Abstract
The oncogene MDM4, also known as MDMX or HDMX, contributes to cancer susceptibility and progression through its capacity to negatively regulate a range of genes with tumour-suppressive functions. As part of a recent genome-wide association study it was determined that the A-allele of the rs4245739 SNP (A>C), located in the 3′-UTR of MDM4, is associated with an increased risk of prostate cancer. Computational predictions revealed that the rs4245739 SNP is located within a predicted binding site for three microRNAs (miRNAs): miR-191-5p, miR-887 and miR-3669. Herein, we show using reporter gene assays and endogenous MDM4 expression analyses that miR-191-5p and miR-887 have a specific affinity for the rs4245739 SNP C-allele in prostate cancer. These miRNAs do not affect MDM4 mRNA levels, rather they inhibit its translation in C-allele-containing PC3 cells but not in LNCaP cells homozygous for the A-allele. By analysing gene expression datasets from patient cohorts, we found that MDM4 is associated with metastasis and prostate cancer progression and that targeting this gene with miR-191-5p or miR-887 decreases in PC3 cell viability. This study is the first, to our knowledge, to demonstrate regulation of the MDM4 rs4245739 SNP C-allele by two miRNAs in prostate cancer, and thereby to identify a mechanism by which the MDM4 rs4245739 SNP A-allele may be associated with an increased risk for prostate cancer.
Introduction
Globally, prostate cancer is the second most common cancer in men (Siegel et al. 2014), with a significant proportion of cases attributed to heritable genetic conditions (Siegel et al. 2014) that influence not only cancer susceptibility but also its clinical course. MDM4 is an oncogene that negatively regulates p53 and several other tumour suppressor genes in prostate cancer and a range of other cancers (Kadakia et al. 2002, Jin et al. 2008, Mancini et al. 2009, Miller et al. 2010). The A-allele of the MDM4 rs4245739 SNP (A>C, GenBank accession no.
MDM4 rs4245739 SNP cancer association
Cancer | Reference | Outcome | Risk allele | Cases | Controls | OR/HR (95% CI) | P value |
---|---|---|---|---|---|---|---|
Ovarian | Wynendaele et al. (2010) | Recurrence | A | 66 | – | HR=4.2 (1.2–13.5) | 0.02 |
Ovarian | Wynendaele et al. (2010) | Tumour-related death | A | 66 | – | HR=5.5 (1.5–20.5) | 0.01 |
Prostate | Eeles et al. (2013) | Risk | A | 25 074 | 24 272 | 1.1 (1.05–1.14) | 2×10−11 |
Breast | Garcia-Closas et al. (2013) | Risk | A | 4193 | 35 194 | 1.14 (1.10–1.18) | 2×10−12 |
Esophageal | Zhou et al. (2013) | Risk (Jinan set) | C | 540 | 550 | OR=0.54 (0.35–0.82) | 0.004 |
Esophageal | Zhou et al. (2013) | Risk (Huaian set) | C | 588 | 600 | OR=0.68 (0.45–0.99) | 0.049 |
Breast | Liu et al. (2013) | Risk (Jinan Set) | C | 1100 | 1400 | OR=0.55 (0.4–0.76) | 2.3×10−4 |
Risk (Huaian Set) | OR=0.41 (0.25–0.67) | 3.1×10−4 |
Oestrogen receptor-negative ovarian cancer patients homozygous for the MDM4 rs4245739 A-allele were shown to have a 4.2-fold increased risk of recurrence and a 5.5-fold increased risk of tumour-related death. Larger scale genome-wide association studies have shown that the rs4245739 SNP A-allele is also associated with an increased risk of prostate and breast cancers. This multi-cancer risk locus has also been identified in a smaller scale oesophageal squamous cell carcinoma study in Chinese populations (two case/control sets – Jinan & Huaian) and a smaller scale breast cancer study in Chinese populations (two case/control sets – Jinan & Huaian) indicating that A/C and C/C genotypes were associated with a decreased risk. (OR, odds ratio; HR, hazard ratio). Ratios and 95% CIs were estimated by multivariate Cox regression and logistic regression.
miRNAs are short approximately 19–24 nucleotide non-coding RNA molecules that post-transcriptionally regulate gene expression via mRNA binding (RNA interference). In cancers, miRNAs have been shown to affect several key oncogenic processes including apoptosis, migration and proliferation (Fonseca-Sanchez et al. 2013, Huang et al. 2013, Pinho et al. 2013). Interestingly, widespread dysregulation of miRNAs has been observed in prostate cancer (Schaefer et al. 2010, Fendler et al. 2011, Wach et al. 2012), and these molecules have potential as both biomarkers and therapeutic targets/treatments (Lichner et al. 2013, Maugeri-Sacca et al. 2013). miR-191 is up-regulated in prostate, breast and colorectal cancers, though its role in these cancers has not been tested (Volinia et al. 2006, Xi et al. 2006, Hui et al. 2009). The capacity of other miRNAs to regulate MDM4 has been demonstrated in breast cancer, although these effects were not SNP-associated (Mandke et al. 2012, Hoffman et al. 2014). As MDM4 has also been proposed as a therapeutic target for several cancers (Garcia et al. 2011, Gembarska et al. 2012), it is important to further explore the MDM4-miRNA axis in prostate cancer.
Herein, we investigated the miR-191-5p/MDM4 rs4245739 interaction in prostate cancer as well as assessing the potential role of additional miRNAs that are predicted to target this sequence. We report herein that miR-191-5p and miR-887 have a specific affinity for the rs4245739 SNP C-allele in prostate cancer, presenting a mechanism by which the un-targeted A-allele may be associated with an increased risk of prostate cancer.
Materials and methods
Cell lines
Unless otherwise stated (see Acknowledgements), all cell lines were originally sourced from the American Type Culture Collection (ATCC) (www.atcc.org). Cell line authentication (STR profiling) was performed by either the Queensland Institute of Medical Research (Brisbane, Queensland, Australia) or DDC Medical (Fairfield, OH, USA).
miRNA target reporter vector assays
The miRNA-SNP online prediction tool, mirsnpscore available at www.bigr.medisin.ntnu.no/mirsnpscore (Thomas et al. 2011), was used to identify miR-191-5p, miR-887 and miR-3669 (Eeles et al. 2013) all predicted to preferentially target the MDM4 rs4245739 SNP C-allele. To validate in silico predictions for miRNA-SNP affinity, miRNA target luciferase reporter vector assays were performed. Reporter vectors were constructed for the major and minor alleles of the MDM4 rs4245739 SNP using the pmirGLO Dual-Luciferase miRNA Target Expression Vector (Promega) with MDM4 3′-UTR inserts pertaining to predicted miRNA binding (C allele insert 5′-GAACATAAAAATGCATTTATTCCGTTCACTTA-3′, A allele insert 5′-GAACATAAAAATGCATTTATTCAGTTCACTTA-3′) synthesised by Integrated DNA Technologies (Baulkham Hills, New South Wales, Australia). The prostate cancer cell line, PC3 (passage 8), was plated at a density of 50 000 cells/well on a 24-well plate and cultured overnight, transiently transfected with 50 ng vector and 60 nM mirVana miRNA Mimics (Life Technologies) using FuGENE transfection reagent (Promega), then analysed 24 h later using the Dual-Luciferase Reporter Assay System (Promega) according to the manufacturer's instructions. Luciferase levels were normalised to Renilla luciferase co-expressed from the same vector. A negative control mirVana miRNA Mimic (Negative Control #1 – Life Technologies) was used for analysis alongside candidate miRNAs as was a perfect match positive control miRNA mimic (Customer-defined mirVana miRNA Mimic – Life Technologies) designed for the rs4245739 SNP, C-allele – 5′-GUGAACGGAAUAAAUGCAUUUU-3′. miR-331-3p which is not predicted to bind with MDM4 was used as a second negative control. For a single experiment, each miRNA/vector treatment was cultured in triplicate, then each sample was assayed in duplicate. A total of three independent experiments were performed.
Cell line genotyping
Genomic DNA was isolated from cultured cells using the DNA portion of the AllPrep DNA/RNA/Protein Mini Kit (Qiagen). The rs4245739 region was PCR amplified using the forward primer 5′-tattcatggaaggacgggccatct-3′ and the reverse primer 5′-gcctaagaacattctctgacaggttgg-3′. The PCR product was purified using the Wizard SV Gel and PCR Clean-up System (Promega – cat # A9282). Samples were then sequenced at the Australian Genome Research Facility (St Lucia, Queensland, Australia) using the forward primer 5′-tgccagaagactaaagaaggctgg-3′.
Western blotting
The prostate cancer cell lines LNCaP and PC3, and the ovarian cancer cell line OV90 were cultured under standard conditions. Cells were plated (passage 8) at a density of approximately 200 000 cells/well on a six-well plate, cultured overnight, and then transiently transfected with 30 nM of mirVana miRNA Mimics using Lipofectamine RNAiMAX transfection reagent (Life Technologies). Forty-eight hours later, total protein was isolated using SDS lysis buffer (1% SDS, 5% glycerol, 10 nM Tris, Roche Complete protease inhibitor), total protein concentration was assessed by the BCA method and 50 μg of total protein was run on a 12% resolving poly-acrylamide gel. Western blotting was performed using Millipore Immobilon FL PVDF membranes with a rabbit anti-MDM4 antibody (Bethyl Labs A300-287A (Bethyl Labs, Montgomery, TX, USA)) diluted 1:3000 and incubated at room temperature for 2 h. Normalisation was performed using either mouse anti β-actin (Abcam – ab8224 (Abcam, Cambridge, UK)) or rabbit anti β-actin (Abcam – ab25894) or mouse anti β-tubulin (Sigma – T4026). Western blots were imaged on an Odyssey Imaging System (LI-COR Biosciences, Lincoln, NE, USA) using fluorescently labelled secondary antibodies (Alexa Fluor 680 and 790 – Invitrogen) with protein band intensities analysed via densitometry using the Odyssey Imaging System software.
RT-qPCR analysis to assess MDM4 expression
Total RNA was isolated using TRIzol Reagent (Life Technologies) and assessed for quality and quantity using a Nanodrop ND-1000 spectrophotometer. Samples (500 ng) of RNA were reversed transcribed using oligo dT primer. qPCR was then performed using the SYBR Green PCR Master Mix (Life Technologies) in triplicate with β-actin used as an endogenous quantitative normalisation control. Relative expression levels were calculated using the comparative threshold cycle (Ct) method. Primers were synthesised by Integrated DNA Technologies. Primer sequences for MDM4 were as follows: forward primer 5′-tgaacatttcaccttgcgcacctg-3′ and reverse primer 5′-caacatctgacagtgcttgcagga-3′ as described previously (Wynendaele et al. 2010). Primer sequences for β-actin were as follows: forward primer 5′-gcgttacaccctttcttgacaaaacct-3′ and reverse primer 5′-gctgtcaccttcaccgttcca-3′.
Analysis of miR-191-5p and miR-887 expression in human prostate tissue datasets
The expression of miR-191-5p and miR-887 in prostatic tissues was assessed in two datasets, one from The Cancer Genome Atlas (TCGA) and one from Memorial Sloan-Kettering Cancer Centre (MSKCC) (Taylor et al. 2010). For the TCGA dataset, normalised small RNA-seq data from 231 tissue samples (181 tumours and 50 normal prostate tissues), generated on an Illumina HiSeq platform, were downloaded from the web portal (https://tcga-data.nci.nih.gov/tcga/tcgaDownload.jsp). Expression values for candidate miRNAs for each sample were set to log2 of reads per million mapped miRNAs. The MSKCC Agilent microarray dataset, which comprises 99 tumours and 28 normal tissues, was processed as described previously (Selth et al. 2012).
RT-qPCR analysis to assess miRNA expression
Total RNA was extracted from cell lines using TRIzol reagent as described earlier in this study. To assess miRNA expression, RT and qPCR (in triplicate) were performed using the TaqMan MicroRNA Reverse Transcription Kit and TaqMan MicroRNA Assays (Life Technologies). Relative expression levels were calculated using the comparative Ct method with the small nuclear RNA RNU24 used as an endogenous quantitative normalisation control as it has been identified as being the most consistent option with the least variability for prostate cancer (Carlsson et al. 2010).
Alamar blue cell viability assay
PC3 cells (passage 8) were plated at a density of 10 000 cells/well on a 96-well plate (black, clear-bottom) and cultured overnight, then transiently transfected with 30 nM of mirVana miRNA Mimics using Lipofectamine RNAiMAX transfection reagent. Twenty-four hours after transfection, cells were treated with Alamar Blue Cell Viability Reagent (Life Technologies) according to the manufacturer's instructions and fluorescent measurements were taken using a FLUOstar Omega plate reader (BMG Labtech, Ortenberg, Germany). Positive and negative control miRNA mimics were used as described above.
Androgen and anti-androgen treatment of LNCaP cells
The androgen receptor-positive, LNCaP prostate cancer cell line was treated with androgen (10 nM dihydrotestosterone (DHT)) (Sigma–Aldrich), or therapeutic anti-androgens (10 μM bicalutamide) (Selleckchem.com, Waterloo, Australia) for 24 h as described previously (Lai et al. 2014). RNA was extracted from cells using Tri-reagent (Life Technologies), and reverse transcribed using superscript III (Life Technologies).
Statistical and other analyses
Unless otherwise stated, for all analyses, three independent experiments were conducted with results expressed as mean±s.d. and analysed using Student's t-test with a P value of <0.05 considered statistically significant. MDM4 mRNA expression analysis (eQTL analysis) was conducted using the Genevar web tool (http://www.sanger.ac.uk/resources/software/genevar/).
Results
MDM4 expression is associated with metastasis and recurrence after radical prostatectomy
Before functional analysis of the MDM4 rs4245739 SNP, we mined data from existing databases to assess MDM4 expression levels in relation to prostate cancer prognosis. Using Oncomine (www.oncomine.org), a dataset comparing MDM4 mRNA expression in 59 primary prostate cancer tissue samples with 34 metastatic samples (Grasso et al. 2012) revealed a 3.35-fold increase in MDM4 expression (P=1.75×10−11) in the metastatic samples (Fig. 1A). A separate dataset comprising gene expression in primary epithelial cultures derived from tissue explanted from 17 patients undergoing radical prostatectomy (Nanni et al. 2006) was also analysed, revealing a 5.422-fold increase in MDM4 mRNA (P=0.002) in patients whose cancer recurred biochemically 1 year after prostatectomy compared with patients with no recurrence, indicating a potential role of MDM4 in the development of aggressive disease and relapse after treatment (Fig. 1B).
miR-191-5p and miR-887 have a higher affinity for the MDM4 rs4245739 SNP C-allele
Using mirsnpscore (www.bigr.medisin.ntnu.no/mirsnpscore; (Thomas et al. 2011)), MDM4 rs4245739 SNP was predicted to affect the binding of three miRNAs to the MDM4 3′-UTR (Eeles et al. 2013). These included miR-191-5p, miR-887 and miR-3669, which were all predicted to have a higher affinity for the C-allele (Fig. 2A). Reporter vector assays were performed using PC3 cells to test the validity of these in silico predictions. miR-191-5p and miR-887 induced a 27 and 28% decrease, respectively, in luciferase activity for the MDM4 C-allele construct. In contrast, these miRNAs did not regulate the A-allele construct, indicating that they have a specific affinity for the rs4245739 SNP C-allele (Fig. 2B). No significant changes were observed for miR-3669 with either SNP variant; therefore, this miRNA was not assessed further.
miR-191-5p and miR-887 affects MDM4 protein levels in C-allele-containing cell lines
We then tested whether miR-191-5p and miR-887 were able to affect endogenous MDM4 protein levels in C-allele-containing cell lines. Six model prostate cancer and six model ovarian cancer cell lines were genotyped for the rs4245739 SNP. All prostate cancer cell lines (LNCaP, LAPC4, DUCaP, DU145 and 22RV1) were homozygous A/A for the rs4245739 SNP except PC3 cells, which were found to be heterozygous A/C. The ovarian cancer cell lines OAW42, OVMZ6, SKOV3 and OVCAR3 were homozygous A/A while OV90 and CAOV3 were found to be heterozygous A/C (Table 2). Interestingly, there were significantly lower MDM4 protein (Supplementary Figure 1A, see section on supplementary data given at the end of this article) and mRNA (Supplementary Figure 1B) levels in the heterozygous A/C lines (PC3 and OV90) compared with the lines homozygous A/A for the rs4245739 SNP.
Genotyping results for the MDM4 rs4245739 SNP in model prostate cancer and ovarian cancer cell lines
Cancer | Cell line | A/A | A/C | C/C |
---|---|---|---|---|
Prostate | LNCaP | ✓ | ||
Prostate | PC3 | ✓ | ||
Prostate | 22RV1 | ✓ | ||
Prostate | Du145 | ✓ | ||
Prostate | LAPC4 | ✓ | ||
Prostate | DUCaP | ✓ | ||
Ovarian | OAW42 | ✓ | ||
Ovarian | OVMZ6 | ✓ | ||
Ovarian | SKOV3 | ✓ | ||
Ovarian | OVCAR3 | ✓ | ||
Ovarian | OV90 | ✓ | ||
Ovarian | CAOV3 | ✓ |
Model cell lines for prostate and ovarian cancers were genotyped for the rs4245739 SNP. Results are expressed as either homozygous A/A or heterozygous A/C or homozygous C/C.
To directly assess the influence of miR-191-5p and miR-887 on MDM4 expression, these miRNAs were over-expressed in PC3 and LNCaP cells as models of A/C and A/A genotypes respectively. miRNA mimic transfection led to over-expression of miR-191-5p and miR-887 by >5000-fold as analysed by RT-qPCR. Over-expression of miR-191-5p and miR-887 resulted in a 38% (P value=0.037) and 52% (P value=0.035) decrease in MDM4 protein, respectively, in PC3 cells (Fig. 3A), whereas no decrease in MDM4 protein was observed following miRNA over-expression in LNCaP cells, which are homozygous A/A (Fig. 3B). The miRNA mimics did not affect MDM4 mRNA levels in PC3 cells (Supplementary Figure 1C), indicating that they act to inhibit protein translation rather than causing mRNA degradation.
Though miR-191-5p affinity for the C-allele had been reported previously in ovarian cancer, a role for miR-887 had not been tested (Wynendaele et al. 2010). We demonstrated that miR-191-5p along with miR-887 can reduce MDM4 protein levels in an ovarian cancer model using the OV90 cell line heterozygous A/C for the rs4245739 SNP (Supplementary Figure 1D). Collectively, these findings indicate that miR-191-5p and miR-887 preferentially target the MDM4 rs4245739 SNP C-allele in prostate and ovarian cancers.
miR-191-5p and miR-887 are up-regulated in prostate cancer
Prostatic expression of miR-191-5p has been reported previously, with levels shown to be up-regulated in prostate cancer compared with normal controls (Volinia et al. 2006). To further assess the biological relevance of miR-191-5p and to gain insight into miR-887 in prostate cancer, we first assessed prostatic expression for these miRNAs in a large cohort from TCGA comprising 181 tumours and 50 benign prostate tissues. In this RNA-seq dataset, both miRNAs were readily detectable in benign and malignant tissues, with miR-191-5p being expressed at considerably higher levels (average 208.5 reads/million mapped miRNA reads; s.d. 111.9; range 32.0–691.1) compared with miR-887 (average 2.3; s.d. 1.7; range 0.25–13.4) (Fig. 4A). In the full TCGA dataset, miR-191-5p was significantly elevated in the tumours compared with normal tissues, but miR-887 was present at similar levels in the two tissues. An examination of a subset of the cohort comprising patient-matched tumour and benign samples (n=50 of each) revealed that both miRNAs were elevated in cancer tissues (Fig. 4B). We also examined the prostatic expression of miR-191-5p and miR-887 in another large publicly available cohort from the MSKCC (Taylor et al. 2010), where miR-887 was found to be expressed at higher levels in metastatic tissues compared with normal tissues or primary tumours (Fig. 4C). In contrast to the TCGA cohort, there was no significant difference in miRNA expression in tumours compared with a patient-matched set of benign samples (Fig. 4D). Collectively, these results indicate that both miR-191-5p and miR-887 are expressed in the normal and malignant prostate, highlighting their potential to play an important role in regulating prostatic MDM4 expression. To further validate these findings, the expression of miR-191-5p and miR-887 was examined in a range of prostatic cell lines. As expected from the patient data, RT-qPCR demonstrated that miR-191-5p and miR-887 were expressed in these lines (PC3, LNCaP, 22RV1 and RWPE1).
We also assessed if there was any negative correlation between miR-191-5p/miR-887 and MDM4 mRNA expression in TCGA and MSKCC cohorts, but no such correlation was observed.
miR-191-5p and miR-887 inhibit PC3 cell growth
If targeting of MDM4 by miR-191-5p and miR-887 is an important pathway regulating growth in prostate cancer, it would be expected that these miRNAs would act as growth suppressors in prostate cancer. To test this concept, we performed Alamar Blue cell viability assays, using PC3 cells transfected with miRNA mimics. Both miR-191-5p and miR-887 were able to induce a reduction in PC3 cell viability similar to the MDM4 siRNA positive control, indicating that these two miRNAs can act as tumour suppressors in this cell line (Fig. 5).
Correlation analysis of the rs4245739 genotype and MDM4 expression
To assess the biological relevance of the MDM4 rs4245739 SNP, we conducted a correlation analysis of its genotype with MDM4 mRNA expression using various publicly available datasets. Using data from the HapMap3 project (for lymphoblastoid cell line expression), we were able to show for two distinct populations (Caucasians of northern and western European descent and Mexican ancestry) (Stranger et al. 2012) a correlation between MDM4 mRNA expression and SNP genotype, with the highest expression levels in subjects with the homozygous A/A genotype and the lowest in those with homozygous C/C (European P=2.7×10−5, Mexican P=0.0236). We also assessed lymphoblastoid cell line expression data from a large nuclear family study (of British decent), which allows analyses focused on heritable expression traits (Liang et al. 2013). A strong correlation of MDM4 mRNA expression with SNP genotype was observed, which again indicated that the highest expression was associated with the A/A genotype (n=400, P=1.82×10−23). A third dataset derived from a study in 856 twins (Grundberg et al. 2012) also displayed a similar correlation for MDM4 mRNA expression with SNP genotype in multiple tissues (highest expression for the A/A genotype – P=adipose 1.44×10−5, lymphocyte 2.16×10−18, skin 1.71×10−7).
miR-191-5p and miR-887 are not androgen-regulated in LNCaP cells
Given the critical role of androgen signalling in prostate cancer, we tested whether miR-191-5p and/or miR-887 were androgen regulated in the androgen-dependent LNCaP cell line. We did not observe any significant change in the expression of either of the miRNAs in LNCaP cells upon treatment with androgen and/or anti-androgen (Fig. 6).
Discussion
MDM4 contributes to cancer susceptibility and progression through its capacity to negatively regulate a range of genes with tumour-suppressive functions. Hence, MDM4 expression levels are a key modulator of these effects. In this study, we found that increased MDM4 expression is associated with metastasis and recurrence after radical prostatectomy, highlighting the significance of this gene in prostate cancer (Nanni et al. 2006, Grasso et al. 2012). To determine how the MDM4 rs4245739 SNP allele ‘A’ may be associated with an increased risk of prostate cancers, we assessed the interaction of miR-191-5p and miR-887 with both the alleles of the MDM4 rs4245739 SNP and found that both these miRNAs have a specific affinity for the C-allele in prostate cancer. We demonstrated that both miRNAs are able to induce a decrease in MDM4 protein expression in PC3 cells (heterozygous A/C) but not in LNCaP cells (homozygous A/A). Homozygous C/C cell lines for prostate cancer were unavailable, which was not surprising considering the minor allele frequency of approximately 0.28 for the C-allele and the increased frequency of the A-allele in prostate cancer as it is associated with an increased risk. However, it has been previously revealed that in esophageal squamous cell carcinoma homozygous C/C as well as the heterozygous A/C rs4245739 genotypes were associated with a decreased risk compared with the A/A genotype, supporting our findings for prostatic cancer regarding the significance of even the heterozygous genotype (Zhou et al. 2013).
Furthermore, our results indicated that not only are miR-191-5p and miR-887 expressed in the normal prostate, but are also elevated in some tumours compared with local benign tissue. The MDM4 protein changes induced by both miR-191-5p and miR-887 indicate that the effects induced by miR-887 may be stronger than those induced by miR-191-5p. The effects of any given miRNA on a specific target gene's expression levels will not simply be a function of binding affinity, but will also be related to the expression levels of the given miRNA and its target genes, and the number and transcription levels of any additional genes targeted by the same miRNA. Thus, although expressed at relatively low levels, miR-887 may still significantly affect MDM4 function in vivo. Interestingly, the miRNA target prediction algorithm Targetscan (Human release 6.2, www.targetscan.org) predicts 54 putative target genes for miR-191-5p, but only ten for miR-887, indicating that the latter may have fewer genes ‘competing’ for binding. Hence, both miRNAs probably possess the capacity to significantly affect MDM4 activity in tumours. As putative tumour suppressor miRNAs in prostate cancer, both miR-191-5p and miR-887 may be assessed in future studies as potential candidate biomarkers or therapies.
In this study, we found only one cell line (PC3) to have the ‘C’ allele for the MDM4 genotype and we demonstrated that both miRNAs are able to induce a decrease in PC3 cell viability, further highlighting their potential as tumour suppressor miRNAs. However, to verify the MDM4-mediated effects of miR-191-5p/miR-887 miRNAs on cell viability, these results need to be replicated in an additional rs4245739 SNP ‘C’ allele-containing cell line. MDM4 has been shown to negatively regulate the tumour suppressor p53 in prostate cancer and several other cancers (Ramos et al. 2001, Wade et al. 2013, Wang et al. 2013). However, the reduction in cell viability following miR-191-5p/miR-887 over-expression cannot be attributed to enhanced p53 activity as PC3 cells are p53-null (Simone et al. 2013). miR-191-5p and miR-887 may therefore be modulating these effects through additional direct gene targets independent of MDM4. Alternatively, other MDM4-associated pathways may be involved. Given that the cell viability changes for both miRNAs were similar to the MDM4 siRNA-positive control, we believe the latter scenario is likely. Besides p53, MDM4 interacts with several other proteins including p21, a cyclin-dependent kinase inhibitor whose expression is associated with reduced proliferation in prostate cancer (Jin et al. 2008, Lee & Lu 2011, Peng et al. 2013), the transcription factor E2F1, which is known to play a role in cell-cycle regulation in prostate cancer (Strachan et al. 2003, Lee et al. 2013), the E3 ubiquitin ligase MDM2 that also harbours polymorphisms associated with the risk of prostate cancer (Badciong & Haas 2002, Yang et al. 2012) and both Smad3 and Smad4, which are major activating components of the transforming growth factor-β (TGF-β) signalling pathway that has been shown to repress growth in prostate cancer (Kadakia et al. 2002, Bjerke et al. 2014, Mishra et al. 2014). As MDM4 probably plays a role in cancer susceptibility and progression via multiple pathways, further study is required to assess the prostate-specific effects of miRNA-mediated MDM4 levels on each of these pathways.
Furthermore, we analysed the correlation between MDM4 mRNA and miR-191-5p and miR-887 in the only two publicly available prostate cancer datasets with matched miRNA:mRNA data – one from MSKCC and the other from TCGA. In both of these datasets, there was no correlation between MDM4 mRNA and miR-191-5p/miR-887. This is consistent with the data shown in Fig. 3 and Supplementary Figure 1C, where we observed that these miRNAs reduced the levels of MDM4 protein but not MDM4 mRNA. These results are not surprising given that the miRNA machinery frequently does not affect mRNA levels in situ but only inhibits translation of these genes (Bartel 2004). Unfortunately, MDM4 protein expression is not tested in the TCGA or MSKCC datasets; therefore, we were unable to test for negative correlations between the miRNAs and MDM4 protein in large patient cohorts. Nevertheless, using a small panel of prostate and ovarian cancer cell lines, we assessed endogenous MDM4 protein levels and detected significantly lower MDM4 levels in the heterozygous A/C lines (PC3 and OV90) compared with the lines homozygous A/A for the rs4245739 SNP, possibly at least in part reflecting endogenous miR-191-5p/miR-887 regulation of MDM4 expression via the C-allele in these heterozygous cell lines. A similar but weaker pattern was observed for MDM4 mRNA genotype-specific expression. Large-scale studies, using larger cell line panels and patient tissues, are now required in order to determine the true significance of correlations between MDM4 protein expression and genotype for prostatic tissues.
We also assessed MDM4 expression correlation with genotype in familial, multi-tissue and multi-population cohorts, revealing a strong correlation between the rs4245739 C-allele and lower MDM4 mRNA expression in multiple non-tumour cells. These observations are further emphasised by a recent large-scale study genotyping approximately 50 000 SNPs in approximately 2100 genes, which revealed significant correlations between mRNA expression and the MDM4 SNP rs2169137 which is in linkage disequilibrium with rs4245739 SNP (Ganesh et al. 2013).
Finally, in an effort to identify the mechanism of regulation of the two identified miRNAs, we analysed effects of androgen and anti-androgen on their expression. We did not observe any significant effects of either DHT or bicalutamide on the expression of miR-191-5p or miR-887. Therefore, the mechanism by which these miRNAs are regulated in prostate cancer remains to be elucidated in future studies.
In conclusion, herein, we identified an association between the MDM4 rs4245739 SNP and two miRNAs, miR-191-5p and miR-887. This not only validates findings from studies of ovarian cancer but also identifies new modes of MDM4 regulation in prostate cancer. Specifically, the down-regulation of MDM4 in C-allele-containing prostate cancer-derived cells induced by miR-191-5p and miR-887 presents a mechanism by which the un-targeted A-allele may be associated with an increased risk of prostate cancer.
Supplementary data
This is linked to the online version of the paper at http://dx.doi.org/10.1530/ERC-15-0013.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Funding
This work was supported by the Cure Cancer and Cancer Australia Priority-driven Collaborative Cancer Research Scheme (PdCCRS) funding (J Batra) and National Health and Medical Research Council (NHMRC) (Career Development Fellowship to J Batra, grant number 1050742), Young Investigator Awards from the Prostate Cancer Foundation (the Foundation 14 award; L A Selth), and the Prostate Cancer Foundation of Australia (grant number YI 0810; L A Selth). A B Spurdle was supported by an NHMRC Senior Research Fellowship; J A Clements is an NHMRC Principal Research Fellow.
Acknowledgements
The authors would like to thank Ying Dong for providing the ovarian cancer cell lines (originally sourced from the ATCC) used in this study and Charles Sawyers for providing the prostate cancer cell line LAPC4 and Matthias Nees for providing the prostate cancer cell line DuCaP. They would also like to thank John Lai and Karen Chambers for technical assistance. Additionally, the results published herein are based partly on data generated by TCGA and established by the National Cancer Institute and the National Human Genome Research Institute, and they are grateful to the specimen donors and relevant research groups associated with this project.
References
Badciong JC & Haas AL 2002 MdmX is a RING finger ubiquitin ligase capable of synergistically enhancing Mdm2 ubiquitination. Journal of Biological Chemistry 277 49668–49675. (doi:10.1074/jbc.M208593200).
Bartel DP 2004 MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116 281–297. (doi:10.1016/S0092-8674(04)00045-5).
Bjerke GA, Yang CS, Frierson HF, Paschal BM & Wotton D 2014 Activation of Akt signaling in prostate induces a TGFβ-mediated restraint on cancer progression and metastasis. Oncogene 33 3660–3667. (doi:10.1038/onc.2013.342).
Carlsson J, Helenius G, Karlsson M, Lubovac Z, Andren O, Olsson B & Klinga-Levan K 2010 Validation of suitable endogenous control genes for expression studies of miRNA in prostate cancer tissues. Cancer Genetics and Cytogenetics 202 71–75. (doi:10.1016/j.cancergencyto.2010.06.009).
Eeles RA, Olama AA, Benlloch S, Saunders EJ, Leongamornlert DA, Tymrakiewicz M, Ghoussaini M, Luccarini C, Dennis J & Jugurnauth-Little S et al. 2013 Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nature Genetics 45 385–391, 391e381–382. (doi:10.1038/ng.2560).
Fendler A, Jung M, Stephan C, Honey RJ, Stewart RJ, Pace KT, Erbersdobler A, Samaan S, Jung K & Yousef GM 2011 miRNAs can predict prostate cancer biochemical relapse and are involved in tumor progression. International Journal of Oncology 39 1183–1192. (doi:10.3892/ijo.2011.1128).
Fonseca-Sanchez MA, Perez-Plasencia C, Fernandez-Retana J, Arechaga-Ocampo E, Marchat LA, Rodriguez-Cuevas S, Bautista-Pina V, Arellano-Anaya ZE, Flores-Perez A & Diaz-Chavez J et al. 2013 microRNA-18b is upregulated in breast cancer and modulates genes involved in cell migration. Oncology Reports 30 2399–2410. (doi:10.3892/or.2013.2691).
Ganesh SK, Tragante V, Guo W, Guo Y, Lanktree MB, Smith EN, Johnson T, Castillo BA, Barnard J & Baumert J et al. 2013 Loci influencing blood pressure identified using a cardiovascular gene-centric array. Human Molecular Genetics 22 1663–1678. (doi:10.1093/hmg/dds555).
Garcia D, Warr MR, Martins CP, Brown Swigart L, Passegue E & Evan GI 2011 Validation of MdmX as a therapeutic target for reactivating p53 in tumors. Genes and Development 25 1746–1757. (doi:10.1101/gad.16722111).
Garcia-Closas M, Couch FJ, Lindstrom S, Michailidou K, Schmidt MK, Brook MN, Orr N, Rhie SK, Riboli E & Feigelson HS et al. 2013 Genome-wide association studies identify four ER negative-specific breast cancer risk loci. Nature Genetics 45 392–398, 398e391–392. (doi:10.1038/ng.2561).
Gembarska A, Luciani F, Fedele C, Russell EA, Dewaele M, Villar S, Zwolinska A, Haupt S, de Lange J & Yip D et al. 2012 MDM4 is a key therapeutic target in cutaneous melanoma. Nature Medicine 18 1239–1247. (doi:10.1038/nm.2863).
Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, Quist MJ, Jing X, Lonigro RJ & Brenner JC et al. 2012 The mutational landscape of lethal castration-resistant prostate cancer. Nature 487 239–243. (doi:10.1038/nature11125).
Grundberg E, Small KS, Hedman AK, Nica AC, Buil A, Keildson S, Bell JT, Yang TP, Meduri E & Barrett A et al. 2012 Mapping cis- and trans-regulatory effects across multiple tissues in twins. Nature Genetics 44 1084–1089. (doi:10.1038/ng.2394).
Hazelett DJ, Rhie SK, Gaddis M, Yan C, Lakeland DL, Coetzee SG, Ellipse/GAME-ON consortium; Practical consortium , Henderson BE, Noushmehr H & Cozen W et al. 2013 Comprehensive functional annotation of 77 prostate cancer risk loci. PLoS Genetics 10 e1004102. (doi:10.1371/journal.pgen.1004102).
Hoffman Y, Bublik DR, Pilpel Y & Oren M 2014 miR-661 downregulates both Mdm2 and Mdm4 to activate p53. Cell Death and Differentiation 21 302–309. (doi:10.1038/cdd.2013.146).
Huang F, Lin C, Shi YH & Kuerban G 2013 MicroRNA-101 inhibits cell proliferation, invasion, and promotes apoptosis by regulating cyclooxygenase-2 in hela cervical carcinoma cells. Asian Pacific Journal of Cancer Prevention 14 5915–5920. (doi:10.7314/APJCP.2013.14.10.5915).
Hui AB, Shi W, Boutros PC, Miller N, Pintilie M, Fyles T, McCready D, Wong D, Gerster K & Waldron L et al. 2009 Robust global micro-RNA profiling with formalin-fixed paraffin-embedded breast cancer tissues. Laboratory Investigation 89 597–606. (doi:10.1038/labinvest.2009.12).
Jin Y, Zeng SX, Sun XX, Lee H, Blattner C, Xiao Z & Lu H 2008 MDMX promotes proteasomal turnover of p21 at G1 and early S phases independently of, but in cooperation with, MDM2. Molecular and Cellular Biology 28 1218–1229. (doi:10.1128/MCB.01198-07).
Kadakia M, Brown TL, McGorry MM & Berberich SJ 2002 MdmX inhibits Smad transactivation. Oncogene 21 8776–8785. (doi:10.1038/sj.onc.1205993).
Lai J, An J, Nelson CC, Lehman ML, Batra J & Clements JA 2014 Analysis of androgen and anti-androgen regulation of KLK-related peptidase 2, 3, and 4 alternative transcripts in prostate cancer. Biological Chemistry 395 1127–1132. (doi:10.1515/hsz-2014-0149).
Lee JH & Lu H 2011 14-3-3γ inhibition of MDMX-mediated p21 turnover independent of p53. Journal of Biological Chemistry 286 5136–5142. (doi:10.1074/jbc.M110.190470).
Lee S, Chun JN, Kim SH, So I & Jeon JH 2013 Icilin inhibits E2F1-mediated cell cycle regulatory programs in prostate cancer. Biochemical and Biophysical Research Communications 441 1005–1010. (doi:10.1016/j.bbrc.2013.11.015).
Liang L, Morar N, Dixon AL, Lathrop GM, Abecasis GR, Moffatt MF & Cookson WO 2013 A cross-platform analysis of 14,177 expression quantitative trait loci derived from lymphoblastoid cell lines. Genome Research 23 716–726. (doi:10.1101/gr.142521.112).
Lichner Z, Fendler A, Saleh C, Nasser AN, Boles D, Al-Haddad S, Kupchak P, Dharsee M, Nuin PS & Evans KR et al. 2013 MicroRNA signature helps distinguish early from late biochemical failure in prostate cancer. Clinical Chemistry 59 1595–1603. (doi:10.1373/clinchem.2013.205450).
Liu J, Tang X, Li M, Lu C, Shi J, Zhou L, Yuan Q & Yang M 2013 Functional MDM4 rs4245739 genetic variant, alone and in combination with P53 Arg72Pro polymorphism, contributes to breast cancer susceptibility. Breast Cancer Research and Treatment 140 151–157. (doi:10.1007/s10549-013-2615-x).
Mancini F, Di Conza G, Pellegrino M, Rinaldo C, Prodosmo A, Giglio S, D'Agnano I, Florenzano F, Felicioni L & Buttitta F et al. 2009 MDM4 (MDMX) localizes at the mitochondria and facilitates the p53-mediated intrinsic-apoptotic pathway. EMBO Journal 28 1926–1939. (doi:10.1038/emboj.2009.154).
Mandke P, Wyatt N, Fraser J, Bates B, Berberich SJ & Markey MP 2012 MicroRNA-34a modulates MDM4 expression via a target site in the open reading frame. PLoS ONE 7 e42034. (doi:10.1371/journal.pone.0042034).
Maugeri-Sacca M, Coppola V, De Maria R & Bonci D 2013 Functional role of microRNAs in prostate cancer and therapeutic opportunities. Critical Reviews in Oncogenesis 18 303–315. (doi:10.1615/CritRevOncog.2013007206).
Miller KR, Kelley K, Tuttle R & Berberich SJ 2010 HdmX overexpression inhibits oncogene induced cellular senescence. Cell Cycle 9 3376–3382. (doi:10.4161/cc.9.16.12779).
Mishra S, Deng JJ, Gowda PS, Rao MK, Lin CL, Chen CL, Huang T & Sun LZ 2014 Androgen receptor and microRNA-21 axis downregulates transforming growth factor beta receptor II (TGFBR2) expression in prostate cancer. Oncogene 33 4097–4106. (doi:10.1038/onc.2013.374).
Nanni S, Priolo C, Grasselli A, D'Eletto M, Merola R, Moretti F, Gallucci M, De Carli P, Sentinelli S & Cianciulli AM et al. 2006 Epithelial-restricted gene profile of primary cultures from human prostate tumors: a molecular approach to predict clinical behavior of prostate cancer. Molecular Cancer Research 4 79–92. (doi:10.1158/1541-7786.MCR-05-0098).
Peng X, Li W, Yuan L, Mehta RG, Kopelovich L & McCormick DL 2013 Inhibition of proliferation and induction of autophagy by atorvastatin in PC3 prostate cancer cells correlate with downregulation of Bcl2 and upregulation of miR-182 and p21. PLoS ONE 8 e70442. (doi:10.1371/journal.pone.0070442).
Pinho FG, Frampton AE, Nunes J, Krell J, Alshaker H, Jacob J, Pellegrino L, Roca-Alonso L, de Giorgio A & Harding V et al. 2013 Downregulation of microRNA-515-5p by the estrogen receptor modulates sphingosine kinase 1 and breast cancer cell proliferation. Cancer Research 73 5936–5948. (doi:10.1158/0008-5472.CAN-13-0158).
Ramos YF, Stad R, Attema J, Peltenburg LT, van der Eb AJ & Jochemsen AG 2001 Aberrant expression of HDMX proteins in tumor cells correlates with wild-type p53. Cancer Research 61 1839–1842.
Schaefer A, Jung M, Mollenkopf HJ, Wagner I, Stephan C, Jentzmik F, Miller K, Lein M, Kristiansen G & Jung K 2010 Diagnostic and prognostic implications of microRNA profiling in prostate carcinoma. International Journal of Cancer 126 1166–1176. (doi:10.1002/ijc.24827).
Selth LA, Townley S, Gillis JL, Ochnik AM, Murti K, Macfarlane RJ, Chi KN, Marshall VR, Tilley WD & Butler LM 2012 Discovery of circulating microRNAs associated with human prostate cancer using a mouse model of disease. International Journal of Cancer 131 652–661. (doi:10.1002/ijc.26405).
Siegel R, Ma J, Zou Z & Jemal A 2014 Cancer statistics, 2014. CA: A Cancer Journal for Clinicians 64 9–29. (doi:10.3322/caac.21208).
Simone CB II, John-Aryankalayil M, Palayoor ST, Makinde AY, Cerna D, Falduto MT, Magnuson SR & Coleman CN 2013 mRNA expression profiles for prostate cancer following fractionated irradiation are influenced by p53 status. Translational Oncology 6 573–585. (doi:10.1593/tlo.13241).
Strachan GD, Jordan-Sciutto KL, Rallapalli R, Tuan RS & Hall DJ 2003 The E2F-1 transcription factor is negatively regulated by its interaction with the MDMX protein. Journal of Cellular Biochemistry 88 557–568. (doi:10.1002/jcb.10318).
Stranger BE, Montgomery SB, Dimas AS, Parts L, Stegle O, Ingle CE, Sekowska M, Smith GD, Evans D & Gutierrez-Arcelus M et al. 2012 Patterns of cis regulatory variation in diverse human populations. PLoS Genetics 8 e1002639. (doi:10.1371/journal.pgen.1002639).
Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, Arora VK, Kaushik P, Cerami E & Reva B et al. 2010 Integrative genomic profiling of human prostate cancer. Cancer Cell 18 11–22. (doi:10.1016/j.ccr.2010.05.026).
Thomas LF, Saito T & Saetrom P 2011 Inferring causative variants in microRNA target sites. Nucleic Acids Research 39 e109. (doi:10.1093/nar/gkr414).
Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C & Ferracin M et al. 2006 A microRNA expression signature of human solid tumors defines cancer gene targets. PNAS 103 2257–2261. (doi:10.1073/pnas.0510565103).
Wach S, Nolte E, Szczyrba J, Stohr R, Hartmann A, Orntoft T, Dyrskjot L, Eltze E, Wieland W & Keck B et al. 2012 MicroRNA profiles of prostate carcinoma detected by multiplatform microRNA screening. International Journal of Cancer 130 611–621. (doi:10.1002/ijc.26064).
Wade M, Li YC & Wahl GM 2013 MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nature Reviews. Cancer 13 83–96. (doi:10.1038/nrc3430).
Wang Y, Zhang YX, Kong CZ, Zhang Z & Zhu YY 2013 Loss of P53 facilitates invasion and metastasis of prostate cancer cells. Molecular and Cellular Biochemistry 384 121–127. (doi:10.1007/s11010-013-1789-1).
Wynendaele J, Bohnke A, Leucci E, Nielsen SJ, Lambertz I, Hammer S, Sbrzesny N, Kubitza D, Wolf A & Gradhand E et al. 2010 An illegitimate microRNA target site within the 3′ UTR of MDM4 affects ovarian cancer progression and chemosensitivity. Cancer Research 70 9641–9649. (doi:10.1158/0008-5472.CAN-10-0527).
Xi Y, Formentini A, Chien M, Weir DB, Russo JJ, Ju J, Kornmann M & Ju J 2006 Prognostic values of microRNAs in colorectal cancer. Biomarker Insights 2 113–121.
Yang J, Gao W, Song NH, Wang W, Zhang JX, Lu P, Hua LX & Gu M 2012 The risks, degree of malignancy and clinical progression of prostate cancer associated with the MDM2 T309G polymorphism: a meta-analysis. Asian Journal of Andrology 14 726–731. (doi:10.1038/aja.2012.65).
Zhou L, Zhang X, Li Z, Zhou C, Li M, Tang X, Lu C, Li H, Yuan Q & Yang M 2013 Association of a genetic variation in a miR-191 binding site in MDM4 with risk of esophageal squamous cell carcinoma. PLoS ONE 8 e64331. (doi:10.1371/journal.pone.0064331).