth curves of xenograft tumours were shown in (C). At 33 days after inoculation, the mice were killed and the tumours were recovered. Images of and weights of xenograft tumours were shown in (D). The expression of SOX12 in xenograft from the nude mice was determined by Western blot (E). Representative images of HE staining and PCNA immunohistochemical staining of xenograft tumours were shown in (F). ** P < 0.01 and *** P < 0.001 compared with shNC.

SOX12 reduced the growth of BT474 xenograft tumours (Figures 4C and 4D). Protein levels of SOX12 were significantly lower in the xenografts formed from shSOX12-1 stable cells than in those from shNC stable cells (Figure 4E). Further, immunohistochemical staining revealed that the PCNA-positive

cells were significantly reduced in the xenografts formed from shSOX12-1 stable cells compared with those from shNC cells (Figure 4F). Together, these results demonstrated that SOX12 shRNA inhibited the proliferation of breast cancer cells and the growth of xenograft breast tumours.

..........................................................................................................................................................................................................................................................................................................................................................................

6

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

Role of SOX12 in breast cancer

Figure 5

SOX12 knockdown induced G1 arrest. BT474 and MCF-7 cells were infected with shSOX12 or shNC virus. (A and B) At 48 h after viral infection, cell cycle distribution of BT474 and MCF-7 cells infected with shSOX12 or shNC virus. (C and D) At 48 h after viral infection, the levels of PCNA, CDK2 and Cyclin D1 were examined with Western blot. GAPDH was used as the loading control. Left panels: real-time PCR results; middle panels: representative images of Western blot; right panels: quantifications of Western blot. WT: wild-type cells; shNC: cells infected with scrambled shRNA virus; shSOX12: cells infected with shSOX12 virus. ** P < 0.01 and *** P < 0.001 compared with shNC.

..........................................................................................................................................................................................................................................................................................................................................................................

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

7

H. Ding and others

Knockdown of SOX12 induces G1-arrest in breast cancer cell lines Then we assessed the effect of SOX12 on the cell cycle of breast cancer cells by flow cytometry analysis. The results show that infection with shSOX12-1 virus caused an accumulation of BT474 and MCF-7 cells at the G0/G1 phase, whereas the percentages of cells in S phase and G2/M phase decreased (Figures 5A and 5B). Moreover, the protein levels of G1/S transition-related proteins were also evaluated by Western blot. Infection with shSOX12-1 virus significantly decreased the protein levels of PCNA, CDK2 and Cyclin D1 (Figures 5C and 5D). These results indicated that SOX12 knockdown contributed to induction of G1-arrest in breast cancer cells.

DISCUSSION SOX12 plays an essential role in embryonic development and cell-fate determination [20]. A recent study has suggested SOX12 may play a role in the tumorigenesis of HCC [21], whereas the association between SOX12 and breast cancer has not been reported. Our results showed that SOX12 mRNA expression was elevated in breast cancer tissues (Figure 1), suggesting that SOX12 may also serve as an oncogene in breast cancer. Metastasis to vital organs is the major cause of mortality from breast cancer [2,3]. Cell migration and invasion are indispensable processes for the metastasis of cancer. In the present study, Transwell assays showed that SOX12 contributed to breast cancer cell migration and invasion, as a reduction in SOX12 protein by RNAi caused significant decreases in the migration and invasion of BT474 and MCF-7 cells (Figures 3A and 3B). Further, the EMT is implicated in promoting carcinoma invasion and metastasis [23,24]. Here, shSOX12 viral infection enhanced the expression of the main factor of EMT (E-cadherin [25]), but decreased the expression of a known inducer of EMT (Twist [26]) (Figures 3C and 3D). The mRNA levels of E-cadherin and Twist were also changed after SOX12 knockdown, indicating that SOX12 may bind to the promoters of these EMT-regulating factors to control their transcription. It has been reported that SOX12 promotes migration and invasion of HCC through upregulating Twist1 and FGFBP1 [21]. Together with the study on HCC, our data suggested that SOX12 may prompt cell invasion through inducing EMT. Previous studies have shown the role of other members of SOX family in the proliferation of tumour cells [12,15,16,19]. Results from the current study demonstrated that knockdown of SOX12 inhibited the proliferation of breast cancer cells in vitro and in vivo (Figure 4). Further, we demonstrated that shSOX12 viral infection significantly decreased mRNA and protein levels of PCNA, CDK2 and Cyclin D1. These results are consistent with the increased percentages of G1 phase shown in the shSOX12-infected cells (Figure 5). SOX12 may bind to the promoter regions of the above genes to influence their transcription, although further investigation is required to verify this hypothesis.

In summary, we revealed that SOX12 expression was upregulated in breast cancer tissues. Moreover, we showed that SOX12 plays an important role in promoting growth, migration and invasion of breast cancer cells. This study suggested that SOX12 may be an important biomarker for breast cancer and may lead to new therapeutic approaches for breast cancer. AUTHOR CONTRIBUTION

Jing Han conceived and designed the experiments. Hanzhi Ding, Hong Quan and Weiguo Yan performed the experiments. Hanzhi Ding analysed the data. Hanzhi Ding and Jing Han wrote the paper.

FUNDING

This work was supported by funds from Research Center for Medical Health, Science and Technology at Ministry of Public Health [grant number W2012FZ042].

REFERENCES 1

Donepudi, M.S., Kondapalli, K., Amos, S.J. and Venkanteshan, P. (2014) Breast cancer statistics and markers. J. Cancer Res. Ther. 10, 506–511 PubMed 2 Culhane, A.C. and Quackenbush, J. (2009) Confounding effects in “A six-gene signature predicting breast cancer lung metastasis”. Cancer Res 69, 7480–7485 CrossRef PubMed 3 Chiang, A.C. and Massague, J. (2008) Molecular basis of metastasis. N. Engl. J. Med. 359, 2814–2823 CrossRef PubMed 4 Lefebvre, V., Dumitriu, B., Penzo-Mendez, A., Han, Y. and Pallavi, B. (2007) Control of cell fate and differentiation by Sry-related high-mobility-group box (Sox) transcription factors. Int. J. Biochem. Cell Biol. 39, 2195–2214 CrossRef PubMed 5 Castillo, S.D. and Sanchez-Cespedes, M. (2012) The SOX family of genes in cancer development: biological relevance and opportunities for therapy. Exp. Opin. Ther. Targets 16, 903–919 CrossRef 6 Dong, C., Wilhelm, D. and Koopman, P. (2004) Sox genes and cancer. Cytogenet. Genome Res. 105, 442–447 CrossRef PubMed 7 Bass, A.J., Watanabe, H., Mermel, C.H., Yu, S., Perner, S., Verhaak, R.G., Kim, S.Y., Wardwell, L., Tamayo, P., Gat-Viks, I. et al. (2009) SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat. Genet. 41, 1238–1242 CrossRef PubMed 8 Rudin, C.M., Durinck, S., Stawiski, E.W., Poirier, J.T., Modrusan, Z., Shames, D.S., Bergbower, E.A., Guan, Y., Shin, J., Guillory, J. et al. (2012) Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat. Genet. 44, 1111–1116 CrossRef PubMed 9 Huang, D.Y., Lin, Y.T., Jan, P.S., Hwang, Y.C., Liang, S.T., Peng, Y., Huang, C.Y., Wu, H.C. and Lin, C.T. (2008) Transcription factor SOX-5 enhances nasopharyngeal carcinoma progression by down-regulating SPARC gene expression. J. Pathol. 214, 445–455 CrossRef PubMed 10 Pei, X.H., Lv, X.Q. and Li, H.X. (2014) Sox5 induces epithelial to mesenchymal transition by transactivation of Twist1. Biochem. Biophys. Res. Commun. 446, 322–327 CrossRef PubMed 11 Wang, D., Han, S., Wang, X., Peng, R. and Li, X. (2015) SOX5 promotes epithelial-mesenchymal transition and cell invasion via regulation of Twist1 in hepatocellular carcinoma. Med. Oncol. 32, 461 PubMed

..........................................................................................................................................................................................................................................................................................................................................................................

8

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

Role of SOX12 in breast cancer

12 Wang, L., He, S., Yuan, J., Mao, X., Cao, Y., Zong, J., Tu, Y. and Zhang, Y. (2012) Oncogenic role of SOX9 expression in human malignant glioma. Med. Oncol. 29, 3484–3490 CrossRef PubMed 13 Matheu, A., Collado, M., Wise, C., Manterola, L., Cekaite, L., Tye, A.J., Canamero, M., Bujanda, L., Schedl, A., Cheah, K.S. et al. (2012) Oncogenicity of the developmental transcription factor Sox9. Cancer Res. 72, 1301–1315 CrossRef PubMed 14 Kopp, J.L., von Figura, G., Mayes, E., Liu, F.F., Dubois, C.L., Morris, J.P., Pan, F.C., Akiyama, H., Wright, C.V., Jensen, K. et al. (2012) Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell 22, 737–750 CrossRef PubMed 15 Wu, Z., Liu, J., Wang, J. and Zhang, F. (2016) SOX18 knockdown suppresses the proliferation and metastasis, and induces the apoptosis of osteosarcoma cells. Mol. Med. Rep. 13, 497–504 PubMed 16 Tsao, C.M., Yan, M.D., Shih, Y.L., Yu, P.N., Kuo, C.C., Lin, W.C., Li, H.J. and Lin, Y.W. (2012) SOX1 functions as a tumor suppressor by antagonizing the WNT/beta-catenin signaling pathway in hepatocellular carcinoma. Hepatology 56, 2277–2287 CrossRef PubMed 17 Qin, Y.R., Tang, H., Xie, F., Liu, H., Zhu, Y., Ai, J., Chen, L., Li, Y., Kwong, D.L., Fu, L. and Guan, X.Y. (2011) Characterization of tumor-suppressive function of SOX6 in human esophageal squamous cell carcinoma. Clin. Cancer Res. 17, 46–55 CrossRef PubMed 18 Stovall, D.B., Cao, P. and Sui, G. (2014) SOX7: from a developmental regulator to an emerging tumor suppressor. Histol. Histopathol. 29, 439–445 PubMed

19 Stovall, D.B., Wan, M., Miller, L.D., Cao, P., Maglic, D., Zhang, Q., Stampfer, M.R., Liu, W., Xu, J. and Sui, G. (2013) The regulation of SOX7 and its tumor suppressive role in breast cancer. Am. J. Pathol. 183, 1645–1653 CrossRef PubMed 20 Hoser, M., Potzner, M.R., Koch, J.M., Bosl, M.R., Wegner, M. and Sock, E. (2008) Sox12 deletion in the mouse reveals nonreciprocal redundancy with the related Sox4 and Sox11 transcription factors. Mol. Cell. Biol. 28, 4675–4687 CrossRef PubMed 21 Huang, W., Chen, Z., Shang, X., Tian, D., Wang, D., Wu, K., Fan, D. and Xia, L. (2015) Sox12, a direct target of FoxQ1, promotes hepatocellular carcinoma metastasis through up-regulating Twist1 and FGFBP1. Hepatology 61, 1920–1933 CrossRef PubMed 22 Schefe, J.H., Lehmann, K.E., Buschmann, I.R., Unger, T. and Funke-Kaiser, H. (2006) Quantitative real-time RT-PCR data analysis: current concepts and the novel “gene expression’s CT difference” formula. J. Mol. Med. (Berl.) 84, 901–910 CrossRef PubMed 23 Guarino, M., Rubino, B. and Ballabio, G. (2007) The role of epithelial-mesenchymal transition in cancer pathology. Pathology 39, 305–318 CrossRef PubMed 24 Yang, J. and Weinberg, R.A. (2008) Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev. Cell 14, 818–829 CrossRef PubMed 25 Thiery, J.P. and Sleeman, J.P. (2006) Complex networks orchestrate epithelial-mesenchymal transitions. Nat. Rev. Mol. Cell Biol. 7, 131–142 CrossRef PubMed 26 Yang, J., Mani, S.A., Donaher, J.L., Ramaswamy, S., Itzykson, R.A., Come, C., Savagner, P., Gitelman, I., Richardson, A., Weinberg, R.A. et al. (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117, 927–939 CrossRef PubMed

Received 22 February 2016/19 August 2016; accepted 26 August 2016 Accepted Manuscript online 31 August 2016, doi 10.1042/BSR20160053

..........................................................................................................................................................................................................................................................................................................................................................................

c 2016 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution  Licence 4.0 (CC BY).

9

Silencing of SOX12 by shRNA suppresses migration, invasion and proliferation of breast cancer cells.

Sex determining region Y-box protein 12 (SOX12) is essential for embryonic development and cell fate determination. The role of SOX12 in tumorigenesis...
1MB Sizes 1 Downloads 11 Views