Abstract
Gene amplification is a hallmark of cancer and is frequently observed in colorectal cancer. Previous whole-genome sequencing of colorectal cancer clinical specimens identified amplification of Ring finger protein 6 (RNF6), a RING-domain E3 ubiquitin ligase. In this study, we showed that RNF6 is upregulated in 73.5% (147/200) of patients with colorectal cancer and was positively associated with RNF6 gene amplification. Furthermore, RNF6 expression and its gene amplification were independent prognostic factors for poor outcome of patients with colorectal cancer. RNF6 promoted cell growth, cell-cycle progression, and epithelial-to-mesenchymal transition in colorectal cancer cells; RNF6 also promoted colorectal tumor growth and lung metastasis in mouse models. Mechanistic investigations revealed that RNF6 bound and ubiquitylated transducin-like enhancer of split 3 (TLE3), a transcriptional repressor of the β-catenin/TCF4 complex. RNF6-mediated degradation of TLE3 significantly suppressed the association of TLE3 with TCF4/LEF, which in turn led to recruitment of β-catenin to TCF4/LEF, triggering Wnt/β-catenin activation. Restoration of TLE3 expression abolished the oncogenic effects of RNF6. Taken together, these results demonstrate that RNF6 plays a pivotal oncogenic role in colorectal tumorigenesis.
Significance: RNF6-mediated ubiquitination and degradation of TLE3 activates the Wnt/β-catenin pathway in colorectal carcinogenesis. Cancer Res; 78(8); 1958–71. ©2018 AACR.
Introduction
Colorectal cancer is the third most common cancer and a major cause of cancer mortality worldwide (1). Both the incidence and death rate of colorectal cancer are increasing rapidly and maintaining an upward trend in Asian countries (2). The genetic mechanisms in colorectal cancer initiation and progression are complex and heterogeneous, involving somatic gene mutations, abnormal gene fusion, deletion or amplification, and epigenetic alterations (3–5). Chromosomal instability (CIN), including somatic DNA copy number amplification, is a hallmark of cancer and is present in approximately 60% to 70% of sporadic colorectal cancer, and the CIN pathway is usually associated with a stepwise process of “adenoma to carcinoma sequence” (6–8). The somatic copy number alterations (SCNA)–high subtype, which represents nearly 60% of primary colorectal cancers, demonstrated significant overlap (62%) with Wnt and MYC activation (7). The Wnt/β-catenin signaling pathway plays a pivotal role in regulation of cell proliferation and cell migration in the intestinal tract and colorectal tumorigenesis (9). Despite the high prevalence of DNA copy number amplification, their role in the pathogenesis of colorectal cancer remains poorly understood. Gene amplification of oncogenes, leading to their overexpression, is one of the potential mechanisms in colorectal cancer (10). Thus, identification of novel driver genes aberrantly upregulated by copy number amplification may uncover oncogenic pathways underlying the initiation and progression of colorectal cancer and discover potential biomarkers for diagnosis, prognosis, and treatment of patients with colorectal cancer.
We recently performed whole-genome sequencing in paired colorectal cancer tissues and identified a novel amplification gene, Ring finger protein 6 (RNF6). RNF6 is located on chromosome 13q12.13, one of the most frequently amplified regions in colorectal cancer (11, 12). RNF6 is a RING-type E3 ubiquitin-protein ligase that belongs to the RNF family, which mediates the ubiquitination of its target proteins and tags them for proteasomal degradation (13, 14). Several RNF family members are involved in the regulation of cell proliferation and differentiation, and they function as oncogenes or tumor suppressors in cancer, depending on their target proteins and the cellular context (15–17). Some RNF proteins are potential prognostic biomarkers and novel therapeutic targets for cancer treatment (18–20). However, the molecular function of RNF6, its target protein substrates, its involvement in signal transduction in cancer, as well as its clinical significance in patients with colorectal cancer are still elusive.
In this study, we uncovered the oncogenic role of RNF6 on colorectal cancer cell functions and tumorigenicity. We examined the molecular mechanisms of RNF6 and revealed that RNF6 directly interacted with Groucho family member transducin-like enhancer of split 3 (TLE3), a critical transcriptional repressor of the β-catenin/TCF4 complex; RNF6 caused TLE3 ubiquitination and degradation, thereby promoting β-catenin recruitment to TCF4/LEF and activating the Wnt/β-catenin signaling cascade. We also unveiled the clinical significance of RNF6 in both the testing and validation cohorts of patients with colorectal cancer.
Materials and Methods
Colorectal cancer cell lines
The colon cancer cell lines (DLD-1, HCT116, HT29, LOVO, SW480, SW620, and SW1116) were obtained from the U.S. ATCC between 2014 and 2015, and cell authentication was confirmed by short tandem repeat profiling. Cells were routinely cultured and maintained in DMEM supplemented with 10% FBS and antibiotics (Gibco BRL) according to the ATCC protocols, except HCT116 cells, which were cultured in complete McCoy's 5A medium (Gibco BRL). Routine Mycoplasma testing was performed by PCR. Cells were grown for no more than 25 passages in total for any experiment.
Clinical samples
A total of 118 colorectal cancer tumor tissues (BeiJing cohort 1) and an additional 150 paired colorectal cancer tumor and adjacent nontumor tissue samples (BeiJing cohort 2) were obtained from Peking University Cancer Hospital, Beijing, China. Moreover, 50 paired primary colorectal cancer and adjacent normal colon tissues (HongKong cohort 1) and 160 primary colorectal cancer tumor tissues (HongKong cohort 2) were collected at Prince of Wales Hospital, The Chinese University of Hong Kong. Samples were confirmed histologically and staged according to the tumor–node–metastasis (TNM) staging system. Patients were followed up regularly, and the median follow-up duration since the time of diagnosis was 55.2 months (range 12.4 to 170 months). The clinicopathologic features of all the patients are provided in Supplementary Table S1. Colorectal cancer dataset was downloaded from The Cancer Genome Atlas (TCGA) colon and rectal cancer database (616 patients). The study was approved by the Clinical Research Ethics Committee of The Chinese University of Hong Kong; The Clinical Research Ethics Committee of Peking University Cancer Hospital and all subjects provided informed consent for obtaining the study specimens. This study was carried out in accordance with the Declaration of Helsinki of the World Medical Association.
Genomic DNA extraction and DNA copy number real-time PCR
Genomic DNA of colorectal cancer tissues was isolated using DNA Mini Kit (Qiagen) according to the manufacturer's protocol. RNF6-specific probe (Hs01093471_cn) was used to assess DNA copy number status in the colorectal cancer tissues. Real-time PCR was performed using TaqMan copy number master mixture (Thermo Fisher Scientific) on ViiA 7 real-time PCR instrument according to the protocol provided by company. Each sample was tested in triplicate. The results were analyzed in CopyCaller software to determine the copy number of the target gene.
Xenograft mouse model
For tumorigenicity assay, HCT116 cells (5 × 106) stably expressing RNF6 or empty vector and DLD-1 cells stably transfected with shRNF6 and shControl were suspended in 0.1-mL PBS and were subcutaneously injected into both dorsal flanks of 4-week-old male athymic Balb/c nude mice (4/group). The tumor volume was measured by the Vernier caliper and calculated according to the formula tumor volume (mm3) = (longer diameter × shorter diameter2)/2. The mice were monitored at indicated time points for tumor growth and sacrificed when the tumors reached a maximal diameter of 1.0 cm. Tumors were excised and weighed. The excised tissues were rapidly frozen in liquid nitrogen until further protein extraction. Tumor sections from paraffin-embedded blocks of the harvested tissues were immunostained with the indicated antibodies. All animal experiments were performed in accordance with guidelines approved by the Animal Ethics Committee of The Chinese University of Hong Kong. All animal studies were approved by the government of the Hong Kong Special Administrative Region Department of Health.
Standard tail vein metastatic assay
RNF6- or vector-stable expression of HCT116 cells were injected at a final concentration of 5 × 106 cells/100-μL PBS into the lateral tail veins of each 4-week-old male Balb/c nude mice (5/group). Mice were sacrificed after 28 days, and the lungs were dissected and fixed in 10% neutral-buffered formalin. Nude mice lung paraffin sections were prepared and analyzed by hematoxylin and eosin staining. Lung metastasis was quantified under microscopy. All animal experiments were performed in accordance with guidelines approved by the Animal Ethics Committee of The Chinese University of Hong Kong. All animal studies were approved by the government of the Hong Kong Special Administrative Region Department of Health.
Coimmunoprecipitation and mass spectrometry
SW480 cells or HCT116 cells were transfected with pcDNA3.1-RNF6–expressing vector and treated with 10 μmol/L MG132 (Cell Signaling Technology) for 10 hours; cell lysates were collected and immunoprecipitated with either 2 μg indicated or IgG antibody at 4°C overnight. The antibodies were then pulled down with 25 μL protein A/G magnetic beads (Thermo Fisher Scientific) for 4 hours at 4°C. After extensive washing, proteins were eluted with low-pH buffer and separated by SDS-PAGE gel, followed by silver staining. Candidate bands were subjected to mass spectrometry analysis for protein identification.
Ubiquitination assay
RNF6 stably transfected HT29 and HCT116 cells or RNF6-depleted SW480 cells and control cells were transfected with HA-ubiquitin plasmid. Two days after transfection, cells were treated with 10 μmol/L MG132 for 10 hours to block proteasomal degradation. Cell lysates were pulled down with either 2 μg HA-tag or IgG antibody. Eluates were separated by SDS-PAGE and immunoblotted with antibodies against TLE3, RNF6, and GAPDH, respectively.
Luciferase reporter activity analysis
Indicated colorectal cancer cells (1 × 105 cells/well) were cotransfected with TOPflash luciferase reporter plasmid (200 ng/well), pRL-CMV vector (5 ng/well), and RNF6 plasmid or control empty vector for overexpression study, or shRNF6 and control shRNA for knockdown study using Lipofectamine 2000 transfection reagent (Life Technologies). TLE3 plasmid and siTLE3 and corresponding controls cotransfect with TOPflash luciferase reporter conduct in the rescue experiments. Cells were harvested at 48 hours posttransfection and luciferase activities were quantified by the Dual-Luciferase Reporter Assay System (Promega). The experiments were conducted three times in triplicates.
Statistical analysis
The results were expressed as mean ± SD. The Pearson correlation coefficient was used to evaluate the correlation between RNF6 amplification and its expression in the clinical samples. Crude relative risks (RR) of prognostic factors associated with RNF6 expression and other predictor variables were estimated by using a univariate Cox proportional hazards regression model and multivariate Cox model. Overall survival or recurrence rate in relation to RNF6 expression or copy number status was evaluated by the Kaplan–Meier survival analysis and the log-rank test. The RNF6 amplification rate between groups was tested byχ2 test. A two-tailed, unpaired, or paired Student t test was performed to compare the variables of two groups, and ANOVA was used for multigroup comparisons. The difference in cell viability and tumor growth rate was determined by repeated measures ANOVA. P < 0.05 was regarded as statistically significant.
Other experimental methods are provided in the Supplementary Information. All primer sequences and the antibodies used and their dilutions are listed in the Supplementary Methods (Supplementary Tables S2 and S3).
Results
RNF6 is overexpressed in primary colorectal tumors
To identify driver oncogenes deregulated by DNA copy number amplification in colorectal cancer, we performed whole-genome sequencing in paired colorectal cancer and adjacent normal samples, which revealed that RNF6 was one of the top outlier genes overexpressed in colorectal cancer, with a high copy ratio compared with baseline copy ratio (Fig. 1A). RNF6 mRNA expression was examined in two independent cohorts of paired colorectal cancer and adjacent normal tissues from Hong Kong (n = 50) and Beijing (n = 150) by real-time PCR. RNF6 mRNA expression was significantly higher in tumor tissues as compared with their adjacent normal tissues in both Hong Kong (37/50, 74%) and Beijing cohorts (110/150, 73.3%; 147/200, 73.5%, in total; Fig. 1B). Consistent with our data, RNF6 mRNA was overexpressed in the TCGA dataset consisting of 380 patients with colorectal cancer (Fig. 1B). In keeping with the increased RNF6 mRNA, RNF6 protein expression was significantly higher in colorectal cancer tumors as compared with adjacent normal tissues in 15 paired colorectal cancer samples by IHC (Fig. 1C).
RNF6 is overexpressed in colorectal cancer, and its overexpression is associated with poor prognosis. A, RNF6 copy number amplification was identified in colorectal cancer (CRC) by whole-genome sequencing. B, mRNA expression of RNF6 in three independent cohorts, including 50 paired samples of adjacent normal tissues and colorectal cancer tumor tissues from HongKong cohort 1, 150 paired samples from BeiJing cohort 2, and unpaired tissues from TCGA colorectal cancer cohort (50 normal and 380 tumor tissues). P < 0.0001, by two-tailed, unpaired, or paired Student t test. C, The protein expression of RNF6 was significantly higher in primary colorectal cancer tumors as compared with their adjacent normal tissue. RNF6 expression in 15 paired colorectal cancer and adjacent normal tissues from the HongKong cohort were assessed by IHC. P < 0.01, by two-tailed, paired Student t test. D, Kaplan–Meier survival analysis of overall survival according to RNF6 mRNA expression in 160 colorectal cancer patients of the testing cohort, which is comprised of the HongKong cohort 1 (one case without survival data) and BeiJing cohort 1 (seven cases without RNF6 mRNA expression). High RNF6 expression predicts poor overall survival in colorectal cancer. The receiver operating characteristic curve was used to define the cutoff, and log-rank analysis was used to test for significance. E, Kaplan–Meier recurrence risk analysis based on RNF6 mRNA expression in 160 colorectal cancer cases. RNF6 overexpression had a higher risk of recurrence. F, RNF6 expression in 160 colorectal cancer tissues of HongKong cohort 2 was assessed by IHC, and representative IHC staining for RNF6 is shown (left). Kaplan–Meier survival curve of overall survival and recurrence risk according to RNF6 nuclear expression status in colorectal cancer tissues; log-rank analysis was used to test for significance (right, three cases without survival data and two cases lost tissue). RNF6 nuclear staining higher than 10% was defined as moderate/strong (high), whereas lower than 10% was defined as negative/weak (low).
RNF6 is overexpressed in colorectal cancer, and its overexpression is associated with poor prognosis. A, RNF6 copy number amplification was identified in colorectal cancer (CRC) by whole-genome sequencing. B, mRNA expression of RNF6 in three independent cohorts, including 50 paired samples of adjacent normal tissues and colorectal cancer tumor tissues from HongKong cohort 1, 150 paired samples from BeiJing cohort 2, and unpaired tissues from TCGA colorectal cancer cohort (50 normal and 380 tumor tissues). P < 0.0001, by two-tailed, unpaired, or paired Student t test. C, The protein expression of RNF6 was significantly higher in primary colorectal cancer tumors as compared with their adjacent normal tissue. RNF6 expression in 15 paired colorectal cancer and adjacent normal tissues from the HongKong cohort were assessed by IHC. P < 0.01, by two-tailed, paired Student t test. D, Kaplan–Meier survival analysis of overall survival according to RNF6 mRNA expression in 160 colorectal cancer patients of the testing cohort, which is comprised of the HongKong cohort 1 (one case without survival data) and BeiJing cohort 1 (seven cases without RNF6 mRNA expression). High RNF6 expression predicts poor overall survival in colorectal cancer. The receiver operating characteristic curve was used to define the cutoff, and log-rank analysis was used to test for significance. E, Kaplan–Meier recurrence risk analysis based on RNF6 mRNA expression in 160 colorectal cancer cases. RNF6 overexpression had a higher risk of recurrence. F, RNF6 expression in 160 colorectal cancer tissues of HongKong cohort 2 was assessed by IHC, and representative IHC staining for RNF6 is shown (left). Kaplan–Meier survival curve of overall survival and recurrence risk according to RNF6 nuclear expression status in colorectal cancer tissues; log-rank analysis was used to test for significance (right, three cases without survival data and two cases lost tissue). RNF6 nuclear staining higher than 10% was defined as moderate/strong (high), whereas lower than 10% was defined as negative/weak (low).
A comparison of RNF6 gene expression in 30 cancer studies based on RNA sequencing results revealed that colorectal cancer is the top cancer associated with RNF6 upregulation (Supplementary Fig. S1A). Therefore, RNF6 can serve as a biomarker overexpressed in human colorectal cancer.
RNF6 overexpression is associated with poor prognosis in patients with colorectal cancer
We first evaluated the clinicopathologic features and prognostic significance of RNF6 in 160 patients with colorectal cancer (HongKong cohort 1 and BeiJing cohort 1) with follow-up information. Kaplan–Meier analysis showed that colorectal cancer patients with high RNF6 expression had significantly poorer survival compared with those with low expression, especially in early stage (I/II) patients with colorectal cancer, but not in advanced stage (III/IV) patients (Fig. 1D). Colorectal cancer patients with high RNF6 expression also showed a higher recurrence rate compared with those with low expression (Fig. 1E). In univariate Cox regression analysis, high RNF6 expression was associated with poor outcomes of patients with colorectal cancer [HR, 1.801; 95% confidence interval (CI), 1.100–2.949; P = 0.019]. Multivariate Cox regression analysis adjusting for age, gender, and TNM stage demonstrated that RNF6 high expression is an independent prognostic factor for poor survival of patients with colorectal cancer (HR 1.646, 95% CI, 1.002–2.703; P = 0.046; Supplementary Table S4). The independent cohort included paired samples of adjacent normal colon and colorectal cancer from 150 patients (BeiJing cohort 2, two cases without survival data). Kaplan–Meier analysis showed that high expression of RNF6 was correlated with poor overall survival (Supplementary Fig. S1B). Furthermore, multivariate Cox regression analysis revealed RNF6 expression to be an independent prognostic factor for poor survival in patients with colorectal cancer (Supplementary Table S5). We analyzed RNF6 mRNA (HongKong cohort 1) and protein (HongKong cohort 2) expression according to tumor stage (Supplementary Fig. S1C). Both mRNA and protein expression of RNF6 was increased in late TNM stage (III/IV) compared with early TNM stage (I/II), suggesting that RNF6 levels increased during tumor progression. The correlation analysis showed that the protein expression of RNF6 by IHC was positively correlated with its mRNA expression by qPCR (Supplementary Fig. S1D). RNF6 protein expression was also evaluated in 160 patients with colorectal cancer (HongKong cohort 2) by IHC tissue microarray. Kaplan–Meier analysis revealed that high RNF6 expression in colorectal cancer was correlated with poor overall survival and greater recurrence risk (Fig. 1F). Both mRNA and protein expression of RNF6 indicated a significantly worse prognosis, suggesting that RNF6 could be regarded as a valuable new prognostic factor for colorectal cancer.
RNF6 amplification contributes to its overexpression and is associated with colorectal cancer recurrence
The RNF6 gene is located on chromosome 13q12.13, a recurrent amplification region in colorectal cancer (11, 12). RNF6 is rarely mutated (1%, 6/631 cases) in colorectal cancer, although analysis of the TCGA pan-cancer dataset revealed that the RNF6 gene is uniquely amplified in colorectal cancer, but not in other human cancers across 12 cancer types (Fig. 2A). Thus, we speculated that gene amplification may be a major mechanism through which RNF6 is activated in colorectal cancer. Notably, there is an increased RNF6 amplification rate in colorectal cancer patients with mutant adenomatous polyposis coli (APC) compared with those with wild-type APC (Fig. 2B). RNF6 amplification occurs in the early stages (I/II) of colorectal tumorigenesis and further increases during colorectal cancer progression (stage III/IV; Fig. 2B). In our BeiJing cohort 1, we observed RNF6 copy number amplification in 33.1% (39/118) of primary colorectal cancer tumors by genomic DNA quantitative PCR (Fig. 2C). RNF6 mRNA levels were significantly higher in colorectal cancer with copy number amplification, and RNF6 mRNA showed positive correlation with its DNA copy number amplification (R = 0.698; P < 0.0001; Fig. 2D). Analysis of the TCGA dataset found that RNF6 DNA copy number amplification is present in 66.3% (390/616) of primary colorectal cancer samples and confirmed a positive association between RNF6 copy number amplification and its mRNA expression (R = 0.842; P < 0.0001; Fig. 2E). Moreover, colorectal cancer patients with RNF6 copy number amplification had a higher recurrence risk compared with those without, especially in early-stage colorectal cancer patients (Fig. 2F). Multivariate Cox regression analysis showed that RNF6 copy number amplification was an independent risk factor associated with high recurrence risk in patients with colorectal cancer (Supplementary Table S6). These data collectively indicate that RNF6 copy number amplification mediates RNF6 overexpression in colorectal cancer and predicts a high risk of recurrence in patients with colorectal cancer.
RNF6 amplification correlates with its upregulation and is associated with a greater risk of recurrence in colorectal cancer. A, Genomic alterations of 13q12.13 across 12 human cancer types in the TCGA pan-cancer dataset. RNF6 showed copy number amplification specifically in colon and rectal cancers. Red, copy number amplification; blue, copy number loss. BLCA, bladder urothelial carcinoma; BRCA, breast invasive carcinoma; CNV, copy number variation; COAD, colon adenocarcinoma; GBM, glioblastoma multiforme; HNSC, head and neck squamous cell carcinoma; KIRC, kidney renal clear cell carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; OV, ovarian serous cystadenocarcinoma; READ, rectum adenocarcinoma; STAD, stomach adenocarcinoma; UCEC, uterine corpus endometrioid carcinoma. B, RNF6 copy number variations were assessed in 297 APC wild-type and APC mutation patients from the TCGA colorectal cancer dataset. RNF6 copy number amplification proportion in early (stage I/II) and late (stage III/IV) tumor stage in the TCGA colorectal cancer cohort (n = 595; P < 0.0001, Chi-square test). C, RNF6 DNA copy number status was examined in 118 colorectal cancer tissues (BeiJing cohort 1) using RNF6-specific probe by genomic DNA real-time PCR. RNF6 was frequently amplified in colorectal cancer. D, RNF6 mRNA expression was higher in the RNF6 DNA copy number amplification group compared with the no amplification group [n = 111 (7 cases without RNF6 mRNA expression); P < 0.0001, two-tailed, unpaired Student t test]. RNF6 copy number more than or approximate to three copies labeled as copy number amplification. RNF6 copy number was positively correlated with its mRNA expression (n = 111; P < 0.0001, Pearson correlation coefficient analysis). E, RNF6 mRNA levels and DNA copy number status were assessed from the TCGA colorectal cancer database. RNF6 amplification was associated with RNF6 upregulation (n = 376; P < 0.0001, Pearson correlation coefficient analysis). F, Kaplan–Meier recurrence risk analysis based on RNF6 copy number status in 111 colorectal cancer tissues. RNF6 copy number amplification was associated with a greater risk of recurrence in patients with colorectal cancer. CRC, colorectal cancer.
RNF6 amplification correlates with its upregulation and is associated with a greater risk of recurrence in colorectal cancer. A, Genomic alterations of 13q12.13 across 12 human cancer types in the TCGA pan-cancer dataset. RNF6 showed copy number amplification specifically in colon and rectal cancers. Red, copy number amplification; blue, copy number loss. BLCA, bladder urothelial carcinoma; BRCA, breast invasive carcinoma; CNV, copy number variation; COAD, colon adenocarcinoma; GBM, glioblastoma multiforme; HNSC, head and neck squamous cell carcinoma; KIRC, kidney renal clear cell carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; OV, ovarian serous cystadenocarcinoma; READ, rectum adenocarcinoma; STAD, stomach adenocarcinoma; UCEC, uterine corpus endometrioid carcinoma. B, RNF6 copy number variations were assessed in 297 APC wild-type and APC mutation patients from the TCGA colorectal cancer dataset. RNF6 copy number amplification proportion in early (stage I/II) and late (stage III/IV) tumor stage in the TCGA colorectal cancer cohort (n = 595; P < 0.0001, Chi-square test). C, RNF6 DNA copy number status was examined in 118 colorectal cancer tissues (BeiJing cohort 1) using RNF6-specific probe by genomic DNA real-time PCR. RNF6 was frequently amplified in colorectal cancer. D, RNF6 mRNA expression was higher in the RNF6 DNA copy number amplification group compared with the no amplification group [n = 111 (7 cases without RNF6 mRNA expression); P < 0.0001, two-tailed, unpaired Student t test]. RNF6 copy number more than or approximate to three copies labeled as copy number amplification. RNF6 copy number was positively correlated with its mRNA expression (n = 111; P < 0.0001, Pearson correlation coefficient analysis). E, RNF6 mRNA levels and DNA copy number status were assessed from the TCGA colorectal cancer database. RNF6 amplification was associated with RNF6 upregulation (n = 376; P < 0.0001, Pearson correlation coefficient analysis). F, Kaplan–Meier recurrence risk analysis based on RNF6 copy number status in 111 colorectal cancer tissues. RNF6 copy number amplification was associated with a greater risk of recurrence in patients with colorectal cancer. CRC, colorectal cancer.
RNF6 promotes colorectal cancer cell proliferation in vitro and tumorigenesis in vivo
As compared with normal colon tissues, RNF6 was overexpressed in five of seven colorectal cancer cell lines as determined by RT-PCR and Western blot analysis (Fig. 3A). The frequent overexpression of RNF6 in colorectal cancer tumors and cell lines prompted us to investigate its oncogenic role in colorectal cancer. We generated HCT116 and HT29 cells stably expressing RNF6 or control vector, whereas RNF6-targeting siRNA or shRNA and corresponding controls were used to knockdown RNF6 in DLD-1 and SW480 cells with high endogenous RNF6 expression (Supplementary Fig. S2A). Ectopic RNF6 expression in HCT116 and HT29 cells significantly promoted cell proliferation, as evidenced by colony formation and cell growth curve assays. Conversely, RNF6 knockdown by siRNA or shRNA in DLD-1 and SW480 cells suppressed cell proliferation (Fig. 3B and C). To assess the effect of RNF6 on tumorigenesis in vivo, we performed subcutaneous xenograft assay in nude mice using HCT116 cells stably expressing RNF6 or empty vector. RNF6 overexpression significantly increased tumor growth and tumor weight (Fig. 3D). Western blot analysis confirmed RNF6 overexpression in the xenograft tumors (Fig. 3E). Moreover, enhanced cell proliferation and reduced cell apoptosis were demonstrated in RNF6-overexpressing tumors using Ki67 and TUNEL staining, respectively (Fig. 3F). On the other hand, knockdown of RNF6 in DLD-1 cells by shRNA inhibited tumor growth, concomitant with reduced cell proliferation (Ki67), and increased cell apoptosis (TUNEL; Supplementary Fig. S2B). These results suggest that RNF6 plays an important oncogenic role in promoting colorectal cancer cell growth and tumorigenicity.
RNF6 promotes colorectal cancer cell growth and induces tumorigenesis in vivo. A, RNF6 was frequently upregulated in colon cancer cell lines as determined by RT-PCR and Western blot analysis. B, Forced expression of RNF6 promoted cell growth, whereas cell growth was inhibited by RNF6 knockdown. C, Ectopic expression of RNF6 promoted colony formation in HT29 and HCT116 cells. Knockdown of RNF6 suppressed colony formation in DLD-1 and SW480 cells. D, Expression of RNF6 promoted tumor growth and increased tumor weight in nude mice (n = 4 per group). E, RNF6, cyclin D1, and active-β-catenin expression in xenograft tumors from RNF6-expressing nude mice was confirmed by Western blot analysis and parallelism controls. GAPDH was used as a loading control. F, Cell proliferation and cell apoptosis in tumor tissues isolated from RNF6-expressing and vector nude mice were determined by Ki67 and TUNEL staining, respectively. Cell proliferation index was quantified by counting the proportion of nuclear Ki67–positive cells. Apoptosis index was quantified by counting the proportion of TUNEL-positive cells. **, P < 0.01. Results are presented as mean ± SD of three independent experiments. CRC, colorectal cancer.
RNF6 promotes colorectal cancer cell growth and induces tumorigenesis in vivo. A, RNF6 was frequently upregulated in colon cancer cell lines as determined by RT-PCR and Western blot analysis. B, Forced expression of RNF6 promoted cell growth, whereas cell growth was inhibited by RNF6 knockdown. C, Ectopic expression of RNF6 promoted colony formation in HT29 and HCT116 cells. Knockdown of RNF6 suppressed colony formation in DLD-1 and SW480 cells. D, Expression of RNF6 promoted tumor growth and increased tumor weight in nude mice (n = 4 per group). E, RNF6, cyclin D1, and active-β-catenin expression in xenograft tumors from RNF6-expressing nude mice was confirmed by Western blot analysis and parallelism controls. GAPDH was used as a loading control. F, Cell proliferation and cell apoptosis in tumor tissues isolated from RNF6-expressing and vector nude mice were determined by Ki67 and TUNEL staining, respectively. Cell proliferation index was quantified by counting the proportion of nuclear Ki67–positive cells. Apoptosis index was quantified by counting the proportion of TUNEL-positive cells. **, P < 0.01. Results are presented as mean ± SD of three independent experiments. CRC, colorectal cancer.
RNF6 accelerates cell-cycle progression and inhibits apoptosis in colorectal cancer cells
To investigate the mechanism by which RNF6 promoted cell growth, we examined the effect of RNF6 on cell-cycle progression by flow cytometry. Ectopic expression of RNF6 in HT29 and HCT116 cells decreased the G0 to G1 phase cell population, with a corresponding increase in S-phase population (Fig. 4A), whereas RNF6 knockdown in DLD-1 and SW480 cells induced G0 to G1 arrest (Supplementary Fig. S3A), indicating that RNF6 promotes G1 to S transition. Consistently, Western blot analysis showed that RNF6 enhanced the expression of proliferating cell nuclear antigen (PCNA), the key factor in DNA replication and cell-cycle regulation (21), and cyclin D1 and cyclin-dependent kinase (CDK4), which promote G1 phase progression, but simultaneously reduced the expression of G1 gatekeepers, such as p53, p21Cip1, and p27Kip1 (Fig. 4B; refs. 22, 23). RNF6 knockdown in DLD-1 and SW480 cells had an opposite effect on the expression of these key cell-cycle regulators (Supplementary Fig. S3A).
RNF6 accelerates cell-cycle progression, inhibits cell apoptosis, and enhances tumor invasion and metastasis in vivo. A, RNF6 decreased cell population in G1 phase and increased cell population in S-phase in HT29 and HCT1116 cells. B, Ectopic expression of RNF6 in HCT116 and HT29 colon cancer cells inhibited cell apoptosis, as determined by Annexin V/7-AAD staining and flow cytometry. C, Ectopic expression of RNF6 enhanced the protein expression of PCNA, cyclin D1, and CDK4 while reducing the expression of p53, p27Kip1, and p21Cip1. RNF6 reduced protein expression of the active forms of caspase-8, caspase-9, caspase-3, caspase-7, and caspase-PARP by Western blot analysis. GAPDH was used as a loading control. D, Expression of RNF6 increased migratory capabilities of HT29 and HCT116 cells. Wound healing assay was performed, and representative images are shown. E, Expression of RNF6 promoted HT29 and HCT116 cell invasion. Matrigel invasion assay was performed, and representative images are shown. F, Representative images of lungs with or without metastasis. Hematoxylin and eosin (H&E) staining of lung tissues from nude mice injected with RNF6-expressing HCT116 cells and empty vector-expressing cells (magnification, ×200). The incidence of lung metastasis was quantified (right; n = 5 per group). G, The effect of ectopic expression of RNF6 on key EMT markers and EMT-associated transcriptional factors was determined by Western blot analysis. GAPDH was used as a loading control. *, P < 0.05; **, P < 0.01; ***, P < 0.001. Results are presented as mean ± SD of three independent experiments.
RNF6 accelerates cell-cycle progression, inhibits cell apoptosis, and enhances tumor invasion and metastasis in vivo. A, RNF6 decreased cell population in G1 phase and increased cell population in S-phase in HT29 and HCT1116 cells. B, Ectopic expression of RNF6 in HCT116 and HT29 colon cancer cells inhibited cell apoptosis, as determined by Annexin V/7-AAD staining and flow cytometry. C, Ectopic expression of RNF6 enhanced the protein expression of PCNA, cyclin D1, and CDK4 while reducing the expression of p53, p27Kip1, and p21Cip1. RNF6 reduced protein expression of the active forms of caspase-8, caspase-9, caspase-3, caspase-7, and caspase-PARP by Western blot analysis. GAPDH was used as a loading control. D, Expression of RNF6 increased migratory capabilities of HT29 and HCT116 cells. Wound healing assay was performed, and representative images are shown. E, Expression of RNF6 promoted HT29 and HCT116 cell invasion. Matrigel invasion assay was performed, and representative images are shown. F, Representative images of lungs with or without metastasis. Hematoxylin and eosin (H&E) staining of lung tissues from nude mice injected with RNF6-expressing HCT116 cells and empty vector-expressing cells (magnification, ×200). The incidence of lung metastasis was quantified (right; n = 5 per group). G, The effect of ectopic expression of RNF6 on key EMT markers and EMT-associated transcriptional factors was determined by Western blot analysis. GAPDH was used as a loading control. *, P < 0.05; **, P < 0.01; ***, P < 0.001. Results are presented as mean ± SD of three independent experiments.
We next determined the effect of RNF6 on the induction of apoptosis using Annexin V/7-aminoactinomycin D (7-AAD) staining and flow cytometry analysis. Ectopic RNF6 expression inhibited both early and late apoptosis in HT29 and HCT116 cells (Fig. 4C). On the contrary, RNF6 knockdown in DLD-1 and SW480 cells increased apoptosis induction (Supplementary Fig. S3B), consistent with an antiapoptotic function of RNF6. Suppression of apoptosis by RNF6 was further evidenced by the reduced protein expression of cleaved forms of caspase-8, caspase-9, caspase-3, caspase-7, and PARP in stably transfected HT29 and HCT116 cells as compared with controls (Fig. 4B), whereas RNF6 knockdown showed increased expression of these apoptosis factors (Supplementary Fig. S3B). Therefore, the oncogenic effect of RNF6 through suppression of cell apoptosis and promotion of cell-cycle progression in colorectal cancer cells.
RNF6 promotes cell migration/invasion and tumor metastasis in vivo
We next investigated the effect of RNF6 on metastasis using cell migration and invasion assays in vitro and experimental lung metastasis assay in vivo. Wound healing and Matrigel invasion assays showed that both the migratory and invasive capabilities of HT29 and HCT116 cells were increased by RNF6 overexpression (Fig. 4D and E). In contrast, RNF6 knockdown markedly abolished cell migration and invasion in DLD-1 and SW480 cells (Supplementary Fig. S3C). In vivo tail vein metastasis assay with HCT116 cells stably expressing RNF6 or control vector demonstrated that RNF6 significantly enhanced the incidence and number of lung metastasis nodules (Fig. 4F).
In keeping with the effect of RNF6 on cell migration/invasion and metastasis, HT29 and HCT116 cells expressing RNF6 demonstrated features of epithelial-to-mesenchymal transition (EMT) including increased expression of N-cadherin and vimentin but decreased expression of E-cadherin and Claudin-1 (Fig. 4G; refs. 24, 25). On the other hand, RNF6 knockdown in DLD-1 and SW480 reversed the EMT phenotype (Supplementary Fig. S3C). We performed Western blot analysis to determine the expression of EMT-associated transcriptional factors. In HCT116 and HT29 cells, RNF6 overexpression induced protein expression of SNAI1, ZEB1, and ZEB2 (Fig. 4G).
RNF6 interacts with TLE3 and induces TLE3 degradation via polyubiquitination
We next sought to determine the molecular mechanisms underlying the oncogenic effect of RNF6. To probe RNF6-interacting partners, we performed coimmunoprecipitation with endogenous RNF6 antibody to pull down potential interacting proteins from SW480 cells, followed by mass spectrometry for protein identification (Fig. 5A). The transcriptional corepressor TLE3 was found to be a high scoring candidate that may interact with RNF6. Coimmunoprecipitation experiments in SW480 and HCT116 (with RNF6 overexpression) cells confirmed that RNF6 binds to TLE3, and reciprocal coimmunoprecipitation further ascertained the interaction between RNF6 and TLE3 using endogenous TLE3 and RNF6 antibodies (Fig. 5B).
RNF6 interacting with TLE3 and inducing TLE3 degradation via polyubiquitination. A, Anti-RNF6 and IgG pulled-down proteins were stained by silver staining. Compared with IgG group, proteins enriched by anti-RNF6 were identified by mass spectrometry (identified proteins are indicated by arrows). B, The interaction between RNF6 and TLE3 was confirmed by coimmunoprecipitation (IP) in SW480 cells and RNF6 stably transfected HCT116 cells. C, RNF6 decreased TLE3 expression but enhanced nuclear β-catenin protein expression. Knockdown of RNF6 yields an opposite effect on these two proteins. Lamin A/C is a nuclear marker, and β-tubulin is a cytoplasmic marker. D, Ectopic expression of RNF6 did not change the mRNA levels of both TLE3 and β-catenin. β-Actin was used as an internal control. **, P < 0.01. E, Treatment with MG132 (10 μmol/L for 12 hours), an inhibitor of the ubiquitin proteasome system, abolished the effect of RNF6 on the degradation of TLE3. F, RNF6-overexpressing HT29 and HCT116 cells or RNF6-silenced SW480 cells were transfected with HA-ubiquitin plasmid for 48 hours. MG132 was added to the cells 6 hours before they were harvested. The cell lysates were pulled down with HA-ubiquitin antibody and immunoblotted with TLE3 antibody. G and H, The interaction between TLE3, β-catenin, and TCF4 was revealed by immunoprecipitation assay. G, Dissociation of β-catenin was found in RNF6-silencing cells caused by TLE3 displacement. H, Depletion of RNF6 simultaneously increased the binding of TLE3 to TCF4 but suppressed the interaction between β-catenin and TCF4.
RNF6 interacting with TLE3 and inducing TLE3 degradation via polyubiquitination. A, Anti-RNF6 and IgG pulled-down proteins were stained by silver staining. Compared with IgG group, proteins enriched by anti-RNF6 were identified by mass spectrometry (identified proteins are indicated by arrows). B, The interaction between RNF6 and TLE3 was confirmed by coimmunoprecipitation (IP) in SW480 cells and RNF6 stably transfected HCT116 cells. C, RNF6 decreased TLE3 expression but enhanced nuclear β-catenin protein expression. Knockdown of RNF6 yields an opposite effect on these two proteins. Lamin A/C is a nuclear marker, and β-tubulin is a cytoplasmic marker. D, Ectopic expression of RNF6 did not change the mRNA levels of both TLE3 and β-catenin. β-Actin was used as an internal control. **, P < 0.01. E, Treatment with MG132 (10 μmol/L for 12 hours), an inhibitor of the ubiquitin proteasome system, abolished the effect of RNF6 on the degradation of TLE3. F, RNF6-overexpressing HT29 and HCT116 cells or RNF6-silenced SW480 cells were transfected with HA-ubiquitin plasmid for 48 hours. MG132 was added to the cells 6 hours before they were harvested. The cell lysates were pulled down with HA-ubiquitin antibody and immunoblotted with TLE3 antibody. G and H, The interaction between TLE3, β-catenin, and TCF4 was revealed by immunoprecipitation assay. G, Dissociation of β-catenin was found in RNF6-silencing cells caused by TLE3 displacement. H, Depletion of RNF6 simultaneously increased the binding of TLE3 to TCF4 but suppressed the interaction between β-catenin and TCF4.
In light of the interaction between RNF6 and TLE3, we tested whether RNF6 could alter TLE3 expression. RNF6 exhibited a predominantly nuclear localization (Fig. 5C). Nuclear TLE3 protein levels were significantly suppressed in RNF6-transfected HT29 and HCT116 cells, whereas depletion of RNF6 in DLD-1 and SW480 cells stabilized TLE3 levels in the nucleus (Fig. 5C). In either case, the mRNA level of TLE3 was not altered (Fig. 5D), indicating that RNF6 negatively regulates TLE3 at posttranscriptional level. Given that RNF6 is an E3 ubiquitin ligase, we hypothesized that RNF6 may function to promote TLE3 ubiquitination and degradation via the ubiquitin–proteasome system (UPS). In line with this possibility, TLE3 expression was restored after treatment with the proteasome inhibitor MG132, suggesting that RNF6-induced TLE3 degradation depends on the UPS (Fig. 5E). To confirm the role of RNF6 in UPS-mediated TLE3 degradation, we assessed TLE3 ubiquitin level. Consistent with our hypothesis, ectopic expression of RNF6 did indeed increase the ubiquitination of TLE3, whilst knockdown of RNF6 decreased the polyubiquitination of TLE3 (Fig. 5F). Taken together, these results indicate that RNF6 functions as an E3 ubiquitin ligase responsible for TLE3 degradation via the ubiquitin proteasome pathway in colorectal cancer cells.
RNF6 promotes binding of β-catenin to the TCF4/LEF complex through TLE3 degradation
It has been previously established that TLE family transcriptional repressors (TLE 1–3) binds to the TCF4/LEF complex with overlapping binding sites with β-catenin (26, 27). Given that RNF6 degrades TLE3, we next investigated how RNF6 might modulate the competition between TLE3 and β-catenin for binding to the TCF4/LEF complex. Congruently, dissociation of β-catenin from TCF4/LEF was found in RNF6-silenced cells with TLE3 enrichment (Fig. 5G). To determine the impact of RNF6 on the binding affinity of β-catenin to the TCF4/LEF complex, we pulled down TCF4 in control and RNF6-silenced SW480 cells. Knockdown of RNF6 significantly increased the binding between TLE3 and TCF4, but with a corresponding decrease in β-catenin/TCF4 interaction (Fig. 5H). RNF6-promoted degradation of TLE3 is thus required for recruitment of β-catenin to TCF4/LEF and stabilization of the β-catenin/TCF4/LEF complex. This connection was reinforced by the observation that nuclear β-catenin level was accumulated when RNF6 was overexpressed, as well as by the reduced nuclear β-catenin in RNF6 knockdown cells (Fig. 5C).
RNF6-mediated TLE3 degradation activates the Wnt signaling pathway
As RNF6-induced TLE3 degradation promotes binding of β-catenin to the TCF4/LEF complex, we speculated that RNF6 regulates Wnt/β-catenin signaling. Congruently, RNF6 accelerated TLE3 turnover when treated with the translational inhibitor cycloheximide (100 μg/mL), whereas β-catenin turnover was reduced, implying that RNF6 helps to stabilize β-catenin mainly by promoting TLE3 degradation (Fig. 6A). Hence, we determined Wnt/β-catenin pathway activity by luciferase reporter assays. As expected, RNF6 overexpression in HCT116 and HT29 cells enhanced activity of Wnt signaling, as demonstrated by TOPflash luciferase reporter assay, whereas knockdown of RNF6 in DLD-1 and SW480 cells dramatically abolished TOPflash activity (Fig. 6B). Consequently, we determined the expression of key downstream Wnt effectors associated with cell proliferation and EMT to illuminate the role of RNF6 in Wnt signaling activation. Consistent with the positive effect of RNF6 on Wnt signaling, Western blot analysis revealed that overexpression of RNF6 upregulated the levels of the Wnt effectors cyclin D1, C-myc, β-catenin, and active β-catenin (28, 29), whereas RNF6 knockdown diminished the expression of these key markers (Fig. 6C). To corroborate our in vitro data, we analyzed TCGA RNA sequencing results of colorectal cancer patients by gene-set enrichment analysis (GSEA). Among the 189 oncogenic signature gene sets, RNF6 was most strongly associated with Wnt/β-catenin, MYC, VEGF, and cell-cycle signaling pathways, whereas it was negatively correlated with apoptosis and cell adhesion signatures (Fig. 6D). RNF6 is therefore essential for Wnt signaling activation and its consequent cascades in colorectal cancer. Owing to its ability to recruit β-catenin to TCF4/LEF, RNF6 facilitates its transcriptional activity, as evidenced by increased the expression of β-catenin/TCF4 target genes, including c-Jun, Axin-2, matrix metalloproteinase-7 (MMP-7), CD44, and VEGF (Fig. 6E; refs. 30–34). Knockdown of RNF6 diminished the expression of β-catenin/TCF4 downstream targets that correlate with cell cycle and EMT process, which is in concordance with the results of the TOPflash assay. Thus, degradation of TLE3 by RNF6 is essential for activation and stabilization of β-catenin to facilitate downstream cell proliferation and EMT-related gene transcription to promote tumor growth and metastasis. In addition, mRNA expression of SNAI1/2, ZEB1, and TWIST1/2 was increased in HCT116 cells overexpressing RNF6, whereas silencing of RNF6 in SW480 decreased mRNA expression of SNAI1/2 and ZEB1 (TWIST1/2 was not detected; Fig. 6E).
RNF6 activates Wnt/β-catenin signaling pathway through TLE3 degradation. A, RNF6 stably transfected HT29 and HCT116 cells were treated with cycloheximide (CHX; 100 μg/mL) for indicated times. The turnover of TLE3 was increased, whereas β-catenin was stabilized compared with control. GAPDH was used as a loading control. B, RNF6 promoted the Wnt signaling pathway in HT29 and HCT116 cells, as determined by TOPflash luciferase reporter assay. RNF6 knockdown suppressed the Wnt signaling pathway in DLD-1 and SW480 cells. TOPflash luciferase reporter with TCF binding site mutation was used as a negative control. C, HT29 and HCT116 cells were stably transfected with pcDNA3.1-RNF6 and pcDNA3.1 empty vector. The key Wnt pathway–related markers were analyzed by Western blot analysis. DLD-1 and SW480 cells were knocked down by shRNF6 and control shRNA. Western blot analysis was performed to measure the key Wnt pathway–related markers. D, Gene-set enrichment analysis (GSEA) of RNF6 in 329 colorectal cancer patients from TCGA. Representative GSEA plots indicated that Wnt/β-catenin, MYC, VEGF and cell-cycle pathways are positively associated with RNF6 among the 189 oncogenic signature gene sets, with the significance defined by false discovery rate (FDR) q < 0.01. The apoptosis and cell adhesion signatures are negatively associated with RNF6 in the hallmark or Kyoto Encyclopedia of Genes and Genomes gene sets analysis, with the significance defined by FDR q < 0.05. NES, normalized enrichment score. E, HCT116 cells were stably transfected with RNF6 or control vector, and SW480 cells were treated with shRNF6 or control shRNA. qRT-PCR was performed to measure the Wnt/β-catenin pathway downstream target genes and EMT-associated transcriptional factors in the indicated colorectal cancer cell lines. β-Actin was used as an internal control. Results are presented as mean ± SD of three independent experiments. F, Upregulation of active β-catenin mediated by RNF6 was attenuated upon restoration of TLE3 in HT29 and HCT116 cells. G, HCT116 cells were stably transfected with pcDNA3.1-RNF6 or pcDNA3.1 empty vector. Knockdown of TLE3 abolished the active effect of RNF6 on Wnt pathway activity, as measured by luciferase reporter assay. Silencing TLE3 by siTLE3 profoundly restored Wnt pathway activity suppressed by RNF6 depletion. H, Restoring TLE3 expression impaired the growth-promoting effect caused by RNF6, as determined by cell viability and colony formation assay in HCT116 cells. Knockdown of TLE3 in SW480 cells weakened RNF6, whose loss caused tumor attenuation. *, P < 0.05; **, P < 0.01; ***, P < 0.001. Results are presented as mean ± SD of three independent experiments. NS, not significant.
RNF6 activates Wnt/β-catenin signaling pathway through TLE3 degradation. A, RNF6 stably transfected HT29 and HCT116 cells were treated with cycloheximide (CHX; 100 μg/mL) for indicated times. The turnover of TLE3 was increased, whereas β-catenin was stabilized compared with control. GAPDH was used as a loading control. B, RNF6 promoted the Wnt signaling pathway in HT29 and HCT116 cells, as determined by TOPflash luciferase reporter assay. RNF6 knockdown suppressed the Wnt signaling pathway in DLD-1 and SW480 cells. TOPflash luciferase reporter with TCF binding site mutation was used as a negative control. C, HT29 and HCT116 cells were stably transfected with pcDNA3.1-RNF6 and pcDNA3.1 empty vector. The key Wnt pathway–related markers were analyzed by Western blot analysis. DLD-1 and SW480 cells were knocked down by shRNF6 and control shRNA. Western blot analysis was performed to measure the key Wnt pathway–related markers. D, Gene-set enrichment analysis (GSEA) of RNF6 in 329 colorectal cancer patients from TCGA. Representative GSEA plots indicated that Wnt/β-catenin, MYC, VEGF and cell-cycle pathways are positively associated with RNF6 among the 189 oncogenic signature gene sets, with the significance defined by false discovery rate (FDR) q < 0.01. The apoptosis and cell adhesion signatures are negatively associated with RNF6 in the hallmark or Kyoto Encyclopedia of Genes and Genomes gene sets analysis, with the significance defined by FDR q < 0.05. NES, normalized enrichment score. E, HCT116 cells were stably transfected with RNF6 or control vector, and SW480 cells were treated with shRNF6 or control shRNA. qRT-PCR was performed to measure the Wnt/β-catenin pathway downstream target genes and EMT-associated transcriptional factors in the indicated colorectal cancer cell lines. β-Actin was used as an internal control. Results are presented as mean ± SD of three independent experiments. F, Upregulation of active β-catenin mediated by RNF6 was attenuated upon restoration of TLE3 in HT29 and HCT116 cells. G, HCT116 cells were stably transfected with pcDNA3.1-RNF6 or pcDNA3.1 empty vector. Knockdown of TLE3 abolished the active effect of RNF6 on Wnt pathway activity, as measured by luciferase reporter assay. Silencing TLE3 by siTLE3 profoundly restored Wnt pathway activity suppressed by RNF6 depletion. H, Restoring TLE3 expression impaired the growth-promoting effect caused by RNF6, as determined by cell viability and colony formation assay in HCT116 cells. Knockdown of TLE3 in SW480 cells weakened RNF6, whose loss caused tumor attenuation. *, P < 0.05; **, P < 0.01; ***, P < 0.001. Results are presented as mean ± SD of three independent experiments. NS, not significant.
Oncogenic effect of RNF6 is dependent on TLE3 degradation
Next, we asked whether RNF6 regulates Wnt/β-catenin signaling through directly modulating TLE3 levels. Restoration of TLE3 attenuated the upregulation of active β-catenin mediated by RNF6 (Fig. 6F). Likewise, knockdown of TLE3 abolished the active effect of RNF6 on the Wnt signaling pathway, as determined by TOPflash activity (Fig. 6G). As a further validation of our findings, silencing of TLE3 restored TOPflash activity caused by RNF6 depletion in SW480 cells (Fig. 6G). Notably, restoration of TLE3 abrogated the oncogenic effect of RNF6 in HCT116 cells, as determined by cell growth and colony formation assays. On the contrary, TLE3 knockdown rescued cell proliferation in SW480 cells with RNF6 loss (Fig. 6H). These results indicate that RNF6 exerts its oncogenic function via mediating TLE3 degradation. Collectively, our data demonstrated that RNF6 mediates its oncogenic effect by activating the Wnt signaling pathway, and this effect is partially dependent on TLE3 degradation.
Discussion
In this study, we identified RNF6 as a novel oncogene that is frequently upregulated by gene amplification in primary colorectal cancer (73.5%) in two independent cohorts. RNF6 is located on chromosome 13q12.13, a genomic region with a higher frequency of amplification in colorectal cancer than in any other cancer type (7, 35). Noticeably, RNF6 amplification occurs in the early stage of colorectal cancer, and patients with early stage colorectal cancer diagnosed with RNF6 copy number amplification manifested a higher risk of recurrence. RNF6 amplification rate further increased in the late tumor stages, which may explain why RNF6 amplification is lower in our cohort (33.1% vs. 66.3%) compared with the TCGA colorectal cancer cohort, since our Beijing cohort 1 consisted of stage II and III colorectal cancer, without any stage IV patients. RNF6 amplification was positively associated with its mRNA expression in colorectal cancer from our cohort and the TCGA dataset, suggesting that gene amplification drives the overexpression RNF6, and RNF6 has a critical role in triggering colorectal cancer initiation and progression.
A series of in vitro and in vivo experiments established RNF6 as an oncogenic factor in colorectal cancer. RNF6 overexpression in colon cancer cells promoted cell proliferation in vitro and tumorigenicity in a xenograft model in mice. RNF6-overexpressing xenograft tumors had elevated cyclin D1 and active-β-catenin expression and a faster growth rate (Fig. 3E), supporting the relationships between RNF6 and Wnt activation in vivo. The oncogenic effect of RNF6 was mediated through accelerating cell-cycle progression and inhibition of apoptosis, involving the inhibition of both intrinsic and extrinsic apoptosis pathways (36). Tumor metastases are the cause of 90% of cancer-related deaths (37). Here, we provided first evidence that RNF6 promoted cell migration and invasion in colorectal cancer cells in vitro and lung metastasis in mice. RNF6 exerted its pro-metastatic effect by promoting EMT, as indicated by loss of the epithelial makers E-cadherin and Claudin-1, and increased expression of the mesenchymal markers N-cadherin, vimentin, SNAI1/2, TWIST1/2, and ZEB1/2, thereby favoring a mesenchymal phenotype that enables increased cell migration and invasion (24, 25, 38, 39). These findings are further verified in TCGA colorectal cancer transcriptome sequencing results, that is, RNF6 positively correlated with cell-cycle signature, whereas it was negatively correlated with apoptosis and cell adhesion signatures. The loss of intercellular adhesion makes it easy for tumor cells to migrate and invade, eventually leading to metastatic dissemination (40). Collectively, RNF6 functions as a versatile oncogene in colorectal cancer by promoting tumor growth and metastasis. RNF6 has also been shown to be a contextual oncogene or tumor suppressor gene in other cancer types, such as prostate cancer (14), leukemia (41), and esophageal squamous cell carcinoma (42, 43). In prostate cancer, RNF6 promoted androgen-independent cancer cell growth via ubiquitination of the androgen receptor (AR), which facilitates the recruitment of coactivators to induce AR pathway downstream gene transcription (14). However, RNF6 was believed to be a tumor suppressor in esophageal squamous cell carcinoma because of its chromosomal location and multiple somatic mutations (42, 43). Hence, the function of RNF6 is likely to be tissue- and cancer-type dependent. The unique amplification of RNF6 in patients with colorectal cancer, together with the compelling oncogenic effect of RNF6 in colorectal cancer cells and mouse models, suggests that RNF6 is particularly important for this specific cancer type.
RNF6 belongs to the RING finger E3 ligase family. Emerging evidence indicates that RING finger E3 ligases participate in numerous cellular processes primarily through facilitating the ubiquitin-dependent degradation of their target proteins (44). Unbiased screening of RNF6-binding proteins in colorectal cancer cells using immunoprecipitation–mass spectrometry unveiled TLE3 as a novel RNF6 interacting partner. Consistent with its function as an E3 ligase, we demonstrated that RNF6 mediated the polyubiquitination of TLE3 and its subsequent degradation in the proteasome in colorectal cancer cells. Strikingly, TLE3 is a powerful repressor of the β-catenin/TCF4 complex, and it negatively regulates Wnt/β-catenin signaling pathway (26, 27). Indeed, RNF6-mediated TLE3 degradation significantly suppressed the association of TLE3 with TCF4/LEF, which in turn, led to recruitment of β-catenin to TCF4/LEF, enhanced Wnt/β-catenin transcriptional activity, and the expression of its downstream factors associated with cell proliferation and EMT (depicted in Fig. 7). Concomitant with RNF6 expression, the clinical colorectal cancers enriched in the Wnt/β-catenin or Wnt-correlated pathways such as MYC and VEGF signatures by the GSEA analysis further supported our findings (29, 34). Furthermore, TLE3 gain- and loss-of-function experiments demonstrated that TLE3 degradation is essential for the oncogenic effect of RNF6. Thus, RNF6/TLE3/β-catenin form a functional axis that exerts progrowth and prometastatic behaviors in colorectal cancer progression.
Proposed mechanistic scheme of RNF6 in promoting the Wnt signaling pathway in colorectal cancer. RNF6 copy number amplification induces high expression of RNF6, and its amplification increased with colorectal cancer progression. RNF6, an E3 ubiquitin ligase, promotes TLE3 ubiquitination and proteasomal degradation, which in turn releases TLE3 from the TCF/LEF complex, thereby increasing recruitment of β-catenin to the TCF4/LEF complex to active Wnt/β-catenin pathway to promote colorectal tumorigenesis.
Proposed mechanistic scheme of RNF6 in promoting the Wnt signaling pathway in colorectal cancer. RNF6 copy number amplification induces high expression of RNF6, and its amplification increased with colorectal cancer progression. RNF6, an E3 ubiquitin ligase, promotes TLE3 ubiquitination and proteasomal degradation, which in turn releases TLE3 from the TCF/LEF complex, thereby increasing recruitment of β-catenin to the TCF4/LEF complex to active Wnt/β-catenin pathway to promote colorectal tumorigenesis.
Loss-of-function mutations of APC or gain-of-function mutations in β-catenin are well-known carcinogenic mechanisms to aberrantly activate canonical Wnt/β-catenin signaling in colorectal cancer (4, 11, 45). APC mutation is involved in aberrant crypts and early adenoma formation, and causes chromosomal instability (46). Concurrence with APC mutation and RNF6 copy number amplification is enhanced in patients with colorectal cancer patients. Mutations in the APC tumor suppressor have been shown to cause chromosomal instability by inducing spindle abnormalities (46). Mutant APC induces chromosome gain or loss randomly; however, it is reasonable to assume that RNF6 becomes specifically amplified through positive selection during tumorigenesis. Given the essentiality of Wnt/β-catenin activation in colorectal cancer development, the participation of RNF6 as a driver oncogene in this pivotal cascade is of particular significance. RNF6-mediated degradation of the Wnt suppressor TLE3 may therefore function as an enhancer to exacerbate Wnt/β-catenin signaling and drive colorectal carcinogenesis. Our results highlighted an underappreciated role of RNF6 gene amplification, which causes aberrant expression of this E3 ligase that specifically targets TLE3 for degradation, which in turn, promotes colorectal cancer development.
Our findings were further strengthened by the clinical impact of RNF6 amplification and overexpression in the survival of patients with colorectal cancer. Consistent with its oncogenic role, RNF6 mRNA expression was an independent prognostic factor that predicts poor survival of patients with colorectal cancer in two independent cohorts. Moreover, colorectal cancer patients with RNF6 copy number amplification had a higher risk of recurrence.
In summary, we demonstrate that RNF6 overexpression due to gene amplification is a common event in colorectal cancer. RNF6 plays a pivotal oncogenic role through TLE3 ubiquitin-mediated degradation, consequently activating Wnt/β-catenin pathway in colorectal carcinogenesis. Therapeutic intervention of RNF6 E3 ligase activity might be a novel strategy for blunting uncontrolled growth and metastasis in Wnt/β-catenin–addicted colorectal cancer. Moreover, RNF6 may serve as an independent prognosis marker for patients with colorectal cancer.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Authors' Contributions
Conception and design: L. Liu, J. Yu
Development of methodology: L. Liu, Y. Zhang, J. Zhang, J. Yu
Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Liu, Y. Zhang, Y. Dong, W. Kang, J. Yu
Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Liu, X. Li
Writing, review, and/or revision of the manuscript: L. Liu, Y. Zhang, C.C. Wong, J. Yu
Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L. Liu, F.K.L. Chan, J.J.Y. Sung, J. Yu
Study supervision: J. Yu
Acknowledgments
The authors would like to thank Professor Wei Jia (University of Hawaii Cancer Center) and Professor Xiaoyong Yang (Comparative Medicine and Cellular & Molecular Physiology, Yale University), who helped discuss the project and gave helpful suggestions.
This project was supported by RGC-GRF Hong Kong (14163817, 14106145, 14111216); HMRF Hong Kong (03140856); 973 Program China (2013CB531401); The National Key Technology R&D Program (2014BAI09B05); Science and Technology Program Grant, Shenzhen (JCYJ20170413161534162); and Shenzhen Virtual University Park Support Scheme to CUHK Shenzhen Research Institute and CUHK direct grant (to J. Yu).
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.