Purpose: Previous study identified E2F1 as a key mediator of non–muscle-invasive bladder cancer (NMIBC) progression. The aim of this study was to identify the E2F1-related genes associated with poor prognosis and aggressive characteristics of bladder cancer.

Experimental Design: Microarray analysis was performed to find E2F1-related genes associated with tumor progression and aggressiveness in the gene expression data from 165 primary patients with bladder cancer. The biologic activity of E2F1-related genes in tumor progression and aggressiveness was confirmed with experimental assays using bladder cancer cells and tumor xenograft assay.

Results: The expression of E2F1 was significantly associated with EZH2 and SUZ12. The overexpression of E2F1, EZH2, and SUZ12 enhanced cancer progression including cell colony formation, migration, and invasiveness. Knockdown of these genes reduced motility, blocked invasion, and decreased tumor size in vivo. E2F1 bound the proximal EZH2 and SUZ12 promoter to activate transcription, suggesting that E2F1 and its downstream effectors, EZH2 and SUZ12, could be important mediators for the cancer progression. In addition, we confirmed an association between these genes and aggressive characteristics. Interestingly, the treatment of anticancer drugs to the cells overexpressing E2F1, EZH2, and SUZ12 induced the expression of CD44, KLF4, OCT4, and ABCG2 known as cancer stem cell (CSC)–related genes.

Conclusions: The link between E2F1, EZH2, and/or SUZ12 revealed that E2f1 directly regulates transcription of the EZH2 and SUZ12 genes. The signature of E2F1EZH2SUZ12 shows a predictive value for prognosis in bladder tumors and the E2F1EZH2SUZ12–driven transcriptional events may regulate the cancer aggressiveness and chemo-resistance, which may provide opportunity for development of new treatment modalities. Clin Cancer Res; 21(23); 5391–403. ©2015 AACR.

Translational Relevance

Non–muscle-invasive bladder cancer (NMIBC) accounts for 80% of bladder cancers, 20% of which experience the progression into muscle-invasive bladder cancer (MIBC) that is responsible for the most cancer-specific deaths. In this study, the activation of E2F1–EZH2–SUZ12 signature was strongly associated with NMIBC-to-MIBC progression. The signature to discriminate distinct molecular subgroups of NMIBC was developed in a training cohort of from 165 patients with bladder cancer and validated in independent cohort. Moreover, we examined E2F1 downstream pathway mediating NMIBC progression and illustrated an association between the overexpression of E2F1–EZH2–SUZ12 and chemoresistance. Thus, we suggest that the transcriptional changes of E2F1–EZH2–SUZ12 clearly predict bladder cancer aggressiveness, as well as anticancer drug resistance. Identification of a high-risk subgroup of patients with NMIBC based on the E2F1–EZH2–SUZ12 signature may improve the application of currently available treatments and provide opportunities for the development of new treatment modalities.

Bladder cancer is the sixth most common cancer in men and women populations. In 2013, 72,570 new cases of bladder cancer were diagnosed and 15,210 deaths were due to bladder cancer in the United States (1). This cancer is characterized by 2 histologically distinct subtypes: non–muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) at initial diagnosis (2). NMIBC is a heterogeneous disease (3), and patients frequently experience disease recurrence and 10% to 30% of them progress to MIBC, which is responsible for most bladder cancer–specific deaths (2). Because MIBC frequently leads to distant metastases (4), a major focus of research has been to understand the mechanisms that promote cancer progression. Although there have been many efforts to construct a robust model to predict progression of NMIBC using clinical information and pathologic classification (3, 5–7), precisely predicting the behavior of heterogeneous NMIBC remains challenging.

Previously, our genome-wide gene expression profile study using microarray technologies successfully identified a gene expression signature that could predict the likelihood of progression of NMIBC (8). Expression of E2F1 was significantly upregulated in the MIBC subtype, strongly indicating that activation of E2F1 might be a critical genetic event in the development of or progression to MIBC (8). Because E2F1 was not uniformly absent in all NMIBCs, we re-examined expression of E2F1 in NMIBCs and subdivided the patients into 2 groups according to the expression level of E2F1. The progression rate in the E2F1-high groups was profoundly higher than in the E2F1-low group, showing that E2F1 is strongly associated with NMIBC-to-MIBC progression (8).

In this study, on the basis of gene-to-gene network analysis, we found that the expression levels of EZH2 and SUZ12, binding partners of polycomb complex PRC2 (polycomb repressive complex 2) and direct targets of E2F1, were significantly higher in the E2F1-high subgroup than in the E2F1-low subgroup. Overexpression of several PcG proteins has been associated with many tumors and has also been identified as prognostic indicator in several tumors (9–13). The PRC2 core components are known such as EZH2, SUZ12, EED, and RBBP4 or RBBP7, which catalyze trimethylation of histone H3 lysine 27 (H3K27me3; ref. 14). Several studies reported that PRC2 was overexpressed in numerous cancer types and played a critical role in the aberrant silencing of tumor suppressor genes (15, 16).

Higher expression of the EZH2 and SUZ12 genes is clearly associated with tumor progression and overall survival (OS) in bladder cancer, but other genes including EED did not show significant level of prediction in this study. Therefore, we investigated the biologic activities of EZH2 and SUZ12, whose expression was significantly associated with poor prognosis and reflected the aggressive characteristics of bladder cancer. Moreover, we examined these genes' downstream pathways to mediate NMIBC progression, illustrating that E2F1 and EZH2 activated cancer stem cell (CSC) signaling pathways in anticancer drug-treated environments. Thus, we suggest that the transcriptional changes of E2F1, EZH2, and SUZ12 clearly predict bladder cancer aggressiveness, as well as anticancer drug resistance.

Cell culture

Human bladder cancer cell lines (EJ and 5637) were obtained from the ATCC. Other cell lines (UC5 and UC9) were provided by H. Barton Grossman (Department of Urology, University of Texas MD Anderson Cancer Center, Houston, TX: deposited into Public Health England, United Kingdom). The cells in this study were used within 6 months in our laboratory and were obtained from a cell bank that performed cell line characterizations. Cells from ATCC were certificated by the results of the short tandem repeat (STR) DNA profiling assay, cytochrome c oxidase I assay, and mycoplasma contamination assay. Eleven of UC series cells were characterized by the STR-PCR method and for mycoplasma contamination.

UC5, UC9 (NMIBC cells), and EJ (MIBC cells) were cultured in DMEM (Hyclone) supplemented with 10% FBS (Hyclone) and 1% penicillin/streptomycin (Hyclone). 5637 (MIBC) cells were cultured in RPMI-1640 medium (Hyclone).

Microarray gene expression profiling

We used a gene expression dataset (GSE13507, n = 256) containing 165 primary patients with bladder cancer in a previous study (8). Among the 165 cancers, 102 were histopathologically proven to be primary NMIBC and remained 63 were primary MIBC [GSE13507; the Korean cohort, n = 165 (102 NMIBCs and 63 MIBCs)]. Clinical data including progression-free survival (PFS), updated in January 2010, were obtained from the Chungbuk National University Hospital. To validate a prognostic value of the signature, 3 other gene expression datasets of patients with bladder cancer from hospitals of the Swedish southern healthcare region [GSE32894, n = 308 (215 NMIBCs and 93 MIBCs); ref. 17], Skane University Hospital [GSE32548, n = 131 (93 NMIBCs and 38 MIBCs); ref. 18], and University Hospital of Lund [GSE19915, n = 146 (97 NMIBCs and 49 MIBCs); ref. 19] were collected. Among them, MIBC data from GSE32894 and GSE32548 were combined, and a total of 58 MIBCs, whose survival time data were available, were used to assess survival rate of MIBC. Additional gene expression dataset including 19 bladder cancer cell lines was also examined in this study (GSE48277, n = 349). All gene expression datasets (GSE13507, GSE32894, GSE32548, GSE19915, and GSE48277) were freely available at NCBI GEO database.

To estimate prognostic values (PFS of NMIBC and OS of MIBC) of a signature combined with E2F1, EZH2, and SUZ12 genes, we adopted a previously developed strategy using the Cox regression coefficient for the genes in the signature [prognostic index (PI); refs. 20, 21]. Additional analysis was carried out as described in Supplementary Methods S1. Gene network–based activation regulator analyses were performed using the Ingenuity Pathway Analysis (IPA) tool.

Plasmid construction and transfection

The plasmid construct was the pcDNA6-V5-His–tagged expression vector with fusion coding sequence (CDS) of E2F1, EZH2, and SUZ12 (Invitrogen) genes. Transfection of plasmids was carried out using the jetPRIME reagent (Polyplus Transfection Inc.) at a ratio of DNA to jetPRIME of 1:3 according to the manufacturer's protocol. The measurement of gene expression at 24 hours posttransfection was normalized with the corresponding empty vectors.

RT-PCR and real-time PCR

The M-MLV Reverse Transcription kit (Beams Biotechnology) along with 3 μg of total RNA and poly(dT) primers were used for synthesis of cDNA. RT-PCR was carried out using an Emerald Amp GT PCR Master Mix (Takara Bio Inc.) to detect the mRNA level of E2F1, EZH2, and SUZ12 with primer sets (in Supplementary Methods S2). PCR cycling conditions were 94°C for 2 minutes to activate DNA polymerase, followed by 25 to 28 cycles of 94°C for 30 seconds, 58°C for 20 seconds, and 72°C for 40 seconds, and 72°C for 7 minutes for postelongation. Real-time PCR was carried out TOPreal premix SYBR Green (Enzynomics) and β-actin was used as control.

Western blot analysis

Proteins from the 5637, EJ, and UC5 cells were homogenized in RIPA buffer containing protease inhibitor (Roche), and the protein concentration was determined by using the BCA Assay (Thermo Scientific; ref. 22). The antibodies used in immunoblotting were against E2F1 (A300-766A, Bethyl Laboratories), EZH2 (4905, Cell Signaling Technology Inc.) SUZ12 (A302-407A, Bethyl Laboratories), and β-actin (4967, Cell Signaling Technology Inc.). Immunoreactivity was detected using the ECL Detection System (GE Healthcare BioSciences Corp.). Films were exposed at multiple time points to ensure that images were not saturated.

RNAi assay

siRNAs targeting E2F1, EZH2, and SUZ12 were used: the SMARTpool ON-TARGET plus siE2F1 (L-003259-00; Dharmacon, GE Healthcare Bio-Sciences Corp.), siEZH2 (L-004218-00; Dharmacon), and siSUZ12 (L006957-00; Dharmacon). The SMARTpool ON-TARGET plus siControl nontargeting pool (D-001810-10) was purchased from Dharmacon. Cell were grown on 60-mm dishes and transfected either with control siRNA, siE2F1, siEZH2, or siSUZ12 (siRNA; 100 nmol/mL). The cells were analyzed 24 hours posttransfection.

We obtained shRNAs for E2F1, EZH2, and SUZ12 from Sigma-Aldrich (MISSION shRNA). Each shRNA for E2F1, EZH2, or SUZ12 was cloned into the pLKO.1-puro vector, using the Polymerase III U6-RNA promoter. A set of 5 shRNAs to each of E2F1, EZH2, and SUZ12 was tested for knockdown, and the shRNA containing the sequences for E2F1, EZH2, and SUZ12 was chosen for these experiments, because both mRNA and protein of E2f1, Ezh2, and Suz12 were effectively decreased. We used nontarget shRNA vector (Cat. No SHC016) as a control and selected stably expressing cells using puromycin (2 μg/mL).

MTT cell viability assay and soft-agar cologenic assay

Cell viability was detected using MTT assay. Cells were seeded in 96-well plates at a density of 1,000 cells per well, and then cells were incubated for 24 and 48 hours. Ten microliters of MTT (5 mg/mL; Sigma-Aldrich) was added to each well and incubated for 3 hours. At the end of the incubation, the supernatants were removed and 100 μL of dimethyl sulfoxide (Sigma-Aldrich) was added to each well, and absorbance at 490 nm was determined for each well using a Wallac Vector 1420 Multilabel Counter (EG&G Wallac). For each experimental condition, 3 wells were used.

For the colony formation assay, UC9 and EJ cells were transfected with expression vector or siRNAs. Trypsin-treated cells were suspended in medium containing DMEM or RPMI-1640 medium with 10% FBS, antibiotics, and 3 mL of 0.35% noble agar (Difco). Cells (1 × 105 cells/well) were plated onto a solidified medium containing 3 mL of 0.7% noble agar in a 60-mm dish. The dishes were incubated at 37°C with 5% CO2, and fresh medium was added every 4 to 5 days. UC9 were grown for 35 days, and EJ were incubated for 21 days before staining with 0.05% crystal violet. We counted forming colonies (>100 μm in diameter) using microscopy.

Invasion and migration assays and tumor xenograft assay

For cell invasion assays, we used a Boyden chamber (NeuroProbe) and membrane (8-μm pore size) precoated with growth factor–reduced Matrigel (BD Biosciences). After 24 hours of transfection, bladder cancer cells in 56 μL of medium without FBS were seeded (5 × 104 cell/well) in the upper chamber. In the lower chamber, 27 μL of medium with 0.1% FBS medium (5637, EJ) and 10% FBS medium (UC5, UC9) was added as a chemoattractant. Then, cells were incubated for 12 hours (5637, EJ) and 24 hours (UC5, UC9).

For cell migration assays, the procedure was similar to the cell invasion assay, except Transwell membranes precoated with collagen (Sigma-Aldrich) were used, and cells were incubated for 12 hours (5637, EJ) and 24 hours (UC5, UC9). After staining the membrane using Diff-Quik reagents (Sysmex Co.), cells adhering to the lower surface were counted using a light microscope at 50× and 200× magnification and at least 4 wells were selected for each experimental group.

For the tumor xenograft assay, 4-week-old male BALB/c nude mice were obtained from SLC (Japan SLC, Inc.) and maintained under pathogen-free conditions. Knockdown- or overexpressed cells (KD-EJ, 1 × 106 cells; UC9, 2 × 106 cells) were suspended in 100 μL PBS. Cells were injected subcutaneously into both flanks on the top and bottom of mice. Tumor diameters were measured every 3 days for 3 weeks postinjection using digital calipers. Tumor volume in cubic millimeters was calculated using the formula: (L × W2) × 0.52, where L is the maximum length and W is the maximum width.

Chromatin immunoprecipitation assay

Chromatin immunoprecipitation (ChIP) assay was carried out as previously described (22) with primer sets (in Supplementary Methods S2) for EZH2 and SUZ12 promoter used for the qPCR.

Chemoresistance assay

Cells were seeded at a density of 1 × 105 cells per well, and transfection of plasmids was carried out using the jetPRIME reagent (Polyplus Transfection Inc.) according to the manufacturer's protocol for 12 hours. After transfection, the medium was changed with 0 or 5 μmol/L of mitomycin C (MMC, Sigma) and 10 μmol/L of cisplatin (Dong-A ST) for 12 hours.

Biologic insights into the gene expression signature associated with disease progression

Using gene expression data of 102 NMIBCs in the Korean cohort (GSE13507; ref. 8), we selected in trans genes correlated with the E2F1 (total 1441 genes by the Pearson correlation test, P < 0.001, r > ∣0.4∣). Gene-to-gene network and upstream regulator analyses were performed using IPA tool displayed several important regulators with their effectors (i.e., gene networks) associated with disease progression of NMIBC (Supplementary Table S1). The path-exploring function of IPA revealed that an interconnection of network hubs composed of E2F1, EZH2, and SUZ12 is involved in a signaling pathway strongly associated with NMIBC progression (Fig. 1A). The patients in the Korean cohort were separated into the EH (E2F1 high expression) subgroup and the EL (E2F1 low expression) subgroup (two-sample t test, P < 0.001; Fig. 1B), indicating that activation of E2F1, the most predominant regulator, might be a key event associated with the progression of NMIBC. E2F1 was interconnected with another gene network hub composed around EZH2 that was interconnected with SUZ12 network (Fig. 1A). EZH2 was more highly expressed in the EH subgroup than in the EL subgroup (two-sample t test, P < 0.001; Fig. 1B). The expression of SUZ12 was also significantly higher in the EH subgroup than in the EL subgroup (two-sample t test, P < 0.001; Fig. 1B).

Figure 1.

Association between gene signature and PFS of NMIBC in the Korean cohort (n = 102). A, a predominant signaling pathway consisting of E2F1, EZH2, and SUZ12 associated with disease progression of NMIBC. Up- and downregulated genes in the EH group are indicated in red and green, respectively. The intensity of color is indicative of the degree of over- or underexpression. Each line and arrow represents functional and physical interactions between the genes and the direction of regulation reported in the literature. B, two-group boxplot comparing expression levels of E2F1, EZH2, and SUZ12 in EH (a high E2F1 cluster) and EL (a low E2F1 cluster) patients. Each P value was obtained by two-sample t test between EH and EL. C, survival estimation, NMIBC progression, MIBC OS, with a signature composed of 3 genes, E2F1, EZH2, and SUZ12.

Figure 1.

Association between gene signature and PFS of NMIBC in the Korean cohort (n = 102). A, a predominant signaling pathway consisting of E2F1, EZH2, and SUZ12 associated with disease progression of NMIBC. Up- and downregulated genes in the EH group are indicated in red and green, respectively. The intensity of color is indicative of the degree of over- or underexpression. Each line and arrow represents functional and physical interactions between the genes and the direction of regulation reported in the literature. B, two-group boxplot comparing expression levels of E2F1, EZH2, and SUZ12 in EH (a high E2F1 cluster) and EL (a low E2F1 cluster) patients. Each P value was obtained by two-sample t test between EH and EL. C, survival estimation, NMIBC progression, MIBC OS, with a signature composed of 3 genes, E2F1, EZH2, and SUZ12.

Close modal

The PFS analysis in NMIBCs (n = 102) using a signature combined with E2F1, EZH2, and SUZ12 (the 3-gene signature) showed a significant difference of progression rates in NMIBC between poor- and good-prognosis subgroups (P = 0.008; Fig. 1C, left). To validate a prognostic value of the signature of 3 genes in NMIBC progression, we tested the signature in independent patient cohorts (GSE32894, GSE32548, and GSE19915). Because all validation cohorts did not contain PFS time data, we alternatively validated the signature by receiver operating characteristic (ROC) analysis comparing PI scores and NMIBC progression events. High or moderate area under curve (AUC) values were observed in all 3 patient cohorts (AUCs = 0.71, 0.59, and 0.65 in GSE32894, GSE32548, and GSE19915, respectively; Supplementary Fig. S1), indicating that the 3-gene signature would be highly associated with NMIBC progression. Using PI scores, patients were divided into 2 groups (poor- or good-prognosis), in which proportions of disease progression were also assessed. Significant differences of progression between poor and good prognosis groups were obtained from the datasets except for GSE32548 (Fisher exact test: P < 0.001, P = 0.459, and P = 0.049 in GSE32894, GSE32548, and GSE19915, respectively). However, the ratios of disease progression in the poor-prognosis group (21.3%, 12.2%, and 22.2%) were still higher than in the good-prognosis group (5%, 5.8%, and 7%) in all 3 validation cohorts (GSE32894, GSE32548, and GSE19915), respectively (Supplementary Fig. S1). In addition to NMIBC progression, we also assessed OS of MIBC using the 3-gene signature. The OS of MIBC (n = 63) in the poor-prognosis subgroup was also significantly worse than that in the good-prognosis subgroup (P = 0.021; Fig. 1C, right). For validation in OS of MIBC, we tested the signature in an independent combined cohort with GSE32984 and GSE32548, in which the survival rate of MIBC in the poor-prognosis subgroup classified by the signature of 3 genes was significantly worse than that in the good-prognosis subgroup (P = 0.044; Supplementary Fig. S2A).

To provide comparative results with other signatures, we additionally illustrated Supplementary Fig. S2 and described a comparative analysis between the signatures in “Comparison of other signatures with the three-gene signature” subsection in Supplementary Text S1. Because a previously published signature for predicting progression of NMIBC (23) consisted of small number of genes (the 11-gene signature) like our signature, we tried to compare them (Supplementary text S1). As shown in Supplementary Text and Supplementary Fig. S2, the 3-gene signature is validated a significant prognostic value.

Characterization of bladder cancer cell lines by unsupervised hierarchical clustering analysis and functional study

We tested the characteristics of various bladder cancer cell lines that were derived from diverse stages of bladder cancer tissues. Unsupervised hierarchical clustering analysis of gene expression data from 19 bladder cancer cell lines yielded 3 major clusters, one representing the more aggressive (MIBC-like) and the other less aggressive (NMIBC-like) cancer cells (Supplementary Fig. S3A). These gene expression patterns may reflect the molecular configurations that are readily distinguishable between more aggressive (MIBC-like) and less aggressive (NMIBC-like) cancer cells.

To determine the microarray data that divided cases into NMIBC and MIBC subgroups, the biologic characteristics of total 12 bladder cancer cell lines were assessed by their invasiveness (Supplementary Fig. S3B). UC5, UC9, UC1, and UC6 cell lines showed characteristics of NMIBC, whereas others (UC3, UC10, UC14, 5637, EJ, T24, KU7, J82) were similar to MIBC (Supplementary Fig. S3B). Thus, we selected 4 cell lines as representative members in 2 characterized subgroups (NMIBC and MIBC) for further studies; UC5 and UC9 were NMIBC and EJ and 5637 were MIBC cells. The expression levels of E2F1, EZH2, and SUZ12 in the 2 groups of bladder cancer cells were evaluated by quantitative RT-PCR and Western blot assay. The mRNA expression of E2F1, EZH2, and SUZ12 in NMIBC was significantly lower than in MIBC cells (P < 0.01; Supplementary Fig. S3C). Protein levels of these genes were also increased in MIBC cells compared with NMIBC cells (Supplementary Fig. S3D). Thus, higher expression of these genes is clearly preserved in invasive cancer cells (Supplementary Fig. S3C and S3D), indicating possible mechanisms that contribute to progression to invasive or metastatic bladder tumors.

The E2F1–EZH2–SUZ12 signature is strongly associated with the progression of noninvasive tumors to invasive tumors

To determine whether the E2F1–EZH2–SUZ12 signature mediates progression from NMIBC to MIBC, we performed a number of in vitro and in vivo assays. As previously described, we examined the endogenous expression levels of E2F1, EZH2, and SUZ12 and compared them between NMIBC and MIBC (Supplementary Fig. S3C and S3D). Expression changes in the overexpressed cells with E2F1, EZH2, and SUZ12 may reflect cancer cell invasion and/or migration properties if these genes are strongly related to tumor progression. Comparisons of the expression levels of these genes between UC9 cells transfected with the pcDNA6 control vector and with pE2F1, pEZH2, or pSUZ12 are shown in Fig. 2A. Cells overexpressing E2F1, EZH2, and SUZ12 showed significant increases in both invasion and migration (Fig. 2B). In addition, in NMIBC UC5 cells, the effects of increasing the expression of E2F1, EZH2, and SUZ12 were also examined (Supplementary Fig. S4).

Figure 2.

Alternative expression of E2F1, EZH2, and SUZ12 regulates invasion and migration in bladder cancer. A and B, effects of overexpression of E2F1, EZH2, and SUZ12 genes in UC9 cells. A, cells overexpressing pE2F1, pEZH2, or pSUZ12 compared with cells with control vector (pcDNA6). Upregulation of mRNA and protein expression were detected by RT-PCR and Western blotting. B, cell overexpressing E2F1, EZH2, and SUZ12 showed increased cell migration and invasion. C and D, effects of knockdown (KD) of E2F1, EZH2, and SUZ12 genes in EJ cells. C, downregulation of mRNA and protein expression was detected in cells with siE2F1, siEZH2, or siSUZ12 compared with cells with control (scRNA). Downregulated E2F1, EZH2, and SUZ12 cells showed reduced invasion and migration. Data are presented as mean ± SD for 3 independent experiments (original magnification, 200×). ***, P < 0.001.

Figure 2.

Alternative expression of E2F1, EZH2, and SUZ12 regulates invasion and migration in bladder cancer. A and B, effects of overexpression of E2F1, EZH2, and SUZ12 genes in UC9 cells. A, cells overexpressing pE2F1, pEZH2, or pSUZ12 compared with cells with control vector (pcDNA6). Upregulation of mRNA and protein expression were detected by RT-PCR and Western blotting. B, cell overexpressing E2F1, EZH2, and SUZ12 showed increased cell migration and invasion. C and D, effects of knockdown (KD) of E2F1, EZH2, and SUZ12 genes in EJ cells. C, downregulation of mRNA and protein expression was detected in cells with siE2F1, siEZH2, or siSUZ12 compared with cells with control (scRNA). Downregulated E2F1, EZH2, and SUZ12 cells showed reduced invasion and migration. Data are presented as mean ± SD for 3 independent experiments (original magnification, 200×). ***, P < 0.001.

Close modal

We also investigated the effects of silencing E2F1, EZH2, and SUZ12 expression using siRNA in EJ (Fig. 2C). EJ cells with decreased E2F1, EZH2, and SUZ12 expression displayed a significant decrease in invasiveness and migration (Fig. 2D). These results demonstrate that the increased expression of E2F1, EZH2, and SUZ12 is related to the invasiveness and migratory characters of bladder cancer cells. In addition, MIBC-5637 was also examined to confirm the effect of silencing of E2F1, EZH2, and SUZ12 (Supplementary Fig. S5).

Redundant role of elevated E2F1, EZH2, and SUZ12 in proliferation, viability, and tumorigenesis of NMIBC cells

To investigate whether the E2F1, EZH2, and SUZ12 affect cell proliferation and viability of NMIBC cells, cell proliferation was evaluated by counting the number of cells every day. As shown in Fig. 3A, top (left), E2F1-, EZH2-, or SUZ12-overexpressing UC9 significantly promoted cell proliferation compared with pcDNA. Otherwise, the depletion of these genes by siRNAs suppressed proliferation of EJ cancer cells (Fig. 3A, top right). We also found that the viability of cells overexpressing these genes was significantly higher than in the control group at 96 hours (Fig. 3A, bottom). In contrast, depletion of these genes by siRNA suppressed the viability of EJ cancer cells, compared with siControl-transfected cells at 72 and 96 hours (Fig. 3A, bottom right). These results suggest that elevated these genes may be related to bladder cancer cell proliferation and viability.

Figure 3.

Effect of alternative expression of E2F1, EZH2, and SUZ12 on cell proliferation and tumorigenesis. A, UC9 cells overexpressing E2F1, EZH2, or SUZ12 showed significantly higher cell proliferation and cell viability than that of empty vector–transfected cells (pcDNA6; left). The siE2F1, siEZH2, or siSUZ12-KD EJ cells showed decreased cell proliferation and cell viability (right). B, tumor volume from xenografted nude mice with control shRNA, shE2F1, shEZH2, or shSUZ12 cell lines. Injection of shE2F1, shEZH2, or shSUZ12 cell lines showed a significant decrease in tumor growth compared with control-treated groups. Volumes of tumors dissected from the sacrificed mice (*, P < 0.01). The decreased RNA and protein levels from the resected tumors are represented in right. C, tumor volume from xenografted nude mice with transfected UC5 cells with control pCDNA, pE2F1, pEZH2, or pSUZ12 overexpression vector. Injection of transfected pE2F1, pEZH2, or pSUZ12 cell lines showed a significant increase in tumor growth compared with control pCDNA groups. Volumes of tumors dissected from the sacrificed mice (*, P < 0.01). The increased RNA and protein levels from the resected tumors are represented in right.

Figure 3.

Effect of alternative expression of E2F1, EZH2, and SUZ12 on cell proliferation and tumorigenesis. A, UC9 cells overexpressing E2F1, EZH2, or SUZ12 showed significantly higher cell proliferation and cell viability than that of empty vector–transfected cells (pcDNA6; left). The siE2F1, siEZH2, or siSUZ12-KD EJ cells showed decreased cell proliferation and cell viability (right). B, tumor volume from xenografted nude mice with control shRNA, shE2F1, shEZH2, or shSUZ12 cell lines. Injection of shE2F1, shEZH2, or shSUZ12 cell lines showed a significant decrease in tumor growth compared with control-treated groups. Volumes of tumors dissected from the sacrificed mice (*, P < 0.01). The decreased RNA and protein levels from the resected tumors are represented in right. C, tumor volume from xenografted nude mice with transfected UC5 cells with control pCDNA, pE2F1, pEZH2, or pSUZ12 overexpression vector. Injection of transfected pE2F1, pEZH2, or pSUZ12 cell lines showed a significant increase in tumor growth compared with control pCDNA groups. Volumes of tumors dissected from the sacrificed mice (*, P < 0.01). The increased RNA and protein levels from the resected tumors are represented in right.

Close modal

To obtain in vivo insight for these observations, nude mice were inoculated with E2F1-KD, EZH2-KD, or SUZ12-KD cell lines. The decreased mRNA and protein levels of shE2F1, shEZH2, or shSUZ12 cells are shown in Supplementary Fig. S6. All mice inoculated with shE2F1, shEZH2, or shSUZ12 cell lines showed a significant decrease in tumor volume compared with control-treated groups (Fig. 3B). Decreased expression levels of E2F1, EZH2, or SUZ12 from the resected tumors were also detected (Fig. 3B, right).

To examine the redundant role of elevated E2F1–EZH2–SUZ12 in proliferation and tumorigenesis in NMIBC cells, we also performed the reverse (overexpression) model in a tumor xenograft assay (Fig. 3C). All mice inoculated with overexpressed elevated E2F1–EZH2–SUZ12 cells showed a significant increase in tumor volume compared with control (pcDNA6) groups (Fig. 3C). Increased mRNA and protein levels from the resected tumors were also represented (Fig. 3C, right).

The alternative expressions of EZH2 and SUZ12 are regulated by E2F1 in bladder cancer cells

To identify whether E2F1 directly regulates transcription of the EZH2 and SUZ12 genes, we used the EZH2 and SUZ12 promoter vectors to drive a luciferase reporter gene in transient cotransfections with an E2F1 expression plasmid in 5637. Ectopic E2F1 strongly upregulated transcription from the both promoters in the 5637 (Fig. 4A). To determine the in vivo interaction of E2F1 with 3 potential E2F1-binding sites in the EZH2 promoter region or 1 potential E2F1-binding site in the SUZ12 promoter region (Fig. 4B), ChIP assays were performed using an E2F1 antibody. The appropriate EZH2 and SUZ12 promoter regions were immunoprecipitated with the E2F1 antibody (Fig. 4C and D). ChIP-qPCR analyses revealed that PCR fragments containing the potential E2F1-binding site at 3 regions in the EZH2 promoter and 1 region in the SUZ12 promoter were markedly increased in DNA samples from E2F1-transfected cells compared with DNA from pcDNA-transfected cells (Fig. 4C and D). No detectable band was observed in the control IgG precipitations.

Figure 4.

E2F1 regulates the expression of EZH2 and SUZ12 by directly binding to their promoter region. A, the E2F1-binding site on the promoter region of EZH2 or SUZ12 was confirmed by luciferase assay in 5637 cells after cotransfection with pE2F1 and a plasmid (EZH2-pro or SUZ12-pro) containing a fragment of the promoter region of EZH2 or SUZ12. B, putative binding sites for E2F1 were found in the promoter regions of EZH2 (−710/−701, −248/−240, and −80/−72) and one site in SUZ12 (−429/421). ChIP-qPCR analyses were performed in untreated UC5 and 5637 cells (C), whereas it was examined in 5637 cells with transfected pcDNA or pE2F1 (D) using the E2F1 antibody. C, the top 2 represent fold enrichment with E2F1 antibody compared with IgG in the promoter regions of EZH2 and SUZ12 versus NTS region in untreated UC5 and 5637 cells, and the bottom shows the relative binding in 5637 cells compared with UC5 cells. D, the top 2 represent fold enrichment with E2F1 antibody compared with IgG with the pcDNA or pE2F1 activities in 5637 cells, and the bottom shows the relative binding in the pE2F1 activities versus pcDNA controls. Data represent the mean ± SE of 3 independent experiments. *, P < 0.05 (t test).

Figure 4.

E2F1 regulates the expression of EZH2 and SUZ12 by directly binding to their promoter region. A, the E2F1-binding site on the promoter region of EZH2 or SUZ12 was confirmed by luciferase assay in 5637 cells after cotransfection with pE2F1 and a plasmid (EZH2-pro or SUZ12-pro) containing a fragment of the promoter region of EZH2 or SUZ12. B, putative binding sites for E2F1 were found in the promoter regions of EZH2 (−710/−701, −248/−240, and −80/−72) and one site in SUZ12 (−429/421). ChIP-qPCR analyses were performed in untreated UC5 and 5637 cells (C), whereas it was examined in 5637 cells with transfected pcDNA or pE2F1 (D) using the E2F1 antibody. C, the top 2 represent fold enrichment with E2F1 antibody compared with IgG in the promoter regions of EZH2 and SUZ12 versus NTS region in untreated UC5 and 5637 cells, and the bottom shows the relative binding in 5637 cells compared with UC5 cells. D, the top 2 represent fold enrichment with E2F1 antibody compared with IgG with the pcDNA or pE2F1 activities in 5637 cells, and the bottom shows the relative binding in the pE2F1 activities versus pcDNA controls. Data represent the mean ± SE of 3 independent experiments. *, P < 0.05 (t test).

Close modal

To further define the mechanistic link between E2F1 and EZH2 or SUZ12, we tested whether expression levels of EZH2 or SUZ12 showed a transient change in E2F1-overexpressing UC9 (pE2F1, Fig. 5A, left) and in siE2F1-treated EJ (siE2F1, Fig. 5A, right). These experiments demonstrated that the overexpression of E2F1 activity induced EZH2 and SUZ12 expression, and the loss of E2F1 activity reduced EZH2 and SUZ12 expression (Fig. 5A). Consistent with the ChIP assays, these results suggest that E2F1 directly regulates transcription of the EZH2 and SUZ12.

Figure 5.

E2F1 controls expression of EZH2 and SUZ12 in bladder cancer cells. A, E2F1 KD decreases the expression of EZH2 and SUZ12 in bladder cancer cells. Left, UC9 cells show upregulation of EZH2 and SUZ12 by E2F1 overexpression. Right, EJ cells show underexpressed EZH2 and SUZ12 expression with E2F1-targeted siRNA (siE2F1) compared with scrambled control (scRNA). B, E2F1 KD (shE2F1) leads to downregulation of EZH2 and SUZ12 when compared with controls (shCon). Downregulation was suppressed by pcDNA6-EZH2 or -SUZ12 overexpression vectors, but not by pcDNA empty vector. C, representative images of entire invaded and stained chambers are shown. Decreased invasion of EJ cells with E2F1-targeted shRNA (shE2F1#1, #2) compared with scrambled shRNA (NTS). Reduced invasive ability was effectively suppressed by EZH2 (+pEZH2) or SUZ12 (+pSUZ12) overexpression but not by pcDNA empty vector. D, results shown on the graph represent means ± SD of 3 independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 5.

E2F1 controls expression of EZH2 and SUZ12 in bladder cancer cells. A, E2F1 KD decreases the expression of EZH2 and SUZ12 in bladder cancer cells. Left, UC9 cells show upregulation of EZH2 and SUZ12 by E2F1 overexpression. Right, EJ cells show underexpressed EZH2 and SUZ12 expression with E2F1-targeted siRNA (siE2F1) compared with scrambled control (scRNA). B, E2F1 KD (shE2F1) leads to downregulation of EZH2 and SUZ12 when compared with controls (shCon). Downregulation was suppressed by pcDNA6-EZH2 or -SUZ12 overexpression vectors, but not by pcDNA empty vector. C, representative images of entire invaded and stained chambers are shown. Decreased invasion of EJ cells with E2F1-targeted shRNA (shE2F1#1, #2) compared with scrambled shRNA (NTS). Reduced invasive ability was effectively suppressed by EZH2 (+pEZH2) or SUZ12 (+pSUZ12) overexpression but not by pcDNA empty vector. D, results shown on the graph represent means ± SD of 3 independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

To assess whether the lack of E2F1 is complemented by EZH2 or SUZ12 in cancer progression, we independently overexpressed the EZH2 or SUZ12 in E2F1-KD cells. In both E2F1-KD cells (shE2F1#1 and shE2F1#2), mRNA expression of EZH2 or SUZ12 was reduced compared with controls (shCon), whereas it was effectively restored by EZH2 or SUZ12 overexpression (Fig. 5B). Then, to verify whether the invasion ability of E2F1 was also rescued by EZH2 or SUZ12, we determined the level of invasion of E2F1-KD cells and cells overexpressing EZH2 or SUZ12 in E2F1-KD cells. E2F1-KD decreased invasion ability, which was significantly restored by EZH2 or SUZ12 overexpression (Fig. 5C and D).

Elevated E2F1, EZH2, and SUZ12 expression is related to sphere formation and chemoresistance in bladder cancer cells

Recent investigations demonstrated that tumorigenicity and tumor progression are driven by CSC characteristics, and the expression of EZH2 was consistently upregulated in CSCs (24–26). It also has been reported that SUZ12 is important for the function of CSCs, and ectopic expression of SUZ12 in transformed cells is sufficient to generate CSCs (27, 28). To elucidate the relationship between the E2F1–EZH2–SUZ12 signature and the CSC characteristics, we analyzed the ability of sphere formation and chemoresistance in bladder cancer cells.

Colony-forming assay was performed to verify whether the elevated expression of E2F1, EZH2, or SUZ12 is critical for sphere formation. Overexpression of E2F1, EZH2, or SUZ12 in UC9 significantly increased the number of large colony (>100 μm of diameter) formation compared with control (pcDNA6-empty; Fig. 6A, top). Otherwise, the depletion of E2F1, EZH2, or SUZ12 in the invasive EJ cells by each siRNAs for E2F1, EZH2, or SUZ12 decreased the number of large colony formation compared with control treated with scRNA (Fig. 6B).

Figure 6.

Colony formation ability and CSC signatures were compared between cells with alternative expressed E2F1, EZH2, or SUZ12 vector and control cells. A, representative images of colony formation of UC9 bladder cancer cells transfected with pcDNA6-V5-His-empty vector and pcDNA6-V5-His-E2F1, -EZH2, or -SUZ12 overexpression vectors. Quantitative analysis of colony numbers is shown as a graph that counted colonies larger than 100 μm in diameter. B, the number of colonies was reduced in EJ cells treated with siE2F1, siEZH2, or siSUZ12 compared with cells treated with siControl. C, UC9 transfected with pcDNA, pE2F1, pEZH2, or pSUZ12 and EJ treated with siE2F1, siEZH2, or siSUZ12 were treated with MMC (5 μmol/L, top) and cisplatin (10 μmol/L, bottom). UC9 cells transfected with pE2F1, pEZH2, or pSUZ12 showed significantly higher cell viability than that of empty vector–transfected cells (pcDNA) by the MTT assay. Controversially, EJ cells treated with siRNA represented significantly lower cell viability than control (scRNA). , pcDNA or scRNA; , pE2F1 or siE2F1; Δ, pEZH2 or siEZH2; , pSUZ12, siSUZ12. D, CSC signatures were analyzed under anticancer drug-treated conditions. UC9 cells (transfected with pcDNA, pE2F1, pEZH2, or pSUZ12) and EJ cells (treated with scRNA, siE2F1, siEZH2, and siSUZ12) were treated with 5 μmol/L MMC. Values are the mRNA expression levels in MMC-treated cells relative to untreated cells. Results shown on the graph represent means ± SD of 3 independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 6.

Colony formation ability and CSC signatures were compared between cells with alternative expressed E2F1, EZH2, or SUZ12 vector and control cells. A, representative images of colony formation of UC9 bladder cancer cells transfected with pcDNA6-V5-His-empty vector and pcDNA6-V5-His-E2F1, -EZH2, or -SUZ12 overexpression vectors. Quantitative analysis of colony numbers is shown as a graph that counted colonies larger than 100 μm in diameter. B, the number of colonies was reduced in EJ cells treated with siE2F1, siEZH2, or siSUZ12 compared with cells treated with siControl. C, UC9 transfected with pcDNA, pE2F1, pEZH2, or pSUZ12 and EJ treated with siE2F1, siEZH2, or siSUZ12 were treated with MMC (5 μmol/L, top) and cisplatin (10 μmol/L, bottom). UC9 cells transfected with pE2F1, pEZH2, or pSUZ12 showed significantly higher cell viability than that of empty vector–transfected cells (pcDNA) by the MTT assay. Controversially, EJ cells treated with siRNA represented significantly lower cell viability than control (scRNA). , pcDNA or scRNA; , pE2F1 or siE2F1; Δ, pEZH2 or siEZH2; , pSUZ12, siSUZ12. D, CSC signatures were analyzed under anticancer drug-treated conditions. UC9 cells (transfected with pcDNA, pE2F1, pEZH2, or pSUZ12) and EJ cells (treated with scRNA, siE2F1, siEZH2, and siSUZ12) were treated with 5 μmol/L MMC. Values are the mRNA expression levels in MMC-treated cells relative to untreated cells. Results shown on the graph represent means ± SD of 3 independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

To determine the chemoresistance which is known as one of CSC characteristics, the cell viability of E2F1-, EZH2-, or SUZ12-overexpressing cells was determined after the treatment of 5 μmol/L MMC (Fig. 6C, top) and 10 μmol/L cisplatin (Fig. 6C, bottom). Overexpression of E2F1, EZH2, or SUZ12 in UC9 and UC5 significantly increased the viability compared with control, whereas the depletion of E2F1, EZH2, or SUZ12 in the invasive EJ cells by each siRNAs decreased viability compared with control (Fig. 6C and Supplementary Fig. S8). Furthermore, mRNA levels of CD44, KLF4, OCT4, and ABCG2 known as CSC markers (29–32) significantly affected in under MMC- or cisplatin-treated conditions in bladder cancer cells with altered E2F1, EZH2, and SUZ12 expression than in matched control group (Fig. 6D and Supplementary Fig. S7).

According to the success of recent genome-wide gene expression profile studies (33–35), we previously reported a prognostic signature for predicting the progression of superficial tumors to invasive ones (8). The higher biologic activity of E2F1 suggests that it may be the major driving force during the progression of bladder cancer. Gene network analyses of the signature revealed that E2F1 and its downstream effectors EZH2 and SUZ12 could be important mediators for the invasive and metastatic progression of superficial tumors (Fig. 1). Consistent with other cancers, the relationship between cancer progression and the overexpression of these genes was observed (36–41). Recently, Santos and colleagues (42) also reported that the increased tumor recurrence and progression in patients with NMIBC is associated with increased E2F and EZH2 expression.

Overexpression of EZH2 and SUZ12 is directly controlled by E2F1 (Fig. 4), and their expression is associated with poor prognosis and indicative of invasion and metastasis in many cancers (Figs. 2 and 3). Our results show that these prognostic molecules can predict the likelihood of progression of NMIBC. Furthermore, unequal distribution of expression patterns reflecting activation of E2F1 in subgroups (EL, EH) with different progression rates supports the notion that distinct molecular features of the tumor govern the clinical phenotypes of NMIBC. As a result, we speculate that the overexpression of E2F1–EZH2–SUZ12 may play a role in proliferation, migration, and invasion of cancer cells.

It has been known that CSC characteristics may lead to cancer aggressiveness, chemoresistance to anticancer drugs, and a high risk of recurrence in patients with cancer (29, 31, 32, 43). Recent reports suggest that tumorigenicity and tumor progression is driven by CSC characteristics, and CSCs consistently showed elevated EZH2 and SUZ12 expression (24–28). It also has been reported that EZH2 and SUZ12 is important for the function of CSCs and its ectopic expression in transformed cells is sufficient to generate CSCs (24–28). Thus, we investigated the association of the E2F1–EZH2–SUZ12 downstream targets with the characteristics of CSCs. To identify the characteristics of CSCs, the abilities of sphere formation and chemoresistance were determined. The capacity of sphere formation is determined by both the proliferation rate and cell adhesion ability and has been used to identify the characteristic of CSCs in the previous studies (44–46). In this study, overexpression of E2F1–EZH2–SUZ12 increased the formation of large sphere, and the depletion of these genes decreased the formation of large sphere, which suggest that these genes might play important roles in sphere formation. Also, an increase in viability of the E2F1–EZH2–SUZ12–overexpressing cells under the treatment of MMC or cisplatin reflects that E2F1, EZH2, or SUZ12 might be related with resistance of the cells to anticancer drug. In addition, the activation of stem cell–like molecules (CD44, OCT4, KLF4, and ABCG2) was detected in bladder cancer cells overexpressing E2F1, EZH2, and SUZ12. Moreover, the activation of ABCG2 and CD44 could be related to the chemoresistance of the cells and, consequently, enriching CSCs by drug treatment might induce cancer aggressiveness (30, 47, 48).

We would suggest that the activation of EZH2 and SUZ12 expression under the control of E2F1 might play a role of switch in the development of bladder cancer according to tumor microenvironment. If the tumor microenvironment is favorable for cancer growth, the overexpression of EZH2–SUZ12 signature by E2F1 regulation contributes to the proliferation and invasiveness of bladder cancer cells. However, under the condition of anticancer drug treatment, the EZH2–SUZ12 signature by E2F1 control might activate CSCs signatures. As results, overexpressed E2F1–EZH2–SUZ12 cells have significantly higher capacities for sphere formation and activate the stem cell–like molecules when cells were put on an anticancer drug-treated condition.

Taken together, the elevated E2F1–EZH2–SUZ12 expression in bladder cancer cells might play important roles in proliferation, migration, and invasiveness. Moreover, the enrichment of cells with the characteristics of CSCs by overexpression of these genes might play critical roles in chemoresistance and tumorigenicity, which might be associated with poor prognosis of bladder cancer cells. Therefore, our findings show that a prognostic molecular signature, E2F1–EZH2–SUZ12, can predict the likelihood of progression of NMIBC. Furthermore, our study could provide useful information to predict an individual's risk of progression and to establish a suitable chemotreatment for disease.

No potential conflicts of interest were disclosed.

Conception and design: J. Heo, I.-S. Chu, S.-H. Leem

Development of methodology: H.-H. Lee, I.-S. Chu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.-R. Lee, Y.-G. Roh, S.-K. Kim, J.-S. Lee, S.-Y. Seol, H.-H. Lee, W.-T. Kim, I.-S. Chu

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.-K. Kim, J.-S. Lee, J. Heo, T.-H. Kang, I.-S. Chu, S.-H. Leem

Writing, review, and/or revision of the manuscript: S.-R. Lee, Y.-G. Roh, S.-K. Kim, J. Heo, T.-H. Kang, I.-S. Chu, S.-H. Leem

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): W.-J. Kim, H.-J. Cha, J.W. Chung, I.-S. Chu, S.-H. Leem

Study supervision: J. Heo, I.-S. Chu, S.-H. Leem

This research was supported by the Mid-Career Researcher Program through the National Research Foundation of Korea (NRF) grant (NRF-2013R1A2A2A04008115), an NRF grant (2008-0062611 and 2011-0019745) funded by the Korea government (MEST), and a grant from the KRIBB Research Initiative Program.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
R
,
Naishadham
D
,
Jemal
A
. 
Cancer statistics, 2013
.
CA Cancer J Clin
2013
;
63
:
11
30
.
2.
Youssef
RF
,
Lotan
Y
. 
Predictors of outcome of non-muscle-invasive and muscle-invasive bladder cancer
.
ScientificWorldJournal
2011
;
11
:
369
81
.
3.
Pow-Sang
JM
,
Seigne
JD
. 
Contemporary management of superficial bladder cancer
.
Cancer Control
2000
;
7
:
335
9
.
4.
Messing
EM
. 
Urothelial tumors of the bladder
. In:
Wein
AJ
,
Kavoussi
LR
,
Novick
AC
,
Partin
AW
,
Peters
CA
,
editor
. 
Campbell-Walsh's urology
.
Philadelphia, PA
:
Saunders
; 
2007
. p.
2417
30
.
5.
Herr
HW
. 
Tumour progression and survival in patients with T1G3 bladder tumours: 15-year outcome
.
Br J Urol
1997
;
80
:
762
5
.
6.
Holmang
S
,
Hedelin
H
,
Anderstrom
C
,
Holmberg
E
,
Busch
C
,
Johansson
SL
. 
Recurrence and progression in low grade papillary urothelial tumors
.
J Urol
1999
;
162
:
702
7
.
7.
Vedder
MM
,
Marquez
M
,
de Bekker-Grob
EW
,
Calle
ML
,
Dyrskjot
L
,
Kogevinas
M
, et al
Risk prediction scores for recurrence and progression of non-muscle invasive bladder cancer: an international validation in primary tumours
.
PloS One
2014
;
9
:
e96849
.
8.
Lee
JS
,
Leem
SH
,
Lee
SY
,
Kim
SC
,
Park
ES
,
Kim
SB
, et al
Expression signature of E2F1 and its associated genes predict superficial to invasive progression of bladder tumors
.
J Clin Oncol
2010
;
28
:
2660
7
.
9.
van Leenders
GJLH
,
Dukers
D
,
Hessels
D
,
van den Kieboom
SWM
,
Hulsbergen
CA
,
Witjes
JA
, et al
Polycomb-group oncogenes EZH2, BMI1, and RING1 are overexpressed in prostate cancer with adverse pathologic and clinical features
.
Eur Urol
2007
;
52
:
455
63
.
10.
Friedman
JM
,
Liang
GN
,
Liu
CC
,
Wolff
EM
,
Tsai
YC
,
Ye
W
, et al
The putative tumor suppressor microRNA-101 modulates the cancer epigenome by repressing the polycomb group protein EZH2
.
Cancer Res
2009
;
69
:
2623
9
.
11.
Varambally
S
,
Dhanasekaran
SM
,
Zhou
M
,
Barrette
TR
,
Kumar-Sinha
C
,
Sanda
MG
, et al
The polycomb group protein EZH2 is involved in progression of prostate cancer
.
Nature
2002
;
419
:
624
9
.
12.
Bachmann
IM
,
Halvorsen
OJ
,
Collett
K
,
Stefansson
IM
,
Straume
O
,
Haukaas
SA
, et al
EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast
.
J Clin Oncol
2006
;
24
:
268
73
.
13.
Kleer
CG
,
Cao
Q
,
Varambally
S
,
Shen
R
,
Ota
I
,
Tomlins
SA
, et al
EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells
.
Proc Natl Acad Sci U S A
2003
;
100
:
11606
11
.
14.
Chase
A
,
Cross
NC
. 
Aberrations of EZH2 in cancer
.
Clin Cancer Res
2011
;
17
:
2613
8
.
15.
Herranz
N
,
Pasini
D
,
Diaz
VM
,
Franci
C
,
Gutierrez
A
,
Dave
N
, et al
Polycomb complex 2 is required for E-cadherin repression by the Snail1 transcription factor
.
Mol Cell Biol
2008
;
28
:
4772
81
.
16.
Villa
R
,
Pasini
D
,
Gutierrez
A
,
Morey
L
,
Occhionorelli
M
,
Vire
E
, et al
Role of the polycomb repressive complex 2 in acute promyelocytic leukemia
.
Cancer Cell
2007
;
11
:
513
25
.
17.
Sjodahl
G
,
Lauss
M
,
Lovgren
K
,
Chebil
G
,
Gudjonsson
S
,
Veerla
S
, et al
A molecular taxonomy for urothelial carcinoma
.
Clin Cancer Res
2012
;
18
:
3377
86
.
18.
Lindgren
D
,
Sjodahl
G
,
Lauss
M
,
Staaf
J
,
Chebil
G
,
Lovgren
K
, et al
Integrated genomic and gene expression profiling identifies two major genomic circuits in urothelial carcinoma
.
PLoS One
2012
;
7
:
e38863
.
19.
Lindgren
D
,
Frigyesi
A
,
Gudjonsson
S
,
Sjodahl
G
,
Hallden
C
,
Chebil
G
, et al
Combined gene expression and genomic profiling define two intrinsic molecular subtypes of urothelial carcinoma and gene signatures for molecular grading and outcome
.
Cancer Res
2010
;
70
:
3463
72
.
20.
Paik
S
,
Shak
S
,
Tang
G
,
Kim
C
,
Baker
J
,
Cronin
M
, et al
A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer
.
N Engl J Med
2004
;
351
:
2817
26
.
21.
Kim
SM
,
Leem
SH
,
Chu
IS
,
Park
YY
,
Kim
SC
,
Kim
SB
, et al
Sixty-five gene-based risk score classifier predicts overall survival in hepatocellular carcinoma
.
Hepatology
2012
;
55
:
1443
52
.
22.
Kim
DH
,
Roh
YG
,
Lee
HH
,
Lee
SY
,
Kim
SI
,
Lee
BJ
, et al
The E2F1 Oncogene Transcriptionally Regulates NELL2 in Cancer Cells
.
DNA Cell Biol
2013
;
32
:
517
23
.
23.
Catto
JW
,
Abbod
MF
,
Wild
PJ
,
Linkens
DA
,
Pilarsky
C
,
Rehman
I
, et al
The application of artificial intelligence to microarray data: identification of a novel gene signature to identify bladder cancer progression
.
Eur Urol
2010
;
57
:
398
406
.
24.
Crea
F
,
Hurt
EM
,
Mathews
LA
,
Cabarcas
SM
,
Sun
L
,
Marquez
VE
, et al
Pharmacologic disruption of Polycomb Repressive Complex 2 inhibits tumorigenicity and tumor progression in prostate cancer
.
Mol Cancer
2011
;
10
:
40
.
25.
Suva
ML
,
Riggi
N
,
Janiszewska
M
,
Radovanovic
I
,
Provero
P
,
Stehle
JC
, et al
EZH2 is essential for glioblastoma cancer stem cell maintenance
.
Cancer Res
2009
;
69
:
9211
8
.
26.
Chang
CJ
,
Yang
JY
,
Xia
W
,
Chen
CT
,
Xie
X
,
Chao
CH
, et al
EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-beta-catenin signaling
.
Cancer Cell
2011
;
19
:
86
100
.
27.
Iliopoulos
D
,
Lindahl-Allen
M
,
Polytarchou
C
,
Hirsch
HA
,
Tsichlis
PN
,
Struhl
K
. 
Loss of miR-200 inhibition of Suz12 leads to polycomb-mediated repression required for the formation and maintenance of cancer stem cells
.
Mol Cell
2010
;
39
:
761
72
.
28.
Martin-Perez
D
,
Sanchez
E
,
Maestre
L
,
Suela
J
,
Vargiu
P
,
Di Lisio
L
, et al
Deregulated expression of the polycomb-group protein SUZ12 target genes characterizes mantle cell lymphoma
.
Am J Pathol
2010
;
177
:
930
42
.
29.
Kumar
SM
,
Liu
S
,
Lu
H
,
Zhang
H
,
Zhang
PJ
,
Gimotty
PA
, et al
Acquired cancer stem cell phenotypes through Oct4-mediated dedifferentiation
.
Oncogene
2012
;
31
:
4898
911
.
30.
Lu
Y
,
Zhu
H
,
Shan
H
,
Lu
J
,
Chang
X
,
Li
X
, et al
Knockdown of Oct4 and Nanog expression inhibits the stemness of pancreatic cancer cells
.
Cancer Lett
2013
;
340
:
113
23
.
31.
Lee
M
,
Nam
EJ
,
Kim
SW
,
Kim
S
,
Kim
JH
,
Kim
YT
. 
Prognostic impact of the cancer stem cell-related marker NANOG in ovarian serous carcinoma
.
Int J Gynecol Cancer
2012
;
22
:
1489
96
.
32.
Hiraga
T
,
Ito
S
,
Nakamura
H
. 
Cancer stem-like cell marker CD44 promotes bone metastases by enhancing tumorigenicity, cell motility, and hyaluronan production
.
Cancer Res
2013
;
73
:
4112
22
.
33.
Alizadeh
AA
,
Eisen
MB
,
Davis
RE
,
Ma
C
,
Lossos
IS
,
Rosenwald
A
, et al
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling
.
Nature
2000
;
403
:
503
11
.
34.
Lee
JS
,
Chu
IS
,
Heo
J
,
Calvisi
DF
,
Sun
Z
,
Roskams
T
, et al
Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling
.
Hepatology
2004
;
40
:
667
76
.
35.
van ‘t Veer
LJ
,
Dai
H
,
van de Vijver
MJ
,
He
YD
,
Hart
AA
,
Mao
M
, et al
Gene expression profiling predicts clinical outcome of breast cancer
.
Nature
2002
;
415
:
530
6
.
36.
Gu
Y
,
Cheng
Y
,
Song
Y
,
Zhang
Z
,
Deng
M
,
Wang
C
, et al
MicroRNA-493 suppresses tumor growth, invasion and metastasis of lung cancer by regulating E2F1
.
PloS One
2014
;
9
:
e102602
.
37.
Alla
V
,
Engelmann
D
,
Niemetz
A
,
Pahnke
J
,
Schmidt
A
,
Kunz
M
, et al
E2F1 in melanoma progression and metastasis
.
J Natl Cancer Inst
2010
;
102
:
127
33
.
38.
Rabello
DD
,
Lucena-Araujo
AR
,
Alves-Silva
JC
,
da Eira
VB
,
de Vasconcellos
MC
,
de Oliveira
FM
, et al
Overexpression of EZH2 associates with a poor prognosis in chronic lymphocytic leukemia
.
Blood Cells Mol Dis
2015
;
54
:
97
102
.
39.
Xu
L
,
Deng
Q
,
Pan
Y
,
Peng
M
,
Wang
X
,
Song
L
, et al
Cancer-associated fibroblasts enhance the migration ability of ovarian cancer cells by increasing EZH2 expression
.
Int J Mol Med
2014
;
33
:
91
6
.
40.
Fan
Y
,
Shen
B
,
Tan
M
,
Mu
X
,
Qin
Y
,
Zhang
F
, et al
TGF-beta-induced upregulation of malat1 promotes bladder cancer metastasis by associating with suz12
.
Clin Cancer Res
2014
;
20
:
1531
41
.
41.
Foster
CS
,
Falconer
A
,
Dodson
AR
,
Norman
AR
,
Dennis
N
,
Fletcher
A
, et al
Transcription factor E2F3 overexpressed in prostate cancer independently predicts clinical outcome
.
Oncogene
2004
;
23
:
5871
9
.
42.
Santos
M
,
Martinez-Fernandez
M
,
Duenas
M
,
Garcia-Escudero
R
,
Alfaya
B
,
Villacampa
F
, et al
In vivo disruption of an Rb-E2F-Ezh2 signaling loop causes bladder cancer
.
Cancer Res
2014
;
74
:
6565
77
.
43.
Obinata
D
,
Takayama
K
,
Urano
T
,
Murata
T
,
Kumagai
J
,
Fujimura
T
, et al
Oct1 regulates cell growth of LNCaP cells and is a prognostic factor for prostate cancer
.
Int J Cancer
2012
;
130
:
1021
8
.
44.
Borena
BM
,
Meyer
E
,
Chiers
K
,
Martens
A
,
Demeyere
K
,
Broeckx
SY
, et al
Sphere-forming capacity as an enrichment strategy for epithelial-like stem cells from equine skin
.
Cell Physiol Biochem
2014
;
34
:
1291
303
.
45.
Liang
X
,
Osman
TA
,
Sapkota
D
,
Neppelberg
E
,
Lybak
S
,
Liavaag
PG
, et al
Rapid adherence to collagen IV enriches for tumour initiating cells in oral cancer
.
Eur J Cancer
2014
;
50
:
3262
70
.
46.
Wang
JL
,
Yu
JP
,
Sun
ZQ
,
Sun
SP
. 
Radiobiological characteristics of cancer stem cells from esophageal cancer cell lines
.
World J Gastroenterol
2014
;
20
:
18296
305
.
47.
Yamashina
T
,
Baghdadi
M
,
Yoneda
A
,
Kinoshita
I
,
Suzu
S
,
Dosaka-Akita
H
, et al
Cancer stem-like cells derived from chemoresistant tumors have a unique capacity to prime tumorigenic myeloid cells
.
Cancer Res
2014
;
74
:
2698
709
.
48.
Oshima
N
,
Yamada
Y
,
Nagayama
S
,
Kawada
K
,
Hasegawa
S
,
Okabe
H
, et al
Induction of cancer stem cell properties in colon cancer cells by defined factors
.
PloS One
2014
;
9
:
e101735
.

Supplementary data