Open Access

SIX1 coordinates with TGFβ signals to induce epithelial-mesenchymal transition in cervical cancer

  • Authors:
    • Shu‑Hua Sun
    • Dan Liu
    • Yun‑Te Deng
    • Xiao‑Xue Zhang
    • Dong‑Yi Wan
    • Bi‑Xin Xi
    • Wei Huang
    • Qian Chen
    • Meng‑Chen Li
    • Ming‑Wei Wang
    • Fei Yang
    • Ping Shu
    • Ke‑Zhi Wu
    • Qing‑Lei Gao
  • View Affiliations

  • Published online on: June 28, 2016     https://doi.org/10.3892/ol.2016.4797
  • Pages: 1271-1278
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epithelial-mesenchymal transition (EMT) plays a critical role in promoting tumor invasion and metastasis. However, the key cofactors that modulate the signal transduction to induce EMT have note been fully explored to date. The present study reports that sine oculis homeobox homolog 1 (SIX1) is able to promote EMT of cervical cancer by coordinating with transforming growth factor (TGF)β-SMAD signals. The expression of SIX1 was negatively correlated with the expression of the epithelial marker E-cadherin in two independent groups of cervical cancer specimens. SIX1 could promote the transition of mesenchymal phenotype in the presence of active TGFβ signals in vitro and in vivo. TGFβ-SMAD signals were required for the SIX1‑mediated promotion of EMT and metastatic capacity of cervical cancer cells. Together, SIX1 and TGFβ cooperated to induce more remarkable changes in the transition of phenotype than each of them alone, and coordinated to promote cell motility and tumor metastasis in cervical cancer. These results suggest that the coordination of SIX1 and TGFβ signals may be crucial in the EMT program, and that SIX1/TGFβ may be considered a valuable marker for evaluating the metastatic potential of cervical cancer cells, or a therapeutic target in the treatment of cervical cancer.

Introduction

The transdifferentiation of epithelial cells into motile mesenchymal cells, a developmental program termed epithelial-mesenchymal transition (EMT), has been demonstrated to play a critical role in promoting tumor invasion and metastasis (1,2). The functional hallmark of the EMT program is to endow stationary epithelial cells with migratory and invasive ability (2). Thus, the transition into a mesenchymal phenotype is considered as a marker of metastatic potential in numerous epithelium-derived carcinomas, including cervical cancer (3,4).

The EMT program is triggered and controlled by multiple signaling pathways that are activated in response to extracellular stimuli (5,6). Among these, the transforming growth factor (TGF)β signaling pathway has a predominant role in promoting epithelial plasticity that may progress to EMT (68). However, a large number of non-invasive tumor cells did not undergo TGFβ-induced EMT in vitro (9), suggesting that TGFβ may not be able to efficiently activate signaling pathways in non-invasive tumor cells. It has been previously observed that the effect of TGFβ1 may be enhanced by other factors that could cooperate with TGFβ1 to induce sufficient activation of multiple signaling pathways (10). While extracellular factors may cooperate with TGFβ (7,8), it remains unclear whether the increased expression of transcription factors in tumor cells could increase the sensitivity of tumor cells to TGFβ stimulation and promote TGFβ-induced EMT.

Sine oculis homeobox homolog 1 (SIX1) is a transcription factor associated with development that is rarely expressed in the majority of adult tissues (11,12). However, overexpression of SIX1 has been observed in various malignancies, and frequently correlates with poor clinical prognosis (1214). In cervical cancer, SIX1 is induced by the E7 oncoprotein of human papillomavirus, and is closely associated with tumorigenesis of cervical epithelium (15). Increased expression of SIX1 could promote tumor growth by accelerating the cell cycle process (15), and enhance the metastatic potential of cervical cancer cells by upregulating the expression of α5β1 integrin (16). Importantly, the present authors recently reported that SIX1 could amplify TGFβ signals, interacting with SMAD2/3 proteins to regulate gene expression (17). Based on these findings, the present study investigated whether SIX1 and TGFβ were responsible for modulating the EMT program in cervical cancer. The data obtained indicated that SIX1 could coordinate with TGFβ to induce EMT, thus enhancing the metastatic capacity of cervical cancer cells.

Materials and methods

Cells and transfection

The human cervical cancer cell line SiHa was purchased from the American Type Culture Collection (Manassas, VA, USA) and cultured in Dulbecco's modified Eagle's medium (DMEM; Hyclone; GE Healthcare Life Sciences, Beijing, China) supplemented with 10% fetal bovine serum (Hyclone; GE Healthcare Life Sciences) at 37°C and 5% CO2. Cells were transduced with CMV-Fluc-IRES-RFP lentiviral particles (GeneChem, Co., Ltd., Shanghai China) expressing luciferase and red fluorescent protein (RFP). Cells with stable transfection, which expressed RFP, were isolated by fluorescence-activated cell sorting on a FACSCalibur flow cytometer and analyzed with CellQuest Pro 6.0 software (BD Biosciences, Franklin Lakes, NJ, USA).

SiHa cells were transfected with plasmid pcDNA3.1-control (Thermo Fisher Scientific, Inc., Waltham, MA, USA), which harboured neomycin-resistance, or with pcDNA3.1-SIX1, which was kindly donated by Dr Kong-Ming Wu (Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA), using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). The expression of TGFβ receptor 1 (TβR1) was knocked down using puromycin-resistance small hairpin (sh)RNA lentiviral particles (GeneChem, Co., Ltd.) targeting 5′-CTGTAATTCTGCTGTAATA-3′. As a negative control (NC), shNC, a shRNA not targeting any known gene, was used. Cells with stable transfection were selected with G418 and puromycin (Sigma-Aldrich, St. Louis, MO, USA). The transfected cells were designated as SiHa-control, SiHa-SIX1, SiHa-SIX1-shNC and SiHa-SIX1-shTβR1. To downregulate the expression of SMAD2 and SMAD3, the corresponding small interfering (si)RNAs (Guangzhou RiboBio Co., Ltd., Guangzhou, China) were transfected into tumor cells using Lipofectamine 2000, according to the manufacturer's protocol.

Bioinformatic analysis

The normalized RNA-sequencing data of cervical cancer samples was publicly available from The Cancer Genome Atlas (TCGA) database (https://tcga-data.nci.nih.gov/tcga/tcgaHome2.jsp). Differential gene expression based on SIX1 expression level was determined as previously described (15) using Bioconductor edgeR software package (18), freely available at www.bioconductor.org. Differences were considered to be statistically significant when false discovery rate was <0.25.

Assay of gene expression by reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from cells with TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Total RNA was treated with Ambion Turbo DNA-free DNase (Thermo Fisher Scientific, Inc.; Austin, TX, USA), according to the manufacturer's instructions. RNA (2 µg) was used for first-strand cDNA synthesis with reverse transcriptase (Promega Corporation, Madison, WI, USA), according to the manufacturer's protocol. cDNA was amplified by qPCR in a CFX96 Touch Real-Time PCR Detection system with CFX Manager 3.0 software (Bio-Rad Laboratories, Inc., Hercules, CA, USA) using iQ SYBR Green Supermix (Bio-Rad Laboratories, Inc.). Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and eukaryotic translation elongation factor 1 alpha 1 (EEF1A1) were used as reference genes, since this was reported to be the most reliable gene combination in cervical cancer (19). The primer sequences for RT-qPCR were as follows: GAPDH, sense 5′-GACAGTCAGCCGCATCTTCT-3′ and antisense 5′-TTAAAAGCAGCCCTGGTGAC-3′; EEF1A1, sense 5′-TGCGGTGGGTGTCATCAAA-3′ and antisense 5′-AGAGTGGGGTGGCAGGTATTG-3′; SIX1, sense 5′-CACCAGTTCTCGCCTCACA-3′ and antisense 5′-CACCCGATATTTGCCCAC-3′; E-cadherin, sense 5′-AGGCCAAGCAGCAGTACATT-3′ and antisense 5′-ATTCACATCCAGCACATCCA-3′; and N-cadherin, sense 5′-CCATCAAGCCTGTGGGAATC-3′ and antisense 5′-GCAGATCGGACCGGATACTG-3′ (Beijing Qing Ke New Industrial Biotechnology Co., Ltd., Beijing, China). The reaction included 5 ng cDNA and 10 pM of each primer. The thermal cycling conditions for qPCR were as follows: Denaturation at 95°C for 10 min; 40 cycles of denaturation at 95°C for 10 sec, annealing at 60°C for 30 sec and extension at 72°C for 30 sec. Gene expression was quantified using the comparative Cq method (19). The expression levels of each mRNA were normalized to those of GAPDH and EEF1A1 mRNAs, and expressed as n-fold difference relative to the control.

Immunohistochemistry

Cervical squamous cell carcinoma samples were acquired from the Clinical Database and Biobank of Patients with Gynecologic Neoplasms (ClinicalTrials.gov Identifier: NCT01267851), with informed consent obtained from all subjects participating in the study. The samples were obtained by surgery from cancer patients without preoperative treatment between February 2007 and August 2009 at Tongji Hospital of Tongji Medical College (Wuhan, China). Tumor samples were processed into tissue microarrays (TMAs) at Shanghai Outdo Biotech Co., Ltd. (Shanghai, China). TMAs and tissue sections were subjected to immunohistochemical analysis using the Vectastain ABC kit (ZSGB-BIO, Beijing, China), according to the manufacturer's protocol. Rabbit polyclonal SIX1 antibody (1:200; catalogue no. HPA001893; Sigma-Aldrich) and rabbit monoclonal E-cadherin antibody (1:500; catalogue no. EP700Y; Abcam, Cambridge, UK) were used as primary antibodies. Low or high protein levels of SIX1 in tissue were evaluated as previously described (17). Since E-cadherin is expressed on the cellular membrane of normal cervical epithelium (3), positive membrane expression of E-cadherin was classified as normal, while significant reduction of membrane expression of E-cadherin was classified as a low-E-cadherin level.

Western blot assay

Western blot assay was performed as previously described (20). Briefly, the cells were harvested and lysed using ice-cold RIPA lysis buffer [50 mM Tris-HCl (pH 7.4), 150 mM sodium chloride, 1% Nonidet P-40 and 0.5% sodium deoxycholate; Google Wuhan Biological Technology Co., Ltd., Wuhan, China]. Following centrifugation at 10,000 × g for 15 min at 4°C, the proteins in the supernatants were quantified using Bradford method, separated on 10% SDS-PAGE gel (Google Wuhan Biological Technology Co., Ltd.) and electrotransferred from the gel to a nitrocellulose membrane (Merck & Co., Inc., Whitehouse Station, NJ, USA). After blocking with 5% skimmed milk (Google Wuhan Biological Technology Co., Ltd.) in phosphate-buffered saline, the membranes were immunoblotted with the primary antibodies at 4°C overnight. The primary antibodies used were as follows: Anti-human SIX1 (1:1,100; catalogue no. HPA001893; Sigma-Aldrich), anti-E-cadherin (1:10,000; catalogue no. ab40772; Abcam), anti-N-Cadherin (1:1,000; catalogue no. ab18203; Abcam), anti-SMAD2 (1:1,000; catalogue no. 5339; Cell Signaling Technology, Inc., Danvers, MA, USA), anti-SMAD3 (1:1,000; catalogue no. 9523; Cell Signaling Technology, Inc.) and β-actin (1:500; catalogue no. ab8229; Abcam). Signals were detected using a SuperSignal West Pico Chemiluminescent Substrate kit (Pierce Biotechnology, Inc., Rockford, IL, USA) with a ChemiDoc XRS+ machine (Bio-Rad Laboratories, Inc.). Protein levels of β-actin were employed as loading controls. The relative protein expression levels were analyzed using Image Lab software (version 4.1; Bio-Rad Laboratories, Inc.).

Migration assay

Migration assay was performed using a Transwell chamber (Corning Incorporated, Corning, NY, USA). Tumor cells (1×104 cells) were placed in the upper chamber. Following 8-h incubation at 37°C in a humidified incubator with 5% CO2, the cells that passed through the membrane were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet (both from Google Wuhan Biological Technology Co., Ltd.) prior to be counted under a microscope (BX53; Olympus Corporation Tokyo, Japan). Cells from three randomly selected fields in each membrane were counted, and the average number of cells per field was calculated.

Animal studies

Female athymic nude (nu/nu) 4-week-old mice (n=40) were purchased from Beijing HFK Bio-technology Co, Ltd. (Beijing, China). The studies were approved by the Committee on the Ethics of Animal Experiments of Tongji Medical College (Wuhan, China). Mice were maintained in the accredited animal facility of Tongji Medical College at 20–26°C in a 12-h light/dark cycle. A lymphatic metastasis model of cervical cancer was used as previously described (16). Briefly, 5×106 tumor cells in 50 µl serum-free DMEM (Boster Biological Technology, Co., Ltd., Wuhan, China)/Matrigel (BD Biosciences) at a 9:1 ratio were injected subcutaneously into the claw pads of the mice. Tumor size (mm3) was measured and calculated by the following formula: Volume = (width)2 × length / 2. When primary tumors reached ~150 mm3 in size, metastases were tracked by optical imaging of luciferase activity originating from tumor cells using the IVIS Spectrum system (Caliper Life Sciences; PerkinElmer, Inc., Waltham, MA, USA). Subsequently, when primary tumors reached ~250 mm3 in size, the mice were euthanized, and their popliteal and inguinal lymph nodes were excised. Metastases of tumor cells in the lymph nodes were confirmed by detection of tumor-expressed RFP under a SZX16 dissecting microscope (Olympus Corporation). The incidence of metastasis-positive mice in each group was calculated.

Statistical analysis

Each experiment was independently repeated ≥3 times. SPSS version 13.0 software (SPSS, Inc., Chicago, IL, USA) was used for statistical analysis. The results were expressed as the mean ± standard deviation, and interpreted by one-way analysis of variance or χ2 test. P<0.05 was considered to indicate a statistically significant difference.

Results

SIX1 expression is associated with EMT of cervical cancer

To determine the effect of SIX1 on EMT of cervical cancer, the effect of SIX1 on the expression of E-cadherin, which is a common feature of EMT, was analyzed. The RNA-sequencing data of cervical cancer specimens was obtained from the TCGA database. The expression profiles of SIX1 and E-cadherin are shown in Fig. 1A. The specimens were divided into SIX1-low expression group and SIX1-high expression group. Using Bioconductor edgeR software package, E-cadherin was identified to be one of the significantly downregulated genes in the SIX1-high expression group (false discovery rate=0.046; Fig. 1B). These results were further confirmed by quantifying the mRNA levels of SIX1 and E-cadherin in the samples (P=0.003; Fig. 1C). A significant decrease in the membrane expression levels of E-cadherin was observed at the primary site of cervical cancer samples that highly expressed SIX1 (P=0.001; Fig. 1D). These results suggest that increased SIX1 expression is associated with mesenchymal phenotype of cervical cancer.

SIX1 enhances TGFβ-induced EMT in cervical cancer cells

TGFβ is one of the most important inducers of EMT (5,21). However, treatment with TGFβ1 only suppressed slightly the expression of the epithelial marker E-cadherin, and negligibly induced the expression of the mesenchymal marker N-cadherin in SiHa cells, a cervical cancer cell line with low expression levels of SIX1 (Fig. 2A) (17). These results suggested that cervical cancer cells may not be sensitive to TGFβ1. Overexpression of SIX1 in SiHa cells affected slightly the expression of E-cadherin and N-cadherin (Fig. 2B). Notably, TGFβ1 could effectively induce SIX1-expressing tumor cells to undergo EMT (Fig. 2B). Similar results were obtained when E-cadherin (SiHa-control 0 vs. 5 days, P=0.016; SiHa-SIX1 0 vs. 5 days, P<0.001) and N-cadherin (SiHa-control 0 vs. 5 days, P=0.071; SiHa-SIX1 0 vs. 5 days, P<0.001) expression was quantified using RT-qPCR (Fig. 2C). Correspondingly, TGFβ1-treated SiHa-SIX1 cells became more fusiform with decreased connection between the cells than TGFβ1-treated SiHa-control cells (Fig. 2D). These results demonstrate that SIX1 coordinates with TGFβ to induce EMT in cervical cancer cells.

TGFβ signals are required for SIX1-induced EMT

The present authors previously reported that SIX1 could interact with SMAD proteins, which are the signal transducers of TGFβ signaling (17). To further clarify the role of TGFβ signals in inducing EMT on SIX1-expressing tumor cells, the components of the TGFβ signaling pathway were inhibited in the present study. When the expression of SMAD2 or SMAD3 was knocked down, the TGFβ1-induced EMT process in SiHa-SIX1 cells was abrogated (Fig. 3A). The same results were obtained when E-cadherin (si-SMAD2, P<0.001; si-SMAD3, P<0.001) and N-cadherin (si-SMAD2, P<0.001; si-SMAD3, P<0.001) expression was quantified using RT-qPCR (Fig. 3B). Albeit not significantly induced EMT in vitro, overexpression of SIX1 alone resulted in a decrease in E-cadherin expression in vivo (P=0.002; Fig. 3C). However, silencing TβR1 completely abrogated the effect of SIX1 (P<0.001; Fig. 3C), further indicating that TGFβ signals are required for SIX1 to induce EMT.

SIX1 coordinates with TGFβ to enhance the metastatic capacity of cervical cancer cells

EMT is associated with the metastatic capacity of tumor cells (5,6). Based on the above results, the effect of SIX1 and TGFβ1 on the metastatic phenotype of cervical cancer cells was further investigated. The migration of SiHa cells was increased following TGFβ1 stimulation. TGFβ1 was more efficient than the control in promoting the migratory capacity of tumor cells if the cells expressed high levels of SIX1 (SiHa-control, P=0.003; SiHa-SIX1, P<0.001; Fig. 4A). The metastatic capacity of tumor cells was further tested using a previously described lymphatic metastasis model (Fig. 4B). The results indicated that SIX1-mediated promotion of lymph node metastasis was completely abolished by silencing TβR1 (SiHa-SIX1 shNC vs. SiHa-SIX1 shTβR1, P=2.61E-4; Fig. 4C). These results demonstrate that SIX1 and TGFβ signals coordinate to enhance the metastatic capacity of cervical cancer cells.

Discussion

An increasing number of studies have provided strong evidence for the critical role of the EMT program in tumor progression and metastasis (5,6). Although TGFβ is regarded as one of the most important inducers of EMT, several non-invasive tumor cells were not able to undergo TGFβ-induced EMT in vitro (9). The present data suggest that the expression levels of the transcription factor SIX1 could determine the sensitivity of tumor cells to TGFβ stimulation.

In the present study, the expression of SIX1 was negatively correlated with that of the epithelial marker E-cadherin in cervical cancer. Consistently, increased SIX1 expression in tumor cells could sufficiently reduce E-cadherin expression and promote lymph node metastasis in vivo. However, increased SIX1 expression in SiHa cells could not significantly induce EMT in vitro, suggesting that SIX1 could not efficiently induce EMT by itself. The present data demonstrated that the main contribution of SIX1 to EMT was to increase the sensitivity of tumor cells to TGFβ stimulation. Increased expression of SIX1 could promote EMT of cervical cancer cells in a TGFβ-dependent manner. Based on the overexpression of SIX1, TGFβ induced more remarkable changes in the transition of phenotype than the SiHa-control group. Therefore, SIX1 and TGFβ coordinated to promote cell motility and tumor metastasis of cervical cancer.

Previous studies have described the roles of TGFβ-activated SMADs in EMT. Increased expression of SMAD2 or SMAD3 was reported to induce EMT, whereas expression of dominant negative versions of SMAD2 or SMAD3 blocked TGFβ-induced EMT (7,22). Additionally, TGFβ activates SMAD and non-SMAD signals, including Rho-like guanosine triphosphatases, phosphatidylinositol-4,5-bisphosphate 3-kinase and mitogen-activated protein kinase signaling pathways, which also contribute to EMT (6,23,24). In the present study, TGFβ could efficiently induce EMT in vitro and lymph node metastasis in vivo if tumor cells expressed high levels of SIX1. However, knocking down the expression of SMAD2 or SMAD3 could completely block the EMT process induced by SIX1 and TGFβ. In a previous study, the present authors demonstrated that SIX1 was involved in the SMAD2/3 protein complex and could enhance TGFβ-SMAD signaling (17). Therefore, enhancing TGFβ-SMAD signaling is an important mechanism by which SIX1 promotes EMT and lymphatic metastasis in cervical carcinoma.

The signal transduction of TGFβ forms a complex network, involving the activation of SMAD and non-SMAD signals, and the crosstalk among various signal transduction pathways such as wingless-related integration site and Notch signaling pathways provides context-dependent effects during EMT (25,26). Therefore, context-specific outcomes may be generated according to the availability of repressors/activators, distinct intensity or duration of SMAD/non-SMAD signaling activity or changes in the levels of interacting protein partners (7). The present data revealed that TGFβ1 stimulation was not able to induce EMT in SiHa cells, which are cervical cancer cells with low expression of SIX1 (17). By contrast, when SiHa-SIX1 cells were stimulated with TGFβ1, significant changes were observed in the expression of EMT markers, cell morphology and metastatic capacity of the cells. In addition, higher expression levels of E-cadherin in SIX1-low clinical samples and in vivo experiments also suggested that environmental factors such as TGFβ were ineffective in inducing EMT in a SIX1-low context. Therefore, the coordination of SIX1 and TGFβ may be critical for inducing EMT in cervical carcinoma.

In summary, the present study revealed the important role of the coordination of SIX1 and TGFβ in inducing EMT in cervical cancer. SIX1 induced EMT of cervical cancer cells in a TGFβ-dependent manner, and increased SIX1 expression significantly enhanced the TGFβ-induced transition of mesenchymal phenotype, indicating that SIX1 and TGFβ coordinate to promote the EMT program and the metastatic capacity of cervical cancer cells. In conclusion, the present results demonstrate that SIX1 plays a crucial role in the progression and metastasis of cervical cancer, and suggest that targeting SIX1 and/or TGFβ signaling may be a valuable strategy in cancer therapy.

Acknowledgements

The present authors would like to thank Dr Qi-Lin Ao and Dr Shuang Guo (Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China) for reviewing the histology data. The present study was supported by the National Science Foundation of China (Beijing, China; grant nos. 81072135 and 81372801).

References

1 

Gao D, Vahdat LT, Wong S, Chang JC and Mittal V: Microenvironmental regulation of epithelial-mesenchymal transitions in cancer. Cancer Res. 72:4883–4889. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Tsai JH and Yang J: Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 27:2192–2206. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Li Y, Wang W, Wang W, Yang R, Wang T, Su T, Weng D, Tao T, Li W, Ma D and Wang S: Correlation of TWIST2 up-regulation and epithelial-mesenchymal transition during tumorigenesis and progression of cervical carcinoma. Gynecol Oncol. 124:112–118. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Zhou XM, Zhang H and Han X: Role of epithelial to mesenchymal transition proteins in gynecological cancers: Pathological and therapeutic perspectives. Tumour Biol. 35:9523–9530. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Xu J, Lamouille S and Derynck R: TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 19:156–172. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Katsuno Y, Lamouille S and Derynck R: TGF-β signaling and epithelial-mesenchymal transition in cancer progression. Curr Opin Oncol. 25:76–84. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Brown KA, Aakre ME, Gorska AE, Price JO, Eltom SE, Pietenpol JA and Moses HL: Induction by transforming growth factor-beta1 of epithelial to mesenchymal transition is a rare event in vitro. Breast Cancer Res. 6:R215–R231. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Feng XX, Liu M, Yan W, Zhou ZZ, Xia YJ, Tu W, Li PY and Tian DA: β3 integrin promotes TGF-β1/H2O2/HOCl-mediated induction of metastatic phenotype of hepatocellular carcinoma cells by enhancing TGF-β1 signaling. PLoS One. 8:e798572013. View Article : Google Scholar : PubMed/NCBI

11 

Ford HL, Landesman-Bollag E, Dacwag CS, Stukenberg PT, Pardee AB and Seldin DC: Cell cycle-regulated phosphorylation of the human SIX1 homeodomain protein. J Biol Chem. 275:22245–22254. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Micalizzi DS, Christensen KL, Jedlicka P, Coletta RD, Barón AE, Harrell JC, Horwitz KB, Billheimer D, Heichman KA, Welm AL, et al: The Six1 homeoprotein induces human mammary carcinoma cells to undergo epithelial-mesenchymal transition and metastasis in mice through increasing TGF-beta signaling. J Clin Invest. 119:2678–2690. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Ng KT, Man K, Sun CK, Lee TK, Poon RT, Lo CM and Fan ST: Clinicopathological significance of homeoprotein Six1 in hepatocellular carcinoma. Br J Cancer. 95:1050–1055. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Behbakht K, Qamar L, Aldridge CS, Coletta RD, Davidson SA, Thorburn A and Ford HL: Six1 overexpression in ovarian carcinoma causes resistance to TRAIL-mediated apoptosis and is associated with poor survival. Cancer Res. 67:3036–3042. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Liu D, Zhang XX, Xi BX, Wan DY, Li L, Zhou J, Wang W, Ma D, Wang H and Gao QL: Sine oculis homeobox homolog 1 promotes DNA replication and cell proliferation in cervical cancer. Int J Oncol. 45:1232–1240. 2014.PubMed/NCBI

16 

Liu D, Zhang XX, Wan DY, Xi BX, Ma D, Wang H and Gao QL: Sine oculis homeobox homolo 1 promotes α5β1-mediated invasive migration and metastasis of cervical cancer cells. Biochem Biophys Res Commun. 446:549–554. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Liu D, Li L, Zhang XX, Wan DY, Xi BX, Hu Z, Ding WC, Zhu D, Wang XL, Wang W, et al: SIX1 promotes tumor lymphangiogenesis by coordinating TGFβ signals that increase expression of VEGF-C. Cancer Res. 74:5597–5607. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Robinson MD, McCarthy DJ and Smyth GK: edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Shen Y, Li Y, Ye F, Wang F, Lu W and Xie X: Identification of suitable reference genes for measurement of gene expression in human cervical tissues. Anal Biochem. 405:224–229. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Wang C, Zhou L, Li S, Wei J, Wang W, Zhou T, Liao S, Weng D, Deng D, Weng Y, et al: C4orf7 contributes to ovarian cancer metastasis by promoting cancer cell migration and invasion. Oncol Rep. 24:933–939. 2010.PubMed/NCBI

21 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Valcourt U, Kowanetz M, Niimi H, Heldin CH and Moustakas A: TGF-beta and the Smad signaling pathway support transcriptomic reprogramming during epithelial-mesenchymal cell transition. Mol Biol Cell. 16:1987–2002. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Derynck R and Zhang YE: Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 425:577–584. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Lamouille S and Derynck R: Emergence of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin axis in transforming growth factor-β-induced epithelial-mesenchymal transition. Cells Tissues Organs. 193:8–22. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Thiery JP and Sleeman JP: Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 7:131–142. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC and de la Pompa JL: Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 18:99–115. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2016
Volume 12 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun SH, Liu D, Deng YT, Zhang XX, Wan DY, Xi BX, Huang W, Chen Q, Li MC, Wang MW, Wang MW, et al: SIX1 coordinates with TGFβ signals to induce epithelial-mesenchymal transition in cervical cancer. Oncol Lett 12: 1271-1278, 2016
APA
Sun, S., Liu, D., Deng, Y., Zhang, X., Wan, D., Xi, B. ... Gao, Q. (2016). SIX1 coordinates with TGFβ signals to induce epithelial-mesenchymal transition in cervical cancer. Oncology Letters, 12, 1271-1278. https://doi.org/10.3892/ol.2016.4797
MLA
Sun, S., Liu, D., Deng, Y., Zhang, X., Wan, D., Xi, B., Huang, W., Chen, Q., Li, M., Wang, M., Yang, F., Shu, P., Wu, K., Gao, Q."SIX1 coordinates with TGFβ signals to induce epithelial-mesenchymal transition in cervical cancer". Oncology Letters 12.2 (2016): 1271-1278.
Chicago
Sun, S., Liu, D., Deng, Y., Zhang, X., Wan, D., Xi, B., Huang, W., Chen, Q., Li, M., Wang, M., Yang, F., Shu, P., Wu, K., Gao, Q."SIX1 coordinates with TGFβ signals to induce epithelial-mesenchymal transition in cervical cancer". Oncology Letters 12, no. 2 (2016): 1271-1278. https://doi.org/10.3892/ol.2016.4797