SPOP promotes tumor progression via activation of β-catenin/TCF4 complex in clear cell renal cell carcinoma

  • Authors:
    • Wencai Zhao
    • Jiancheng Zhou
    • Zhuo Deng
    • Yang Gao
    • Yongyi Cheng
  • View Affiliations

  • Published online on: July 6, 2016     https://doi.org/10.3892/ijo.2016.3609
  • Pages: 1001-1008
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Renal cell carcinoma (RCC) is the most common type of kidney cancer, about one third of the cases are diagnosed at advanced stages with metastases and effective treatments for metastatic RCC are lacking. The molecular events supporting RCC progression remain poorly understood. SPOP, an E3 ubiquitin ligase component, was recently showed to sufficiently promote RCC tumorigenesis, however, other potential functions of SPOP in RCC have not been studied. In the present investigation, by assessing the immunohistochemical staining of SPOP in urological tumors, we found the protein was highly expressed in RCC, in particular, it was specifically expressed in clear cell RCC. cDNA microarray data showed that SPOP mRNA level was significantly increased in clear cell RCC compared to normal kidney tissues, which might be the result of the abnormal DNA copy number of this gene. More interestingly, SPOP was positive in tumors with local invasion or metastasis, and it was associated with tumor recurrence-free survival of clear cell RCC patients. Further in vitro assays demonstrated that SPOP drove RCC epithelial-mesenchymal transition (EMT) and promoted cell invasion. Mechanistically, SPOP enhanced β-catenin protein expression as well as its nuclear translocation, and elevated TCF4 expression. Both β-catenin and TCF4 upregulated the critical EMT-inducing transcription factor ZEB1, which functioned as an effector of β-catenin/TCF4 signaling in RCC invasion. These data identified SPOP as a new marker and prognostic factor for clear cell RCC, and its functions provide new insight into the molecular mechanisms of RCC progression, in which SPOP appears to be an EMT activator.

Introduction

Renal cell carcinoma (RCC) is the most common type of kidney cancer, >200,000 new cases and 100,000 deaths are estimated to occur worldwide each year (1). Up to 30% of RCC patients present with metastases at the time of diagnosis and nearly half of the rest will subsequently develop metastases in their course. When metastasis occurs, it is largely incurable, with a very poor 5-year survival rate (2). RCC is highly resistant to chemotherapy and radiotherapy, immunotherapies such as interleukin-2 and interferon α are once used as first-line treatments for metastatic RCC (mRCC), however, the response rates are extremely low (3). Newly developed targeted-therapies based on the understanding of molecular mechanisms of RCC progression make significant improvements over immunotherapies for mRCC. Unfortunately, <40% of patients have response to targeted-therapies and nearly all patients will eventually develop resistance (4,5). It appears that a therapeutic ceiling has been reached for mRCC, thus, it is important to comprehensively study the mechanisms of how RCC develops metastasis, and explore promising therapeutic approaches for this disease.

SPOP, a BTB/POZ domain containing speckle-type POZ protein, was first identified as a component for the E3 ubiquitin ligase (6). In Drosophila, D-SPOP (ortholog of human SPOP) has been shown to promote the ubiquitination and degradation of Cubitus interruptus (Ci) in the Hedgehog pathway, and JNK phosphatase puckered (Puc) in the tumor necrosis factor (TNF) pathway, respectively (79). In human, SPOP has been recently shown to mediate ubiquitination of the death domain-associated protein (Daxx) (10), the polycomb group protein BMI-1, the histone variant MacroH2A (11), and the transcription factor Gli (9).

In RCC, hypoxia-inducible factor (HIF) and mammalian target of rapamycin (mTOR) pathways are considered as the most predominant pathways controlling RCC development and progression (12,13), and therapies targeting these two pathways have brought clinical benefits to mRCC (14). A more recent study shows that SPOP is a direct target of HIF, and cytoplasmic SPOP promotes RCC tumorigenesis through the ubiquitination and degradation of multiple regulators of cellular proliferation and apoptosis, including the tumor suppressor PTEN, ERK phosphatases DUSP7, the proapoptotic molecule Daxx, and the Hedgehog pathway transcription factor Gli2 (15). However, other potential functions of SPOP in RCC have not been studied. In this study, we aim to determine whether SPOP promotes invasion and metastasis in RCC.

Materials and methods

Human RCC specimens and immunohistochemistry staining

Forty-seven human RCC and 11 matched normal kidney specimens were obtained from patients who underwent surgical resection with the approval of the Institutional Review Board (IRB). Immunohistochemistry was performed as described previously (16). Briefly, paraffin-embedded sections were subjected to deparaffinization, rehydration, and heat-induced antigen retrieval. After blocking of endogenous peroxidase with 3% hydrogen peroxide, sections were subsequently incubated with primary SPOP antibody (Santa Cruz Biotechnology), horseradish-peroxidase-labeled dextran polymer (Dako EnVision™) and developed with 3,3′-diaminobenzidine chromogen followed by counter staining with hematoxylin. All stains were assessed by an independent pathologist according to the histologic scoring system (H-score) based on the product of staining intensity (0, no staining; 1, weak; 2, moderate; and 3, strong) and percentage of stained cells (0, 0%; 1, 1–30%; 2, 31+70%; and 3, 71–100%). The expression of SPOP in each tissue was considered either negative (H-score, <2) or positive (H-score, >2).

Cell culture

Human normal kidney cell HK-2, RCC cells 786-0, A498, RCC4 and 769-P were maintained in RPMI-1640 medium (Gibco) supplied with 10% fetal bovine serum (FBS), RCC cells ACHN, CAKI-1, CAKI2 and A498 were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco) supplied with 10% FBS. All the cells were cultured in a humidified incubator containing 5% CO2 at 37°C.

cDNA constructs, siRNA and transfection

Human SPOP cDNA cloned into pDONR221 vector was obtained from the DNASU Plasmid Repository. Control siRNA (si-NC) and siRNA specifically targeting SPOP (si-SPOP) were from Guangzhou RiboBio Co., Ltd. For transfections, 2×105 cells were seeded in 6-well plate and cultured overnight, SPOP plasmids or siRNAs were transfected into cells by Xfect™ transfection reagent (Clontech) according to the manufacturer’s instructions. Twenty-four hours (h) after the transfection, cell protein or RNA was collected for further assays.

Transwell invasion assay

Matrigel-coated Transwell chambers were applied to examine RCC cell in vitro invasive ability. RCC cells were pre-transfected with the indicated plasmids or siRNAs, 100 μl 0.5% FBS medium of 3×104 cell suspension was then planted into the upper chamber, and 600 μl of 10% FBS medium was supplied to the lower chamber. Cells were cultured at 37°C in 5% CO2 for 24 h. Invaded cells onto the lower surface of the upper chambers were stained with 0.5% crystal violent (Sigma) and photographed and counted.

Immunofluorescence

Microslide cultured cells were fixed with 4% paraformaldehyde, permeabilized with 0.3% Triton X-100 and blocked with 5% bovine serum albumin. Cells were incubated with β-catenin primary antibody (Cell Signaling Technology) overnight at 4°C and subsequently incubated with AlexaFluor 488-conjugated secondary antibody (Sigma) for 1 h at room temperature, followed by nuclear staining with 4,6-diamidino-2-phenylindole and fluorescence was visualized by fluorescence microscopy (Olympus Optical Co.)

Reverse transcriptional (RT) real-time PCR

Cell total RNA was extracted with RNeasy mini kit (Qiagen) and reverse transcribed with cDNA synthesis kit (Invitrogen). Real-time PCR analysis was set up with SYBR Green qPCR Supermix kit (Invitrogen) supplied with commercial primers specific for the indicated genes, and carried out in the iCycler thermal cycler (Bio-Rad). The relative level of mRNA expression of each gene was determined by normalizing with an internal control gene GAPDH.

Western blotting

Western blotting was performed as previously described (17). Cells were first lysed and total proteins were collected, equivalent amounts of protein were separated on 10% NuPAGE Bis-Tris gels (Invitrogen) and transferred to nitrocellulose membranes. Membranes were blocked with 3% skim-milk (w/v), and incubated with primary antibodies overnight at 4°C. After washing, membranes were incubated with appropriate secondary antibodies conjugated with horseradish peroxidase and signals were then detected by chemiluminescence (Pierce). Primary SPOP, vimentin, pan-cytokeratin (Pan-CK), TCF4 and GAPDH antibodies were purchased from Santa Cruz Biotechnology; E-cadherin and α-SMA antibodies were from BD Biosciences, ZEB1 and MMP-2 antibodies were from Cell Signaling Technology.

Bioinformatic and statistical analyses

The RNA-sequencing-based mRNA expression data for SPOP, TCF4, ZEB1 genes and the reverse phase protein array-based protein expression data for β-catenin of human clear cell RCC samples were all retrieved from The Cancer Genome Atlas (TCGA) Data Portal (18). Gene microarray data for SPOP of human normal kidney and clear cell RCC tissues were retrieved from the GEO datasets (GSE14994, GSE781 and GSE15641). SPOP gene microarray data and DNA copy number data of multiple types of cancer cell lines were retrieved from the Cancer Cell Line Encytopedia (CCLE) datasets. The Kaplan-Meier analysis (long-rank test) was performed to analyze recurrence-free survival. Pearson’s correlation coefficient was used to test the association between genes. Data from in vitro assay are presented as the mean ± SEM from three independent experiments, and the differences between two groups were compared by Student’s two-tail t-test. All statistical analyses were performed by GraphPad Prism6 (GraphPad Software).

Results

SPOP is highly expressed in clear cell RCC

Previous studies suggest that overexpression of SPOP may lead to dysregulation of pathways involved in tumorigenesis (8,9). We assessed SPOP expression in urological tumors including prostate cancer, bladder cancer, RCC and normal kidney tissues by immunohistochemistry. Interestingly, we found SPOP was negative in prostate cancer, bladder cancer and normal kidney tissues, but it was highly expressed in RCC tissues (Fig. 1A). RCC is a heterogeneous group of tumors with distinct histological subtypes, including clear cell, papillary, chromophobe, and other rare subtypes in addition to oncocytoma (19). When RCC subtypes were stratified, we found that the papillary, chromphobe or oncocytoma RCC were weak or negative for SPOP, but clear cell RCC was positively stained with this protein (Fig. 1B). In total we analyzed 47 clear cell RCC and matched 11 normal kidney tissues, and the results showed that 83% clear cell RCC were positive for SPOP, while only 18% normal tissues were positive (Table I). This indicates that SPOP is highly expressed in clear cell RCC and may serve as a specific biomarker for this type of RCC.

Table I

SPOP IHC staining in normal and clear cell RCC tissues.

Table I

SPOP IHC staining in normal and clear cell RCC tissues.

SPOP

Positive (%)Negative (%)P-value
Normal tissues2 (18)9 (82)<0.001
Clear cell RCC39 (83)8 (17)

In addition to the determination of SPOP protein status in clear cell RCC, we checked SPOP mRNA expression by analysis of gene microarray data of normal kidney and clear cell RCC from the GEO datasets. Results from three independent datasets consistently showed that SPOP mRNA was significantly upregulated in clear cell RCC compared to normal kidney (Fig. 2A). To further explore whether SPOP upregulation is due to genomic abnormality, we analyzed the association of SPOP mRNA level and its DNA copy number in multiple types of cancer cell lines including 1,014 samples from the CCLE datasets, and found there was a positive correlation between SPOP mRNA level and its DNA copy number (Fig. 2B), and in RCC cell lines, a similarly positive correlation was also found (Fig. 2C). These data suggest that the upregulation of SPOP in RCC may be due to the genomic variation.

SPOP is associated with progressive clear cell RCC

Previous studies indicate that SPOP plays important roles during tumor cell apoptosis and proliferation (7,10,15), we further investigated the potential functions of SPOP in tumor progression. The expression of SPOP in human clear cell RCC with local invasion (tumor cell invaded into perirenal fat, renal capsule or regional lymph node) was detected by immunohistochemistry, notably, the results showed that almost all the RCC with local invasion were SPOP-positive (Fig. 3A). We compared SPOP expression in RCC with different pathological stages according to the 2010 AJCC TNM classfication (20), and found RCC in T3–4 stages (primary tumors with local invasion) showed much high frequency of SPOP positive staining compared to RCC in T1–2 stages (without local invasion). Similarly, RCC with lymph node invasion (N1) or distant metastasis (M1) showed very high frequency of SPOP positive staining compared to RCC in N0 (without lymph node invasion) or M0 (without distant metastasis) (Table II). These date suggest SPOP is associated with clear cell RCC invasion and metastasis. We further analyzed the association of SPOP expression and tumor recurrence-free survival, and the result demonstrated that SPOP was negatively correlated with RCC recurrence-free survival (Fig. 3B), indicating SPOP as novel prognostic marker for RCC patients.

Table II

SPOP IHC staining in clear cell RCC with local invasion/metastasis.

Table II

SPOP IHC staining in clear cell RCC with local invasion/metastasis.

SPOP

Positive (%)Negative (%)
Tumor stage
 T1–217 (70)7 (30)
 T3–422 (96)1 (4)
Lymph nodes
 N028 (80)7 (20)
 N111 (92)1 (8)
Metastasis
 M031 (79)8 (21)
 M18 (100)0 (0)
SPOP promotes the invasiveness of RCC

To confirm the biological function of SPOP in RCC invasion, in vitro cell line based assays were performed. Profile of SPOP expression in a series of RCC cell lines by RT-PCR showed ACHN cells with low SPOP expression, thus, it was applied for SPOP overexpression model, while A498 cells with high SPOP was applied for SPOP silencing model (Fig. 4A and B). In vitro Transwell invasion assays demonstrated that overexpression of SPOP promoted ACHN invasion (Fig. 4C), while silencing of SPOP by siRNA in A498 cells suppressed cell invasion (Fig. 4D). It has been well documented that epithelial-mesenchymal transition (EMT) is a process thought to initiate metastasis, and it is characterized by the gain of mesenchymal markers (e.g., vimentin, α-SMA, MMP2) and the loss of epithelial markers (e.g., E-cadherin, cytokeratins), as well as increased motility and invasion of cancer cells (21,22). We examined the expression of EMT markers in SPOP overexpressing or silencing cells, and the results showed that SPOP downregulated epithelial markers, such as E-cadherin and Pan-CK, and upregulated mesenchymal makers, such as vimentin, α-SMA and MMP-2 (Fig. 4E). These data indicate SPOP as an inducer for the invasiveness of RCC cells.

Mechanisms of SPOP in regulation of RCC invasion

We further dissected the molecular mechanisms of SPOP in regulation of RCC invasion. Firstly, the associations between SPOP and a panel of EMT markers and EMT-inducing transcription factors (EMT-TFs) were analyzed in human clear cell RCC samples from TCGA datasets by Pearson correlation analyses. The results showed negative correlations between SPOP and epithelial marker CDH1 (E-cadherin), and positive correlations between SPOP and mesenchymal makers VIM (vimentin), ACTA2 (α-SMA) and MMP-2 (Table III). Importantly, SPOP was positively correlated with many critical EMT-TFs, such as TCF4 and ZEB1 (Table III and Fig. 5A). Further cell line based assays confirmed that overexpression of SPOP upregulated TCF4 and ZEB1 expression (Fig. 5B). ZEB1 is well known as one of the most critical transcriptional factors driving EMT in many cancer cells (23), and β-catenin/TCF4 complex has been demonstrated to bind ZEB1 gene promoter region and promote its transcription (24). Our data showed that silencing of TCF4 in RCC cells suppressed ZEB1 expression (Fig. 5C), and there was an extremely positive correlation between TCF4 and ZEB1 mRNA in clear cell RCC samples (Fig. 5D), these data again confirmed that TCF4 is an upstream regulator for ZEB1 expression. Additionally, we observed that SPOP could also upregulate cytosolic β-catenin protein expression and promote its nuclear translocation (Fig. 5E). Silencing of β-catenin suppressed ZEB1 expression (Fig. 5F), and there was a positive correlation between β-catenin protein and ZEB1 expression in clear cell RCC samples (Fig. 5G), suggesting β-catenin as upstream regulator for ZEB1. Furthermore, although overexpression of SPOP upregulated ZEB1 expression as well as cell invasion, co-transfection of TCF4 siRNA or β-catenin siRNA could ablate the effects of SPOP on ZEB1 gene expression and cell invasive ability (Fig. 5H and I). Taken together, our results indicate that SPOP promotes ZEB1 to drive RCC cell invasion via activating the β-catenin/TCF4 complex (Fig. 6).

Table III

Pearson correlation analyses of the mRNA expression of SPOP and EMT related genes in human RCC samples from TCGA dataset (RNA Seq V2 RSEM).

Table III

Pearson correlation analyses of the mRNA expression of SPOP and EMT related genes in human RCC samples from TCGA dataset (RNA Seq V2 RSEM).

Pearson r95% CIP-valueSignificant (α=0.05)No. of samples
SPOP vs. CDH1−0.1875−0.27 to −0.10<0.0001Yes534
SPOP vs. VIM0.13930.06 to 0.220.0013Yes534
SPOP vs. ACTA20.1850.10 to 0.27<0.0001Yes534
SPOP vs. MMP20.27340.19 to 0.35<0.0001Yes534
SPOP vs. MMP9−0.0442−0.13 to 0.040.3079No534
SPOP vs. TCF40.42770.36 to 0.49<0.0001Yes534
SPOP vs. ZEB10.39420.32 to 0.46<0.0001Yes534
SPOP vs. ZEB20.28840.21 to 0.36<0.0001Yes534
SPOP vs. SMAD40.20860.13 to 0.29<0.0001Yes534
SPOP vs. SNAI10.25810.18 to 0.34<0.0001Yes534
SPOP vs. SNAI20.30630.23 to 0.38<0.0001Yes534
SPOP vs. TWIST10.10920.025 to 0.190.0115Yes534

Discussion

RCC is a clinicopathologically heterogeneous disease with distinct histological subtypes, including clear cell which accounts for the 70% of cases, and other rare subtypes, such as papillary, chromophobe, and oncocytoma (25). Although different subtypes of RCC exhibit certain distinguishing morphology, diagnostic difficulties arise when one subtype displays morphologic features that overlap with others. Recent advances are paving the way for seeking specific molecular abnormalities based on improved knowledge of the cytogenetics and molecules to recognize distinct molecular subtypes. A panel of immunohistochemical markers are used to differentiate the major subtypes of RCC. Unfortunately, these markers lack specificity and sensitivity. For example, carbonic anhydrase IX has been proposed as a sensitive marker for clear cell RCC, but it is not positive for all cases (26). Vimentin, a broad mesenchymal marker, is expressed in 87–100% clear cell and papillary RCC, but also in 73% oncocytoma (27). PAX2 is found to be a good marker for kidney cancers, but it is also positive for normal kidney tissues (28). We find that SPOP is negative in prostate cancer and bladder cancer but positive in 83% of the clear cell RCC, and all the cases with local invasion included for this study are positive. Although a large cohort of samples is required for further study, results from small number of samples in this study indicate SPOP may serve as a new marker for clear cell RCC, especial for metastatic cases.

Epithelial-mesenchymal transition (EMT) is a process thought to initiate metastasis, and it is characterized by the gain of mesenchymal markers (e.g., vimentin, α-SMA, MMP2) and the loss of epithelial markers (e.g., E-cadherin, cytokeratins), as well as increased motility and invasion of cancer cells. EMT is driven by many EMT-inducing transcription factors (EMT-TFs) (21,23). The well documented EMT-TFs include the Zinc-finger factors Snail, Slug, ZEB2 and ZEB1, and the HLH factors Twist and E12/E47. All of which directly bind to E-boxes in the promoter of the E-cadherin gene and repress its expression (29). By analyses of the associations of SPOP and EMT markers and a panel of EMT-TFs in a large cohort of clear cell RCC samples, we find SPOP is negatively correlated with epithelial maker and positively correlated with mesenchymal markers and all the EMT-TFs, suggesting SPOP indeed plays essential roles in inducing EMT and promoting RCC progression, which is consistent with the finding that SPOP predicts a poor recurrence-free survival of RCC patient.

Within the SPOP associated EMT-TFs, we further confirm ZEB1 is the critical downstream effector of SPOP to drive RCC cell invasion. ZEB1 has been reported to be regulated by the TGF-β signaling pathway (30) and Wnt/β-catenin signaling pathway (24). The activation of Wnt signaling inactivates the glycogen synthase kinase-3β, and leads to the stabilization of β-catenin protein in cytoplasm followed by the nuclear translocation to complex with TCF4 and enhance the transcriptional activity of TCF4 (31). β-catenin/TCF4 complex has been demonstrated to bind the ZEB1 promoter region and promote its transcription in intestinal tumor cells (24). We confirm that SPOP upregulates ZEB1 in clear cell RCC by promoting β-catenin protein nuclear translocation and TCF4 mRNA expression. SPOP, as an E3 ubiquitin ligase component, and appears to promote RCC tumorigenesis by the ubiquitination and degradation of PTEN, DUSP7 and Daxx as previously reported (15). However, results in this study indicate new actions of SPOP in RCC, of which SPOP seems to regulate β-catenin in posttranscriptional level and TCF4 in transcriptional level, further studies are required to illuminate the details of the mechanism.

Acknowledgements

This study was supported by the Shaanxi Provincial Key Scientific Foundation (grant no. 2013KTCL03-04 to Y. Xu), the National Natural Science Foundation of China (grant no. NSFC81172436 to Y. Sun) and the Shaanxi Provincial Natural Science Foundation (grant no. 2016JQ8011 to J. Zhou).

References

1 

Rini BI, Campbell SC and Escudier B: Renal cell carcinoma. Lancet. 373:1119–1132. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Hollingsworth JM, Miller DC, Daignault S and Hollenbeck BK: Five-year survival after surgical treatment for kidney cancer: A population-based competing risk analysis. Cancer. 109:1763–1768. 2007. View Article : Google Scholar : PubMed/NCBI

3 

McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, Kirkwood JM, Gordon MS, Sosman JA, Ernstoff MS, et al: Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J Clin Oncol. 23:133–141. 2005. View Article : Google Scholar

4 

Kroeger N, Choueiri TK, Lee JL, Bjarnason GA, Knox JJ, MacKenzie MJ, Wood L, Srinivas S, Vaishamayan UN, Rha SY, et al: Survival outcome and treatment response of patients with late relapse from renal cell carcinoma in the era of targeted therapy. Eur Urol. 65:1086–1092. 2014. View Article : Google Scholar

5 

Koshkin VS and Rini BI: Emerging therapeutics in refractory renal cell carcinoma. Expert Opin Pharmacother. 17:1225–1232. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Mains PE, Kemphues KJ, Sprunger SA, Sulston IA and Wood WB: Mutations affecting the meiotic and mitotic divisions of the early Caenorhabditis elegans embryo. Genetics. 126:593–605. 1990.PubMed/NCBI

7 

Kent D, Bush EW and Hooper JE: Roadkill attenuates Hedgehog responses through degradation of Cubitus interruptus. Development. 133:2001–2010. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Liu J, Ghanim M, Xue L, Brown CD, Iossifov I, Angeletti C, Hua S, Nègre N, Ludwig M, Stricker T, et al: Analysis of Drosophila segmentation network identifies a JNK pathway factor overexpressed in kidney cancer. Science. 323:1218–1222. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Zhang Q, Zhang L, Wang B, Ou CY, Chien CT and Jiang J: A hedgehog-induced BTB protein modulates hedgehog signaling by degrading Ci/Gli transcription factor. Dev Cell. 10:719–729. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Kwon JE, La M, Oh KH, Oh YM, Kim GR, Seol JH, Baek SH, Chiba T, Tanaka K, Bang OS, et al: BTB domain-containing speckle-type POZ protein (SPOP) serves as an adaptor of Daxx for ubiquitination by Cul3-based ubiquitin ligase. J Biol Chem. 281:12664–12672. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Hernández-Muñoz I, Lund AH, van der Stoop P, Boutsma E, Muijrers I, Verhoeven E, Nusinow DA, Panning B, Marahrens Y and van Lohuizen M: Stable X chromosome inactivation involves the PRC1 Polycomb complex and requires histone MACROH2A1 and the CULLIN3/SPOP ubiquitin E3 ligase. Proc Natl Acad Sci USA. 102:7635–7640. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Gordan JD, Lal P, Dondeti VR, Letrero R, Parekh KN, Oquendo CE, Greenberg RA, Flaherty KT, Rathmell WK, Keith B, et al: HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma. Cancer Cell. 14:435–446. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, Shimamura T, Sato-Otsubo A, Nagae G, Suzuki H, et al: Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 45:860–867. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Su D, Stamatakis L, Singer EA and Srinivasan R: Renal cell carcinoma: Molecular biology and targeted therapy. Curr Opin Oncol. 26:321–327. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Li G, Ci W, Karmakar S, Chen K, Dhar R, Fan Z, Guo Z, Zhang J, Ke Y, Wang L, et al: SPOP promotes tumorigenesis by acting as a key regulatory hub in kidney cancer. Cancer Cell. 25:455–468. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Zhou J, Luo J, Wu K, Yun EJ, Kapur P, Pong RC, Du Y, Wang B, Authement C, Hernandez E, et al: Loss of DAB2IP in RCC cells enhances their growth and resistance to mTOR-targeted therapies. Oncogene. Feb 15–2016.(Epub ahead of print). View Article : Google Scholar

17 

Zhou J, Zhu G, Huang J, Li L, Du Y, Gao Y, Wu D, Wang X, Hsieh JT, He D, et al: Non-canonical GLI1/2 activation by PI3K/AKT signaling in renal cell carcinoma: A novel potential therapeutic target. Cancer Lett. 370:313–323. 2016. View Article : Google Scholar

18 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Muglia VF and Prando A: Renal cell carcinoma: Histological classification and correlation with imaging findings. Radiol Bras. 48:166–174. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Eggener S: TNM staging for renal cell carcinoma: Time for a new method. Eur Urol. 58:517–519; discussion 519–521. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Kang Y and Massagué J: Epithelial-mesenchymal transitions: Twist in development and metastasis. Cell. 118:277–279. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Tania M, Khan MA and Fu J: Epithelial to mesenchymal transition inducing transcription factors and metastatic cancer. Tumour Biol. 35:7335–7342. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Sánchez-Tilló E, de Barrios O, Siles L, Cuatrecasas M, Castells A and Postigo A: β-catenin/TCF4 complex induces the epithelial-to-mesenchymal transition (EMT)-activator ZEB1 to regulate tumor invasiveness. Proc Natl Acad Sci USA. 108:19204–19209. 2011. View Article : Google Scholar

25 

Cohen HT and McGovern FJ: Renal-cell carcinoma. N Engl J Med. 353:2477–2490. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Al-Ahmadie HA, Alden D, Qin LX, Olgac S, Fine SW, Gopalan A, Russo P, Motzer RJ, Reuter VE and Tickoo SK: Carbonic anhydrase IX expression in clear cell renal cell carcinoma: An immunohistochemical study comparing 2 antibodies. Am J Surg Pathol. 32:377–382. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Hes O, Michal M, Kuroda N, Martignoni G, Brunelli M, Lu Y, Adley BP, Alvarado-Cabrero I and Yang XJ: Vimentin reactivity in renal oncocytoma: Immunohistochemical study of 234 cases. Arch Pathol Lab Med. 131:1782–1788. 2007.PubMed/NCBI

28 

Ozcan A, Zhai J, Hamilton C, Shen SS, Ro JY, Krishnan B and Truong LD: PAX-2 in the diagnosis of primary renal tumors: Immunohistochemical comparison with renal cell carcinoma marker antigen and kidney-specific cadherin. Am J Clin Pathol. 131:393–404. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Huber MA, Kraut N and Beug H: Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol. 17:548–558. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Peinado H, Quintanilla M and Cano A: Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: Mechanisms for epithelial mesenchymal transitions. J Biol Chem. 278:21113–21123. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Kypta RM and Waxman J: Wnt/β-catenin signalling in prostate cancer. Nat Rev Urol. 9:418–428. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 49 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao W, Zhou J, Deng Z, Gao Y and Cheng Y: SPOP promotes tumor progression via activation of β-catenin/TCF4 complex in clear cell renal cell carcinoma. Int J Oncol 49: 1001-1008, 2016
APA
Zhao, W., Zhou, J., Deng, Z., Gao, Y., & Cheng, Y. (2016). SPOP promotes tumor progression via activation of β-catenin/TCF4 complex in clear cell renal cell carcinoma. International Journal of Oncology, 49, 1001-1008. https://doi.org/10.3892/ijo.2016.3609
MLA
Zhao, W., Zhou, J., Deng, Z., Gao, Y., Cheng, Y."SPOP promotes tumor progression via activation of β-catenin/TCF4 complex in clear cell renal cell carcinoma". International Journal of Oncology 49.3 (2016): 1001-1008.
Chicago
Zhao, W., Zhou, J., Deng, Z., Gao, Y., Cheng, Y."SPOP promotes tumor progression via activation of β-catenin/TCF4 complex in clear cell renal cell carcinoma". International Journal of Oncology 49, no. 3 (2016): 1001-1008. https://doi.org/10.3892/ijo.2016.3609