Skip to main content

Selenium-binding protein 1 transcriptionally activates p21 expression via p53-independent mechanism and its frequent reduction associates with poor prognosis in bladder cancer

Abstract

Background

Recent studies have shown that selenium-binding protein 1 (SELENBP1) is significantly down-regulated in a variety of solid tumors. Nevertheless, the clinical relevance of SELENBP1 in human bladder cancer has not been described in any detail, and the molecular mechanism underlying its inhibitory role in cancer cell growth is largely unknown.

Methods

SELENBP1 expression levels in tumor tissues and adjacent normal tissues were evaluated using immunoblotting assay. The association of SELENBP1 expression, clinicopathological features, and clinical outcome was determined using publicly available dataset from The Cancer Genome Atlas bladder cancer (TCGA-BLCA) cohort. DNA methylation in SELENBP1 gene was assessed using online MEXPRESS tool. We generated stable SELENBP1-overexpression and their corresponding control cell lines to determine its potential effect on cell cycle and transcriptional activity of p21 by using flow cytometry and luciferase reporter assay, respectively. The dominant-negative mutant constructs, TAM67 and STAT1 Y701F, were employed to define the roles of c-Jun and STAT1 in the regulation of p21 protein.

Results

Here, we report that the reduction of SELENBP1 is a frequent event and significantly correlates with tumor progression as well as unfavorable prognosis in human bladder cancer. By utilizing TCGA-BLCA cohort, DNA hypermethylation, especially in gene body, is shown to be likely to account for the reduction of SELENBP1 expression. However, an apparent paradox is observed in its 3′-UTR region, in which DNA methylation is positively related to SELENBP1 expression. More importantly, we verify the growth inhibitory role for SELENBP1 in human bladder cancer, and further report a novel function for SELENBP1 in transcriptionally modulating p21 expression through a p53-independent mechanism. Instead, ectopic expression of SELENBP1 pronouncedly attenuates the phosphorylation of c-Jun and STAT1, both of which are indispensable for SELENBP1-mediated transcriptional induction of p21, thereby resulting in the G0/G1 phase cell cycle arrest in bladder cancer cell.

Conclusions

Taken together, our findings provide clinical and molecular insights into improved understanding of the tumor suppressive role for SELENBP1 in human bladder cancer, suggesting that SELENBP1 could potentially be utilized as a prognostic biomarker as well as a therapeutic target in future cancer therapy.

Background

Bladder cancer is one of the most common urological malignancies, and is the leading cause of deaths in patients with urinary tract disease. The incidence of bladder cancer has steadily increased worldwide in recent decades [1]. According to the report from American Cancer Society in 2018, there are an estimated 81,190 of new cases diagnosed with bladder cancer and approximately 17,240 of bladder cancer-related deaths in the United States [2]. For non-muscle-invasive and muscle-invasive bladder cancer, grade and depth of tumor invasion are the most important prognostic factors, respectively [3]; the 5-year overall survival rate is approximately 8.1% among patients with distant metastasis [4]. Therefore, the identification of novel therapeutic targets or predictive biomarkers, and in-depth elucidation of the underlying molecular mechanisms are of tremendous importance for reducing the mortality of this disease.

The human selenium-binding protein 1 (SELENBP1) gene, located on chromosome 1 at q21-22, encodes a highly-conserved member of the selenium-binding protein family [5], and is the homologue of the mouse SP56 gene that has been originally reported as a 56 kDa mouse protein with the ability to bind 75selenium stably [6, 7]. The human SELENBP1 protein is ubiquitously expressed in various tissue types, especially higher in heart, lung, liver and kidney [5]. Since the initial report identifying SELENBP1 as a tumor-associated protein in prostate cancer [8], the reduced expression or even lost of SELENBP1 has been consistently observed in a variety of solid tumors as compared to corresponding normal tissues, including those of the skin [9], lung [10], esophagus [11], stomach [12,13,14], colon [15, 16], liver [17], breast [18] and ovary [19]. Thereafter, accumulating evidence has convincingly demonstrated that reduced expression of SELENBP1 is an independent predictive of poor clinical outcome in multiple malignant diseases [13,14,15,16,17,18, 20,21,22]. Furthermore, an increasing number of in vitro and in vivo studies has also consistently shown that increasing the levels of SELENBP1 significantly suppresses the malignant characteristics of cancer cells, leading scientific community to the consensus that SELENBP1 may act as a putative tumor suppressor involved in the regulation of cell proliferation, senescence, epithelial–mesenchymal transition, migration and apoptosis [23]. However, the clinical significance of SELENBP1 in human bladder cancer has not yet been characterized in any detail. Additionally, the molecular mechanisms underlying the tumor-suppressive role for SELENBP1 in cancer cells are still largely undefined.

Here we show that SELENBP1 is significantly down-regulated in human bladder cancer tissues and cell lines, and its frequent reduction is further associated with tumor progression as well as poor clinical outcome among patients with bladder cancer. In addition, the reduced expression of SELENBP1 is inversely associated with DNA hypermethylation in its promoter region, but more significantly correlated negatively with DNA methylation in gene body. Importantly, an apparent paradox is observed in 3′-UTR region, in which DNA methylation is positively related to SELENBP1 expression. Furthermore, we verify the growth inhibitory role for SELENBP1 in human bladder cancer, and report a novel function for SELENBP1 in transcriptionally modulating p21 expression through a p53-independent mechanism. Instead, ectopic expression of SELENBP1 pronouncedly attenuates the phosphorylation of c-Jun and STAT1, both of which are indispensable for SELENBP1-mediated transcriptional induction of p21, thereby resulting in the G0/G1 phase cell cycle arrest in bladder cancer cell.

Methods and materials

Cell lines, cell culture and plasmids

Human bladder cancer cell lines (UMUC3, RT4, RT112, TCCSUP, J82 and 5637) used in this study were described previously [24, 25]. Human bladder cancer T24 and its metastatic derivative T24T cell lines [26], and human colon cancer wild-type HCT116 (HCT116 WT), p21 knockout (HCT116 p21−/−) and p53 knockout (HCT116 p53−/−) cell lines [27] were described in our prior studies, and were cultured in corresponding medium supplemented with 10% heat-inactivated fetal bovine serum, 2 μM l-glutamine and 25 μg/mL gentamycin at 37 °C in a humidified atmosphere of 5% CO2. As previously described [28, 29], UMUC3 and T24T cell lines were authenticated by Genetica DNA Laboratories (Burlington, NC, USA). The constructs encoding human HA-tagged SELENBP1 (HA-SELENBP1) [30] or dominant-negative form of c-Jun (TAM67) [31] were previously described. The plasmid encoding dominant-negative STAT1 (DN-STAT1 Y701F) and its control empty vector (pEGFP-C1) were gifts from Alan Perantoni (Addgene plasmid #12302) [32]. Full-length (2.4 Kb) and truncated-length (1.8 Kb, 1.3 Kb and 200 bp) of p21 promoter-driven luciferase reporter plasmids were generously provided by Dr. Jennifer A. Pietenpol at Vanderbilt Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine (Nashville, TN) [33].

Human bladder cancer tissue specimens

Twelve pairs of primary invasive bladder cancer specimens and their paired adjacent normal bladder tissues were obtained from patients who received radical cystectomy from 2012 to 2013 at the Department of Urology of the Union Hospital of Tongji Medical College (Wuhan, China) [34]. After surgical procedure, all of specimens were immediately collected and stored in liquid nitrogen, and the pathological diagnoses were further confirmed by a certified clinical pathologist according to the 2004 World Health Organization Consensus Classification and Staging System for bladder neoplasms [35]. Before collecting these specimens, ethical approval has been obtained from the Medical Ethics Committee of China, and all patients have given their informed consent for translational research.

Transfection and luciferase reporter assay

All of stable or transient transfections were performed using PolyJet™ DNA in vitro transfection reagent (SignaGen Laboratories, Rockville, MD) according to manufacturer’s instructions. Stable transfectants were selected with puromycin, hygromycin B or G418 for 3–4 weeks [26, 27], and surviving cells from the antibiotics selection were pooled as stable mass transfectants, with at least two passages prior to utilization for further experiments. For the determination of p21 promoter-mediated luciferase activity, cells were transfected with the related luciferase reporter in combination with the pRL-TK vector (Promega, Madison, WI). After 24 h of transfection, luciferase activity was determined using a dual-luciferase reporter assay system kit (Promega, Madison, WI) as described previously [36, 37]. The results were normalized by internal TK signal, and expressed as mean ± standard deviation from at least three independent experiments.

Immunoblotting assay and antibodies

Whole-cell extracts were prepared with cell lysis buffer (10 mM Tris–HCl (pH 7.4), 1% SDS, and 1 mM Na3VO4), and were then subjected to immunoblotting analysis as described in our previous studies [24, 27]. Briefly, equal aliquots of cell extracts were resolved by SDS-PAGE, transferred to PVDF membranes (Bio-Rad, Hercules, CA) and probed with primary antibodies against one of the following proteins: SELENBP1 (Medical and Biological Laboratories, Japan), p21, p27, Cyclin D1, CDK4, CDK6, p53, Sp1, JunB, Jun D, c-Jun, phospho-c-Jun at S73, STAT1/3/5/6, phospho-STAT1 at Y701, phospho-STAT3 at Y705, phospho-STAT5 at Y694, phospho-STAT6 at Y641, SOCS1, GAPDH and HA-tag (Cell Signaling Technology, Beverly, MA). Primary-antibody-bound proteins were further incubated with an alkaline-phosphatase-conjugated secondary antibody and detected using an ECF immunoblotting system (Amersham, Piscataway, NJ). Images were acquired using Typhoon FLA 7000 (GE Healthcare, Pittsburgh, PA). The results shown are representative of three independent experiments. The density of bands was quantified relative to that of loading control by using Quantity One software (Life Science, Hercules, CA) [31].

RT-PCR

Total RNA was extracted using TRIzol reagent (Invitrogen, Grand Island, NY) according to the manufacturer’s instructions. Total RNA (5.0 μg) was used for first-strand cDNA synthesis with oligo (dT) 20 primer by SuperScript First-Strand Synthesis system (Invitrogen). The mRNA levels were determined by semi-quantitative reverse transcription (RT)-PCR, as described previously [38]. These specific primers used in this study were as follows: human p21 (forward, 5′-CCC AGT GGA CAG CGA GCA GC-3′; reverse, 5′-ACT GCA GGC TTC CTG TGG GC-3′), and human GAPDH (forward, 5′-AGA AGG CTG GGG CTC ATT TG-3′; reverse, 5′-AGG GGC CAT CCA CAG TCT TC-3′).

Anchorage-independent growth assay

Anchorage-independent growth assay in soft agar (soft agar assay) was performed as described in our previous study [31, 39]. Briefly, 1 × 104 of indicated cells were re-suspended in 1 mL of 10% FBS basal modified Eagle’s medium (BME) containing 0.33% soft agar and seeded on top of 0.5% agar in 10% FBS BME in each well of 6-well plates. The plates were maintained in a 5% CO2 incubator at 37 °C for 2–3 weeks. Colonies with more than 32 cells were scored and presented as mean ± standard deviation from at least three independent experiments.

Cell-cycle analysis

As previously described [27], the indicated cells were harvested and fixed in 75% ethanol overnight, and were suspended in the staining buffer containing 0.1% Triton X-100, 0.2 mg/mL RNase A, and 50 mg/mL propidium iodide (PI) at 4 °C. DNA content was then determined by a flow cytometry Epics XL flow cytometer (Beckman Coulter Inc., Miami, FL) and the results were analyzed with EXPO32 (v1.2) software.

Bioinformatics analysis of data mining in TCGA-bladder cancer (TCGA-BLCA) cohort

The TCGA-BLCA clinical data and SELENBP1 expression data were obtained from UCSC Xena Browser (https://xenabrowser.net; last updated on April 5th 2019) [40]. UCSC Xena browser shows that TCGA has 406 patients with bladder cancer, in which 19 of patients have SELENBP1 expression data of primary tumor and matched normal bladder tissues. Patients are categorized into low or high SELENBP1 expression group using cut-off value of X-2.33SD (X, mean; SD, standard deviation) calculated from SELENBP1 expression in the matched normal bladder tissues. These two groups were applied for Kaplan–Meier survival analysis (10-year survival). The correlation between SELENBP1 expression and clinicopathological parameters in TCGA-BLCA were also examined. Besides, we analyzed the association between SELENBP1 expression and DNA methylation using the MEXPRESS tool (http://mexpress.be/), a user-friendly tool for the visualization and interpretation of DNA methylation in TCGA data [41].

Statistical analysis

Statistical analysis was performed using SPSS 19.0 software (SPSS, Chicago, IL). Student’s t-test or Fisher’s exact test was used to determine significant differences. p < 0.05 is considered statistically. The results are expressed as the mean ± standard deviation from at least three independent experiments.

Results

SELENBP1 is frequently down-regulated in human bladder cancer, and low expression of SELENBP1 predicts poor clinical prognosis

To determine SELENBP1 expression levels in human bladder cancer, we evaluated the protein expression of SELENBP1 in 12 pairs of bladder cancer tissues and matched adjacent normal tissues using immunoblotting assay. As shown in Fig. 1a, b, SELENBP1 protein levels were significantly down-regulated in 9 out of 12 bladder cancers relative to matched adjacent normal tissues (p = 0.0021). We further examined SELENBP1 mRNA levels in The Cancer Genome Atlas bladder cancer (TCGA-BLCA) cohort, including 406 patients with SELENBP1 expression data, in which 19 of patients have SELENBP1 expression data of primary tumor and matched normal bladder tissues. Consistently, reduction of SELENBP1 expression was also frequently observed in a majority of tumor tissues from TCGA-BLCA cohort (p < 0.0001; Fig. 1c). Utilizing the gene expression data in matched normal tissues, we created two groups based on SELENBP1 gene expression and analyzed overall survival (OS) curves of differential SELENBP1 expression levels. Log-rank test of the OS curves in TCGA-BLCA cohort indicated that bladder cancer patients with low SELENBP1 expression were remarkably related to worse prognosis than those with high SELENBP1 expression (Fig. 1d), implying that SELENBP1 might serve as a tumor suppressor in bladder cancer.

Fig. 1
figure 1

SELENBP1 is frequently down-regulated in human bladder cancer, and low expression of SELENBP1 predicts poor clinical prognosis. a Twelve pairs of bladder cancers (T) and adjacent matched normal tissues (N) were extracted and subjected to immunoblotting assay for determining the levels of SELENBP1 protein expression. GAPDH was used as a loading control. b Relative intensity of SELENBP1 in a was determined using Quantity One software, and normalized to GAPDH. The relative intensity of SELENBP1 in normal tissues was set to 1.0. Paired t test was performed and p value was indicated in top panel. cSELENBP1 mRNA levels were extracted from TCGA bladder cancer (TCGA-BLCA) cohort, including 19 of normal bladder tissues and 406 of bladder cancer tissues. Non-paired t test was performed and p value was indicated in the panel. d Kaplan–Meier 10-year survival analysis among patients with high and low SELENBP1 expression in TCGA-BLCA cohort. The log-rank test was performed and p value was indicated in the panel. Hazard ration (HR) and 95% confidence interval (CI) were also calculated

Low expression of SELENBP1 correlates with unfavorable clinicopathological characteristics of bladder cancer patients

Next, we analyzed the relationship between SELENBP1 expression and clinicopathological features in bladder cancer by using TCGA-BLCA cohort (Table 1), and found that low expression of SELENBP1 was markedly correlated with non-papillary subtype (Papillary: 52% vs. Non-papillary: 64%; p = 0.023), and advanced tumor-node-metastasis (TNM) stage (I/II: 53% vs. III/IV: 64%; p = 0.029), and distant metastasis with marginal statistical significance (p = 0.057), but not with age, gender, lymphovascular invasion (LVI) or other clinicopathological features. These results strongly suggest that the reduced expression of SELENBP1 is associated with the aggressive phenotype of bladder cancer.

Table 1 The correlation of SELENBP1 expression with clinicopathological features in TCGA-BLCA cohort (n = 406)

SELENBP1 expression inversely associates with DNA methylation in the promoter and gene body, but positively correlates with DNA methylation in 3′-UTR region

DNA methylation has been recognized as a major mechanism responsible for epigenetic silencing of SELENBP1 expression in esophageal and colorectal cancer [11, 42]. Therefore, we addressed this association between SELENBP1 expression and DNA methylation in the bladder cancer by using the MEXPRESS tool (http://mexpress.be/), a user-friendly tool for the visualization and interpretation of DNA methylation in TCGA data [41]. As shown in Fig. 2, it was clear that normal samples cluster towards higher SELENBP1 expression than tumor tissues (Wilcoxon rank-sum test, p = 9.01e−7). Moreover, there was a significant inverse association between SELENBP1 expression and DNA methylation in its promoter region located close to the transcription start site (TSS) and the first exon (Pearson correlation coefficients ranging from − 0.339 to − 0.505; p < 0.001). Interestingly, we observed a more significant negative relationship between gene body methylation and SELENBP1 expression with Pearson r = − 0.619 (p < 0.001). However, DNA methylation in its 3′ untranslated region (UTR) was positively associated with SELENBP1 expression (Pearson r = + 0.482; p < 0.001). These observations highlight that those multiple genomic regions of DNA methylation may be differentially involved in modulating SELENBP1 expression.

Fig. 2
figure 2

Visualization of the association between SELENBP1 expression and DNA methylation in TCGA-BLCA cohort using the MEXPRESS tool. SELENBP1 gene together with its transcripts is indicated in the left bottom panel. Next to the gene, blue line plots show the methylation data for the location of each probe (Infinium Human Methylation 450 microarray data; infinium 450 k), whose relative location in SELENBP1 gene is also indicated in the far right panel. The yellow line plot displays the RNA-seq-derived expression data, and the samples are ordered by SELENBP1 expression value. This view shows how SELENBP1 expression and DNA methylation in different parts of SELENBP1 gene are correlated, which is confirmed by the Pearson correlation test. The correlation coefficients (r) are indicated in the right panel. The symbol (***) shows a significant correlation (p < 0.001). TSS, transcription start site; UTR, untranslated region

Overexpression of SELENBP1 inhibits bladder cancer cell growth

To determine the biological function of SELENPB1 in bladder cancer, we firstly employed immunoblotting assay to evaluate SELENBP1 expression in 8 of human bladder cancer cell lines, including T24, T24T, UMUC3, RT4, RT112, TCCSUP, J82 and 5637. As expected, the majority of bladder cancer cell lines exhibited undetectable SELENBP1 protein levels, except TCCSUP and J82 cell lines (Fig. 3a). Then, we established stable transfectants expressing HA-tagged SELENBP1 in UMUC3 cells (Fig. 3b). As shown in trypan blue exclusion assay (Fig. 3c), ectopic expression of SELENBP1 in UMUC3 cells led to a significant decrease in anchorage-dependent growth. Additionally, we also observed that overexpression of SELENBP1 markedly inhibited anchorage-independent growth of UMUC3 cells (Fig. 3d), further verifying the tumor-suppressive role for SELENBP1 in human bladder cancer cellular transformation.

Fig. 3
figure 3

Overexpression of SELENBP1 inhibits bladder cancer cell growth. a Cell lysates of human bladder cancer cell lines were extracted and subjected to immunoblotting assay for determining the levels of SELENBP1 protein expression. GAPDH was used as a loading control. b UMUC3 cells were stably transfected with empty vector (Vector) or construct encoding HA-tagged SELENBP1 (HA-SELENBP1). Cell lysates of these stable transfectants were extracted and subjected to immunoblotting assay by using anti-HA antibody. GAPDH was used as a loading control. c The number of 1 × 104 stable UMUC3 (Vector) and UMUC3 (HA-SELENBP1) cells per well was seeded in 6-well plates. The number of viable cells at the indicated time points was determined by trypan blue exclusion assay. The values were shown as mean ± standard deviation from at least three independent experiments. The asterisk (*) indicates a significant difference in comparison to that of control group (p < 0.05). d Anchorage-independent cell growth of UMUC3 cells stably transfected with empty vector or construct encoding HA-SELENBP1 was determined by soft agar assay. The colony formation was observed and captured under an inverted microscope (×40 magnification; left panel) and numbers of colonies were scored on day 15, and presented as colonies (×100)/104 cells (right panel). The values were shown as mean ± standard deviation from at least three independent experiments. The asterisk (*) indicates a significant difference in comparison to that of control transfectants (p < 0.05)

Induction of p21 protein is crucial for SELENBP1-mediated G0/G1 phase cell cycle arrest

To elucidate the underlying mechanism responsible for its inhibitory role in cancer cell growth, the potential effect of SELENBP1 on cell cycle was determined by flow cytometry analysis. The results showed that overexpression of SELENBP1 induced a significant G0/G1 phase arrest in UMUC3 cells (Fig. 4a), suggesting that G0/G1 phase arrest induction might be associated with tumor suppressive role of SELENBP1. To further elucidate the crucial players that are involved in SELENBP1-mediated G0/G1 phase arrest, we then evaluated the regulatory effect of SELENBP1 on multiple cell cycle-associated proteins, including cyclin-dependent kinase inhibitor 1A (CDKN1A; also known as p21), cyclin-dependent kinase inhibitor 1B (CDKN1B or p27), cyclin D1, cyclin-dependent kinase 4 (CDK4) and CDK6. As shown in Fig. 4b, overexpression of SELENBP1 had no observable regulatory role on p27, Cyclin D1, CDK4 or CDK6, but did result in a dramatic induction of p21 protein in both of UMUC3 and T24T cells. This finding suggests that p21 protein is likely to be a crucial player in SELENBP1-mediated cancer cell growth inhibition. To verify this notion, we established stable transfectants expressing HA-tagged SELENBP1 protein in human colon cancer HCT116 wild-type cells (HCT116 WT) and p21 knockout cells (HCT116 p21−/−), in both of which endogenous protein levels of SELENBP1 were undetectable [30]. As expected, overexpression of SELENBP1 consistently stimulated p21 protein expression and further significantly inhibited anchorage-independent cell growth in HCT116 WT cells (Fig. 4c, d). However, SELENBP1 failed to modulate cancer cell growth in p21-deficiency cells (Fig. 4d), revealing that p21 protein serves as a crucial downstream mediator responsible for the growth-inhibitory role of SELENBP1 in cancer cells. Collectively, these results demonstrate that SELENBP1 specifically up-regulates p21 protein levels and induces G0/G1 phase arrest, thereby leading to attenuation of cancer cell growth.

Fig. 4
figure 4

Induction of p21 protein is crucial for SELENBP1-mediated G0/G1 phase cell cycle arrest. a UMUC3 (Vector) and UMUC3 (HA-SELENBP1) cells were collected at day 4 as described in “Fig. 3c”, and subjected to flow cytometry assay following the staining of propidium iodide. b Cell lysates of stable transfectants UMUC3 (Vector), UMUC3 (HA-SELENBP1), T24T (Vector) and T24T (HA-SELENBP1) were extracted and subjected to immunoblotting assay for determining expression levels of G0/G1 phase arrest-associated regulatory proteins. GAPDH was used as a loading control. Densitometric quantification of p21 (relative to GAPDH) is shown under each blot. c HCT116 WT and HCT116 p21−/− cells were transfected with empty vector or HA-SELENBP1, each along with pGIPZ, and stable transfectants were established under the selection of puromycin. Cell lysates of these stable transfectants were extracted and then subjected to immunoblotting assay with anti-SELENBP1, anti-p21 or anti-GAPDH antibodies. d Anchorage-independent cell growth of stable transfectants described in “c” was determined by soft agar assay. Representative images of colonies were visualized under an inverted microscope (×40 magnification; left panel) and relative numbers of colonies were calculated on day 12 (right panel). The asterisk (*) indicates a significant difference in comparison to that of control transfectants (p < 0.05)

SELENBP1 transcriptionally stimulates p21 expression in a p53-independent manner

To investigate the mechanisms underlying SELENBP1-mediated induction of p21 protein, we first examined p21 mRNA levels, and found that p21 mRNA was markedly up-regulated in both UMUC3 and T24T stable transfectants expressing HA-SELENBP1 (Fig. 5a). To further verify whether SELENBP1-mediated induction of p21 expression occurs at transcriptional level, dual luciferase activities were evaluated in stable UMUC3 (Vector) and UMUC3 (HA-SELENBP1) transfectants using one full-length (2.4 Kb) and three differential truncated-length (1.8 Kb, 1.3 Kb and 200 bp) of human p21 promoter-driven luciferase reporters (Fig. 5b). As indicated in Fig. 5c, overexpression of SELENBP1 significantly led to an approximately 2.3-fold increase in full-length (2.4 Kb), 1.8 Kb- or 1.3 Kb-length of p21 promoter-driven luciferase activities, whereas this regulatory effect was not observed in 200 bp-length reporter assay. These findings reveals that SELENBP1 transcriptionally stimulates p21 expression through the potential responsive elements located within approximately ~ 1300 bp to ~ 200 bp region of the p21 upstream promoter. It has been well established that tumor suppressor p53 is able to binding to the p21 promoter and directly activate its expression [43]. However, we noticed that p53-responsive elements, located in − 1394 bp and − 2285 bp region of p21 promoter [33, 43], are outside of identified SELENBP1-responsive region in p21 promoter (Fig. 5b), leading us to the notion that SELENBP1-mediated p21expression is likely to occur through a p53-independent mechanism. To verify this notion, we re-introduced HA-tagged SELENBP1 in human colon cancer HCT116 p53 knockout cells (HCT116 p53−/−), and found that SELENBP1 still retained the ability to activate p21 expression even in the absence of p53 (Fig. 5d). This result indicates that p53 protein might not be required for SELENBP1-mediated regulation of p21 expression. Collectively, despite the well-established putative role for p53 in the transcriptional regulation of p21 expression, our results demonstrate that SELENBP1 triggers transcriptional induction of p21 expression through a p53-independent pathway.

Fig. 5
figure 5

SELENBP1 transcriptionally stimulates p21 expression in a p53-independent manner. a Total RNA was isolated from indicated stable transfectants, and then subjected to RT-PCR assay for determining p21 mRNA levels. The mRNA levels of GAPDH were used as a loading control. b The potential binding sites of multiple transcription factors in the p21 promoter are shown in upper panel, and full-length (2.4 Kb), 1.8 Kb, 1.3 Kb and 200 bp of p21 promoter-driven luciferase reporters are schematically shown in lower panel. c Stable UMUC3 (Vector) and UMUC3 (HA-SELENBP1) cells were transiently co-transfected with pRL-TK and p21 promoter-driven luciferase reporters as described in “b”. Following 36 h of transient transfection, luciferase reporter activity was determined and normalized to that of corresponding UMUC3-Vector. The asterisk (*) indicates a significant difference in comparison to that of corresponding control transfectants (p < 0.05). d Cell lysates of HCT116 WT and HCT116 p53−/− were subjected to immunoblotting assay for identification of p53 expression (left panel). Following 48 h of transient transfection with HA-SELENBP1 or empty vector, HCT116 p53−/− cells were extracted and then subjected to immunoblotting assay with anti-SELENBP1, anti-p21 or anti-GAPDH antibodies (right panel). Densitometric quantification of p21 (relative to GAPDH) is shown

Overexpression of SELENBP1 results in the suppression of c-Jun and STAT1 phosphorylation

In addition to p53 protein, bioinformatics analysis revealed that p21 promoter region also contains multiple consensus binding sites for other transcriptional factors [44], including signal transductions and activators of transcription (STATs), activating protein-1 (AP-1) and specificity protein 1 (Sp1) (Fig. 5b). To identify whether these transcriptional factors are involved in SELENBP1-mediated transcriptional induction of p21, we used immunoblotting assay to evaluate the levels of transcription factors Sp1 and AP-1 subunits-JunB, JunD and c-Jun, as well as STAT family members, including STAT1, STAT3, STAT5 and STAT6. The results showed that both c-Jun phosphorylation at Ser73 and STAT1 phosphorylation at Tyr701 were significantly down-regulated in UMUC3 cells expressing HA-SELENBP1, whereas other evaluated transcription factors showed no observable changes (Fig. 6a). This result suggests that c-Jun and/or STAT1 may be involved in SELENBP1-mediated transcriptional induction of p21.

Fig. 6
figure 6

The restoration of SELENBP1 leads to the suppression of c-Jun and STAT1 phosphorylation, both of which are required for SELENBP1-mediated transcriptional induction of p21 protein. a Cell lysates of stable UMUC3 (Vector) and UMUC3 (HA-SELENBP1) cells were subjected to immunoblotting assay with indicated antibodies that might be involved in transcriptional regulation of p21 expression. b UMUC3 cells stably transfected with dominant-negative mutant form of c-Jun (TAM67) or pcDNA3.1(Vector) were extracted and then subjected to immunoblotting analysis with indicated antibodies. Densitometric quantification of p21 (relative to GAPDH) is shown. c UMUC3 cells were stably transfected with dominant-negative mutant STAT1 (DN-STAT1) or pEGFP-C1 construct (Vector), and were then extracted for immunoblotting assay with indicated antibodies. Densitometric quantification of p21 and SOCS1 (relative to GAPDH) is shown under each blot. d TAM67 (1 μg) and pcDNA3.1 (1 μg) plasmids were transiently transfected into stable UMUC3 (DN-STAT1) and UMUC3 (pEGFP-C1), respectively. Following 48 h of transient transfection, cells were collected and then subjected to immunoblotting assay. Densitometric quantification of p21 (relative to GAPDH) is shown. e Indicated amounts of empty vector (pcDNA3.1) and TAM67 was transiently co-transfected into stable UMUC3 (DN-STAT1) or UMUC3 (Vector) cells, together with 1.3 Kb of p21 promoter-driven luciferase reporter and pRL-TK, as an internal control. Thirty-six hours post transfection, luciferase reporter activity was determined. Luciferase activities of UMUC3 (Vector) and UMUC3 (DN-STAT1) groups are normalized to those of corresponding control group that transfected with 0.6 μg of pcDNA3.1, respectively. The asterisk (*) indicates a significant difference as compared to UMUC3 (DN-STAT1) group transfected with 0.6 μg of pcDNA3.1 (p < 0.05). The symbol (#) indicates p < 0.05 when compared with UMUC3 (Vector) group transfected with 0.6 μg of pcDNA3.1. The values are shown as mean ± standard deviation from four independent experiments. f Following 48 h of transient transfection with indicated plasmids, UMUC3 cells were collected and then subjected to flow cytometry assay

Combined suppression of c-Jun and STAT1 is required for SELENBP1-mediated transcriptional induction of p21 protein

Next, to define the roles of c-Jun and STAT1 in the regulation of p21 protein, dominant-negative mutants of c-Jun (TAM67) [31] and STAT1 (DN-STAT1 Y701F) [32] were stably transfected into UMUC3 cells, respectively. The results showed that TAM67 efficiently down-regulated c-Jun phosphorylation at Ser73, but inhibited the expression of p21 protein (Fig. 6b), indicating that inhibition of c-Jun activity alone is not sufficient to mimic the effect of SELENBP1-mediated induction of p21 protein. Conversely, blockade of STAT1 activity alone by DN-STAT1 Y701F slightly increased the expression of p21 protein (Fig. 6c), though dominant-negative STAT1 resulted in an effective down-regulation of suppressor of cytokine signaling 1 (SOCS1), a negative regulator of JAK/STAT signaling [45]. These observations led us to hypothesize whether simultaneous blockade of c-Jun and STAT1 activity is required for the induction of p21 protein. To verify this notion, we introduced pcDNA3.1 control and TAM67 into stable UMUC3 (Vector) and UMUC3 (DN-STAT1 Y701F) transfectants, respectively. As shown in Fig. 6d, concurrent blockade of c-Jun and STAT1 activities resulted in a significant increase in the expression of p21 protein, revealing that suppression of both c-Jun and STAT1 activity is indispensable for SELENBP1-mediated induction of p21 protein. Consistent with the induction of p21 protein, the truncated-length (1.3 Kb) of human p21 promoter-driven luciferase reporter assay revealed that p21 promoter activity increased significantly following the introduction of increasing amounts of TAM67 into stable UMUC3 (DN-STAT1 Y701F) cells (Fig. 6e). Meanwhile, we noticed that inhibition of c-Jun phosphorylation by TAM67 alone attenuated p21 promoter activity in stable UMUC3 (Vector) cells (Fig. 6e). This observation was, however, in agreement with TAM67-mediated inhibition of p21 protein (Fig. 6b), suggesting that c-Jun regulates p21 expression in a context-dependent manner, at least dependent on the STAT1 activity. More importantly, we found that simultaneous blockade of c-Jun and STAT1 activity induced a significant G0/G1 phase arrest in UMUC3 cells (Fig. 6f). These mechanistic insights further highlight the indispensable roles for both c-Jun and STAT1 in SELENBP1-mediated transcriptional induction of p21 protein and subsequent G0/G1 phase arrest. Taken together, our results demonstrate overexpression of SELENBP1 triggers transcriptional activation of p21 through concurrent suppression of c-Jun and STAT1 phosphorylation.

Discussion

Reduced expression of selenium-binding protein 1 (SELENBP1), since first cloned in 1997, has been frequently observed in a variety of solid tumors, and is significantly related to unfavorable clinical outcome in multiple cancer types [23]. However, the evidence concerning the clinical relevance of SELENBP1 in human bladder cancer is lacking. In the present study, we are the first to show that the reduction of SELENBP1 expression is also a frequent event in human bladder cancer tissues (Fig. 1), but is more likely to occur in patients with non-papillary subtype and advanced TNM stage (Table 1). Furthermore, restoration of SELENBP1 expression results in a significant G0/G1 phase arrest and subsequent cell growth inhibition in human bladder cancer cells (Figs. 3 and 4). Collectively, we report here that SELENBP1 may serve as a promising prognostic biomarker in predicting poor clinical outcome among patients with bladder cancer. Nevertheless, how SELENBP1 is clinically incorporated into bladder cancer management together with other prognostic biomarkers merits further investigation.

Cytosine methylation is a DNA modification generally associated with epigenetic transcription silencing [46]. The publicly available TCGA-BLCA data, analyzed by the online MEXPRESS tool [41], indicates that multiple locations of SELENBP1 gene are hyper-methylated in a majority of bladder cancer tumors (Fig. 2). In line with prior reports in esophageal and colorectal cancer [11, 42], DNA methylation in the immediate vicinity of the TSS is inversely associated with SELENBP1 expression (Pearson correlation coefficients ranging from − 0.339 to − 0.505; p < 0.001). Intriguingly, this inverse association is much more significant between methylation in gene body and SELENBP1 expression (Pearson r = − 0.619, p < 0.001), suggesting that DNA hypermethylation, especially in gene body, is likely to account for the reduction of SELENBP1 expression in bladder cancer. However, there is a significant and positive relationship between DNA methylation in 3′-UTR region and SELENBP1 expression (Pearson r = + 0.482; p < 0.001). This apparent paradox supports that DNA methylation in different genomic contexts might be differentially involved in the regulation of SELENBP1 expression [47]. Recently, DNA methylation-based biomarker studies have predominantly been focused on the effects of hypermethylation of promoter in tumour-suppressor genes [48]. Therefore, our finding provides valuable information when developing SELENBP1 as a genomic methylation-based biomarker for tumor progression and prognostic outcome.

To date, the molecular functions of SELENBP1 in cancer are relatively poorly understood, although it has been consistently reported to attenuate aberrant cell growth and suppress cell migration in a variety of cancer cells [49,50,51]. We and others have previously shown that SELENBP1 has the ability to form a physical interaction with glutathione peroxidase 1 selenoprotein [17, 52], a well-characterized enzyme that detoxify hydrogen peroxide to nontoxic water [53]. These findings implies a novel role for SELENBP1 in modulating the cellular redox microenvironment, which is further supported by our recent report showing that SELENBP1 regulates the levels of extracellular reduced form of glutathione [30]. More recently, SELENBP1 has been recognized as a novel human methanethiol oxidase with an enzymatic role in sulfur metabolism [54]. Here, we establish a novel biochemical link between SELENBP1 and p21 protein, and further determine the indispensable role for p21 in SELENBP1-mediated growth inhibition in cancer cells (Fig. 4). However, whether glutathione or sulfur metabolism is involved in SELENBP1-mediated induction of p21 protein warrants further investigation.

The p21 protein, also known as p21WAF1/CIP1, is encoded by the CDKN1A gene mapped to chromosome 6p21.2, and belongs to the Cip/Kip family of CDK inhibitors [55]. A substantial number of studies have demonstrated p21 as a multifunctional, broad-acting protein that plays key roles in cell cycle regulation, cell migration and apoptosis [44]. Thus, p21 has been shown to be intricately regulated via p53-dependent and –independent pathways [56]. Here, we verify that SELENBP1 activates transcriptional induction of p21 through a p53-independent mechanism using p53-deficiency cancer cell model (Fig. 5). Given the high prevalence of TP53 mutation and/or lose of function across almost all of cancer types, our finding suggests that SELENBP1 probably retains the growth-inhibitory capacity even in p53-mutated cancer cells.

Independent of p53, a variety of transcription factors have been reported to transcriptionally activate p21 protein expression [56], including Sp1. However, there is no observable effect on this player following the restoration of SELENBP1 in bladder cancer cells (Fig. 6). By contrast, SELENBP1 specifically inhibits the phosphorylation of c-Jun and STAT1, belonging to AP-1 and STAT family, respectively. Our further analysis suggests that combined suppression of c-Jun and STAT1 activities may be essential for SELENBP1-mediated transcriptional induction of p21 protein. C-Jun has been previously shown to interact with STAT3 and co-operatively regulate the transcription of their target genes in bladder cancer [57]. Given that c-Jun responsive element is closely located at the potential STAT1 binding sites in the promoter of p21, it is intriguing to further determine whether there is a physical interaction between c-Jun and STAT1 in the context of SELENBP1-mediated p21 expression, and the results from which would improve our understanding of the tumor-suppressive role for SELENBP1 in cancer.

Conclusion

In summary, our results have revealed that the reduction of SELENBP1 is a frequent event and significantly correlates with tumor progression as well as unfavorable prognosis among patients with bladder cancer. Further bioinformatics analysis suggests that DNA hypermethylation, especially in gene body, is likely to account for the reduction of SELENBP1 expression in bladder cancer. More importantly, we have established a novel biochemical link between SELENBP1 and p21 protein, in which SELENBP1 activates transcriptional induction of p21 expression in a p53-independent mechanism. Taken together, our results provide clinical and molecular evidence for an improved understanding of the tumor-suppressive role of SELENBP1 in bladder cancer, suggesting that SELENBP1 could potentially be used as a prognostic biomarker as well as a therapeutic target in future cancer therapy. Therefore, how SELENBP1 is clinically incorporated into bladder cancer management merits further investigation.

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

SELENBP1:

selenium-binding protein 1

p21:

cyclin-dependent kinase inhibitor 1A

p27:

cyclin-dependent kinase inhibitor 1B

STAT1:

signal transduction and activator of transcription 1

SOCS1:

suppressor of cytokine signaling 1

AP-1:

activating protein-1

Sp1:

specificity protein 1

CDK4:

cyclin-dependent kinase 4

CDK6:

cyclin-dependent kinase 6

UTR:

untranslated region

TSS:

transcription start site

OS:

overall survival

TCGA-BLCA:

The Cancer Genome Atlas bladder cancer

References

  1. Abdollah F, Gandaglia G, Thuret R, et al. Incidence, survival and mortality rates of stage-specific bladder cancer in United States: a trend analysis. Cancer Epidemiol. 2013;37(3):219–25.

    Article  PubMed  Google Scholar 

  2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7–30.

    Article  PubMed  Google Scholar 

  3. Kamat AM, Hahn NM, Efstathiou JA, et al. Bladder cancer. Lancet. 2016;388(10061):2796–810.

    Article  PubMed  Google Scholar 

  4. Alfred Witjes J, Lebret T, Comperat EM, et al. Updated 2016 EAU guidelines on muscle-invasive and metastatic bladder cancer. Eur Urol. 2017;71(3):462–75.

    Article  CAS  PubMed  Google Scholar 

  5. Chang PW, Tsui SK, Liew C, Lee CC, Waye MM, Fung KP. Isolation, characterization, and chromosomal mapping of a novel cDNA clone encoding human selenium binding protein. J Cell Biochem. 1997;64(2):217–24.

    Article  CAS  PubMed  Google Scholar 

  6. Bansal MP, Mukhopadhyay T, Scott J, Cook RG, Mukhopadhyay R, Medina D. DNA sequencing of a mouse liver protein that binds selenium: implications for selenium’s mechanism of action in cancer prevention. Carcinogenesis. 1990;11(11):2071–3.

    Article  CAS  PubMed  Google Scholar 

  7. Bansal MP, Oborn CJ, Danielson KG, Medina D. Evidence for two selenium-binding proteins distinct from glutathione peroxidase in mouse liver. Carcinogenesis. 1989;10(3):541–6.

    Article  CAS  PubMed  Google Scholar 

  8. Yang M, Sytkowski AJ. Differential expression and androgen regulation of the human selenium-binding protein gene hSP56 in prostate cancer cells. Cancer Res. 1998;58(14):3150–3.

    CAS  PubMed  Google Scholar 

  9. Schott M, de Jel MM, Engelmann JC, et al. Selenium-binding protein 1 is down-regulated in malignant melanoma. Oncotarget. 2018;9(12):10445–56.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Chen G, Wang H, Miller CT, et al. Reduced selenium-binding protein 1 expression is associated with poor outcome in lung adenocarcinomas. J Pathol. 2004;202(3):321–9.

    Article  CAS  PubMed  Google Scholar 

  11. Silvers AL, Lin L, Bass AJ, et al. Decreased selenium-binding protein 1 in esophageal adenocarcinoma results from posttranscriptional and epigenetic regulation and affects chemosensitivity. Clin Cancer Res. 2010;16(7):2009–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. He QY, Cheung YH, Leung SY, Yuen ST, Chu KM, Chiu JF. Diverse proteomic alterations in gastric adenocarcinoma. Proteomics. 2004;4(10):3276–87.

    Article  CAS  PubMed  Google Scholar 

  13. Zhang J, Dong WG, Lin J. Reduced selenium-binding protein 1 is associated with poor survival rate in gastric carcinoma. Med Oncol. 2011;28(2):481–7.

    Article  PubMed  CAS  Google Scholar 

  14. Xia YJ, Ma YY, He XJ, Wang HJ, Ye ZY, Tao HQ. Suppression of selenium-binding protein 1 in gastric cancer is associated with poor survival. Hum Pathol. 2011;42(11):1620–8.

    Article  CAS  PubMed  Google Scholar 

  15. Kim H, Kang HJ, You KT, et al. Suppression of human selenium-binding protein 1 is a late event in colorectal carcinogenesis and is associated with poor survival. Proteomics. 2006;6(11):3466–76.

    Article  PubMed  Google Scholar 

  16. Li T, Yang W, Li M, et al. Expression of selenium-binding protein 1 characterizes intestinal cell maturation and predicts survival for patients with colorectal cancer. Mol Nutr Food Res. 2008;52(11):1289–99.

    Article  CAS  PubMed  Google Scholar 

  17. Huang C, Ding G, Gu C, et al. Decreased selenium-binding protein 1 enhances glutathione peroxidase 1 activity and downregulates HIF-1alpha to promote hepatocellular carcinoma invasiveness. Clin Cancer Res. 2012;18(11):3042–53.

    Article  CAS  PubMed  Google Scholar 

  18. Zhang S, Li F, Younes M, Liu H, Chen C, Yao Q. Reduced selenium-binding protein 1 in breast cancer correlates with poor survival and resistance to the anti-proliferative effects of selenium. PLoS ONE. 2013;8(5):e63702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Huang KC, Park DC, Ng SK, et al. Selenium binding protein 1 in ovarian cancer. Int J Cancer. 2006;118(10):2433–40.

    Article  CAS  PubMed  Google Scholar 

  20. Chen F, Chen C, Qu Y, et al. Selenium-binding protein 1 in head and neck cancer is low-expression and associates with the prognosis of nasopharyngeal carcinoma. Medicine. 2016;95(35):e4592.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Zhang XY, Gao PT, Yang X, et al. Reduced selenium-binding protein 1 correlates with a poor prognosis in intrahepatic cholangiocarcinoma and promotes the cell epithelial–mesenchymal transition. Am J Transl Res. 2018;10(11):3567–78.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ha YS, Lee GT, Kim YH, et al. Decreased selenium-binding protein 1 mRNA expression is associated with poor prognosis in renal cell carcinoma. World J Surg Oncol. 2014;12:288.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Elhodaky M, Diamond AM. Selenium-binding protein 1 in human health and disease. Int J Mol Sci. 2018;19(11):3437.

    Article  PubMed Central  CAS  Google Scholar 

  24. Zhang D, Wang Y, Liang Y, et al. Loss of p27 upregulates MnSOD in a STAT3-dependent manner, disrupts intracellular redox activity and enhances cell migration. J Cell Sci. 2014;127(Pt 13):2920–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sun W, Li S, Yu Y, et al. MicroRNA-3648 is upregulated to suppress TCF21, resulting in promotion of invasion and metastasis of human bladder cancer. Mol Ther Nucleic Acids. 2019;16:519–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jin H, Yu Y, Hu Y, et al. Divergent behaviors and underlying mechanisms of cell migration and invasion in non-metastatic T24 and its metastatic derivative T24T bladder cancer cell lines. Oncotarget. 2015;6(1):522–36.

    Article  PubMed  Google Scholar 

  27. Zhang R, Wang Y, Li J, Jin H, Song S, Huang C. The Chinese herb isolate yuanhuacine (YHL-14) induces G2/M arrest in human cancer cells by up-regulating p21 protein expression through an p53 protein-independent cascade. J Biol Chem. 2014;289(10):6394–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhu J, Tian Z, Li Y, et al. ATG7 promotes bladder cancer invasion via autophagy-mediated increased ARHGDIB mRNA stability. Adv Sci. 2019;6(8):1801927.

    Article  CAS  Google Scholar 

  29. Zhu J, Li Y, Chen C, et al. NF-kappaB p65 overexpression promotes bladder cancer cell migration via FBW7-mediated degradation of RhoGDIalpha protein. Neoplasia. 2017;19(9):672–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang Y, Fang W, Huang Y, et al. Reduction of selenium-binding protein 1 sensitizes cancer cells to selenite via elevating extracellular glutathione: a novel mechanism of cancer-specific cytotoxicity of selenite. Free Radic Biol Med. 2015;79:186–96.

    Article  CAS  PubMed  Google Scholar 

  31. Wang Y, Xu J, Gao G, et al. Tumor-suppressor NFkappaB2 p100 interacts with ERK2 and stabilizes PTEN mRNA via inhibition of miR-494. Oncogene. 2016;35(31):4080–90.

    Article  CAS  PubMed  Google Scholar 

  32. Timofeeva OA, Plisov S, Evseev AA, et al. Serine-phosphorylated STAT1 is a prosurvival factor in Wilms’ tumor pathogenesis. Oncogene. 2006;25(58):7555–64.

    Article  CAS  PubMed  Google Scholar 

  33. Westfall MD, Mays DJ, Sniezek JC, Pietenpol JA. The Delta Np63 alpha phosphoprotein binds the p21 and 14-3-3 sigma promoters in vivo and has transcriptional repressor activity that is reduced by Hay-Wells syndrome-derived mutations. Mol Cell Biol. 2003;23(7):2264–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Liang Y, Zhu J, Huang H, et al. SESN2/sestrin 2 induction-mediated autophagy and inhibitory effect of isorhapontigenin (ISO) on human bladder cancers. Autophagy. 2016;12(8):1229–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Jiang G, Wu AD, Huang C, et al. Isorhapontigenin (ISO) inhibits invasive bladder cancer formation in vivo and human bladder cancer invasion in vitro by targeting STAT1/FOXO1 axis. Cancer Prev Res. 2016;9(7):567–80.

    Article  CAS  Google Scholar 

  36. Song L, Li J, Ye J, et al. p85alpha acts as a novel signal transducer for mediation of cellular apoptotic response to UV radiation. Mol Cell Biol. 2007;27(7):2713–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu J, Zhang D, Mi X, et al. p27 suppresses arsenite-induced Hsp27/Hsp70 expression through inhibiting JNK2/c-Jun- and HSF-1-dependent pathways. J Biol Chem. 2010;285(34):26058–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zeng X, Xu Z, Gu J, et al. Induction of miR-137 by isorhapontigenin (ISO) directly targets Sp1 protein translation and mediates its anticancer activity both in vitro and in vivo. Mol Cancer Ther. 2016;15(3):512–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang D, Li J, Costa M, Gao J, Huang C. JNK1 mediates degradation HIF-1alpha by a VHL-independent mechanism that involves the chaperones Hsp90/Hsp70. Cancer Res. 2010;70(2):813–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Goldman M, Craft B, Hastie M, et al. The UCSC Xena platform for public and private cancer genomics data visualization and interpretation. bioRxiv. 326470. https://doi.org/10.1101/326470.

  41. Koch A, De Meyer T, Jeschke J, Van Criekinge W. MEXPRESS: visualizing expression, DNA methylation and clinical TCGA data. BMC Genomics. 2015;16:636.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Pohl NM, Tong C, Fang W, Bi X, Li T, Yang W. Transcriptional regulation and biological functions of selenium-binding protein 1 in colorectal cancer in vitro and in nude mouse xenografts. PLoS ONE. 2009;4(11):e7774.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. El-Deiry WS, Tokino T, Velculescu VE, et al. WAF1, a potential mediator of p53 tumor suppression. Cell. 1993;75(4):817–25.

    Article  CAS  PubMed  Google Scholar 

  44. Kreis NN, Louwen F, Yuan J. Less understood issues: p21(Cip1) in mitosis and its therapeutic potential. Oncogene. 2015;34(14):1758–67.

    Article  CAS  PubMed  Google Scholar 

  45. Endo TA, Masuhara M, Yokouchi M, et al. A new protein containing an SH2 domain that inhibits JAK kinases. Nature. 1997;387(6636):921–4.

    Article  CAS  PubMed  Google Scholar 

  46. Schubeler D. Function and information content of DNA methylation. Nature. 2015;517(7534):321–6.

    Article  CAS  PubMed  Google Scholar 

  47. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484–92.

    Article  CAS  PubMed  Google Scholar 

  48. Koch A, Joosten SC, Feng Z, et al. Analysis of DNA methylation in cancer: location revisited. Nat Rev Clin Oncol. 2018;15(7):459–66.

    Article  CAS  PubMed  Google Scholar 

  49. Yang W, Diamond AM. Selenium-binding protein 1 as a tumor suppressor and a prognostic indicator of clinical outcome. Biomark Res. 2013;1(1):15.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Jeong JY, Zhou JR, Gao C, Feldman L, Sytkowski AJ. Human selenium binding protein-1 (hSP56) is a negative regulator of HIF-1alpha and suppresses the malignant characteristics of prostate cancer cells. BMB Rep. 2014;47:411.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Ansong E, Ying Q, Ekoue DN, et al. Evidence that selenium binding protein 1 is a tumor suppressor in prostate cancer. PLoS ONE. 2015;10(5):e0127295.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Fang W, Goldberg ML, Pohl NM, et al. Functional and physical interaction between the selenium-binding protein 1 (SBP1) and the glutathione peroxidase 1 selenoprotein. Carcinogenesis. 2010;31(8):1360–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lubos E, Loscalzo J, Handy DE. Glutathione peroxidase-1 in health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2011;15(7):1957–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Pol A, Renkema GH, Tangerman A, et al. Mutations in SELENBP1, encoding a novel human methanethiol oxidase, cause extraoral halitosis. Nat Genet. 2018;50(1):120–9.

    Article  CAS  PubMed  Google Scholar 

  55. Moussa RS, Park KC, Kovacevic Z, Richardson DR. Ironing out the role of the cyclin-dependent kinase inhibitor, p21 in cancer: novel iron chelating agents to target p21 expression and activity. Free Radic Biol Med. 2019;133:276–94.

    Article  CAS  PubMed  Google Scholar 

  56. Warfel NA, El-Deiry WS. p21WAF1 and tumourigenesis: 20 years after. Curr Opin Oncol. 2013;25(1):52–8.

    Article  CAS  PubMed  Google Scholar 

  57. Itoh M, Murata T, Suzuki T, et al. Requirement of STAT3 activation for maximal collagenase-1 (MMP-1) induction by epidermal growth factor and malignant characteristics in T24 bladder cancer cells. Oncogene. 2006;25(8):1195–204.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Dr. Kuok In Ngo for critical reading this manuscript.

Funding

This study was supported by funding from National Natural Science Foundation of China (81602645), Natural Science Foundation of Guangdong Province (2018A030313292) and Research Funds from Guangzhou Municipal Science and Technology Project (201804010430). The funding body has no roles in the design of the study and collection, analysis and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

YW and GZ designed and performed experiments. GJ participated in sample collection, sample processing, clinical information collection. YW, WZ, XC, GW, GJ and GZ analyzed and interpreted data. YW, WZ, XC, GW and GZ participated in manuscript writing. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yulei Wang or Guochun Zhang.

Ethics declarations

Ethics approval and consent to participate

Before collecting these specimens, ethical approval has been obtained from the Medical Ethics Committee of China, and all patients have given their informed consent for translational research.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Zhu, W., Chen, X. et al. Selenium-binding protein 1 transcriptionally activates p21 expression via p53-independent mechanism and its frequent reduction associates with poor prognosis in bladder cancer. J Transl Med 18, 17 (2020). https://doi.org/10.1186/s12967-020-02211-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-020-02211-4

Keywords