Abstract
Although prostate cancer (CaP) is the most frequently diagnosed malignant tumor in American men, the mechanisms underlying the development and progression of CaP remain largely unknown. Recent studies have shown that downregulation of the microRNA miR-124 occurs in several types of human cancer, suggesting a tumor suppressive function of miR-124. Until now, however, it has been unclear whether miR-124 is associated with CaP. In the present study, we completed a series of experiments to understand the functional role of miR-124 in CaP. We detected the expression level of miR-124 in clinical CaP tissues, evaluated the influence of miR-124 on the growth of CaP cells and investigated the mechanism underlying the dysregulation of miR-124. We found that (i) miR-124 directly targets the androgen receptor (AR) and subsequently induces an upregulation of p53; (ii) miR-124 is significantly downregulated in malignant prostatic cells compared to benign cells, and DNA methylation causes the reduced expression of miR-124; and (iii) miR-124 can inhibit the growth of CaP cells in vitro and in vivo. Data from this study revealed that loss of miR-124 expression is a common event in CaP, which may contribute to the pathogenesis of CaP. Our studies also suggest that miR-124 is a potential tumor suppressive gene in CaP, and restoration of miR-124 expression may represent a novel strategy for CaP therapy.
Similar content being viewed by others
Introduction
Prostate cancer (CaP) is the most frequently diagnosed malignant tumor and was the second leading cause of cancer-related death in American men in 2011.1 Metastatic CaP can be treated effectively with androgen ablation. However, one of the most troubling aspects of this disease is that after hormone treatment, the tumor inevitably progresses from an androgen-dependent to an incurable castration-resistant (CR) form.2 The mechanism underlying the progression has been poorly understood. In the past decades, therefore, an important task in CaP research has been to elucidate the molecular alteration occurring in the development of CR tumors. Considerable insight into the progression of CaP has been recently achieved, including the discovery of aberrantly expressed microRNAs (miRNAs).
The human genome may encode over 1000 miRNAs, which negatively regulate ∼60% of human genes.3, 4 Thus, miRNAs are involved in almost all important cellular processes, not only in physiological conditions but also in diseases including cancer.5 Indeed, a number of cancer-related miRNAs have been identified recently. Some miRNAs have also been reported to be aberrantly expressed in human CaP.6 These miRNAs, which act as tumor suppressor genes (TSGs) or oncogenes, contribute to the pathogenesis of CaP by directly targeting some proliferation-related genes or apoptotic molecules.7 In spite of these exciting findings, the role of miRNAs in the development and progression of CaP has been largely unexplored. Thus, identifying CaP-associated miRNAs and investigating their roles in CaP will help us understand the mechanisms related to the development and progression of this disease.
MiRNA-124 (miR-124) is a highly conserved miRNA whose in vivo function is poorly defined. This small non-coding RNA was first reported to be highly expressed in neuronal cells.8 Recent studies revealed that miR-124 was significantly downregulated in several types of human cancers.9, 10, 11, 12 Moreover, it regulates some proliferation-related genes such as cyclin-dependent kinase 6,13 forkhead box A214 and solute carrier family 16, member 1 (SLC16A1).13 Thus, miR-124 is classified as a tumor suppressor miRNA.13 Until now, however, the role of miR-124 in CaP has been totally unknown, although it was reported previously to be undetectable in 22Rv1 CaP cells.15
In the present study, we aimed to explore the role of miR-124 in CaP. We found that miR-124 directly targets the androgen receptor (AR). We also detected the expression level of miR-124 in clinical CaP tissues and found that a majority of these CaP tissues express low levels of miR-124. In addition, we observed that synthetic miR-124 mimics inhibit the proliferation of CaP cells. As the AR has a crucial role in the pathogenesis of CaP, our studies established a link between dysregulated miR-124, overexpressed AR and CaP cell proliferation. Our results imply that miR-124 is a potential TSG and downregulation of miR-124 contributes to the development and progression of CaP.
Results
MiR-124 directly targets the AR and inhibits proliferation of CaP cells
Overexpression of the AR is well known to have an important role in the pathogenesis of CaP. We are very interested in determining whether certain miRNA(s) contribute to upregulation of the AR. From two algorithms, TargetScan (Release 5.2) and miRanda (August 2010 Release), that predict miRNA-binding sites in the first 436 base of the AR 3′ UTR (NM_000044.2), we identified three broadly conserved miRNA-binding sites for six miRNAs (miR-130/miR-301, miR-124/miR-506 and miR-30/miR-384). In order to assess the ability of these miRNAs to regulate AR expression, AR-positive C4–2B CaP cells cultured in androgen-deprived medium were separately treated with four chemically modified miRNA mimics (miR-124, miR-130, miR-384 or miR-506, purchased from Ambion, Austin, TX, USA). As shown in Figure 1a, compared with miRNA-negative control (miR-NC), treatment with miR-124 mimic resulted in a reduction of AR protein by ∼70%, while the other three miRNA mimics induced 20–40% decrease in AR expression, suggesting a potent downregulation of the AR by miR-124. To determine the influence of these miRNAs on proliferation of CaP cell, AR-positive CaP cell lines (LNCaP, C4–2B and 22Rv1) were transiently transfected with each of these miRNA mimics. Consistent with its effect on AR level, transfection of miR-124 mimic inhibited proliferation to a greater extent than the other miRNA mimics tested (Supplementary Figure 1). We thus focused on miR-124 in this study.
To determine whether miR-124-mediated downregulation of the AR affects the AR activity, both AR-positive LNCaP and C4–2B were treated with miR-124 mimic. Western blot analysis demonstrated that miR-124 induced downregulation of the AR, which was concomitant with a reduced PSA level (Figure 1b). As AR regulates miR-125b,16 we used C4–2B and cds2 cells that express increased miR-125b16 to examine the effect of miR-124 mimic on endogenous miR-125b. We found that miR-124 induced downregulation of miR-125b by 30% in C4–2B and 54% in cds2 (Figure 1c). To verify that the putative miR-124 binding site in the 3′ UTR of AR mRNA is responsible for regulation by miR-124, the 3′ UTR was cloned into a luciferase reporter vector and then cotransfected with miR-124 mimic into C4–2B cells. Luciferase activity was measured 2 days after the transfection. As shown in Figure 1d, transfection of miR-124 mimic resulted in 40% reduction of the enzyme activity, indicating a direct interaction between miR-124 and AR mRNA. Taken together, these data indicate that miR-124 targets the AR and affects its activity.
MiR-124 is significantly downregulated in CaP cells
We determined the expression level of miR-124 in CaP cells. The abundance of miR-124 was first examined in seven prostate cell lines (two benign and five malignant) using quantitative PCR (qPCR). We found a reduced expression of miR-124 in the malignant cell lines compared with the benign cell lines (Figure 2a). We also performed northern blot analysis of these cell lines and similar results were obtained (Supplementary Figure 2A). Next, we tested whether miR-124 is downregulated in clinical prostate samples. We reviewed the miRNA expression profiling of our initial clinical tissue specimens, and found that the abundance of miR-124 is markedly reduced in four CaP samples relative to that in one benign prostatic hyperplasia (BPH) tissue (Supplementary Figure 2B). We then determined whether downregulation of miR-124 is common in clinical CaP tissues. The levels of miR-124 were detected in 79 clinical prostatic tissues, including 19 BPHs, 44 primary CaPs, 6 lymph node metastases and 10 CR tumors. Although miR-124 abundances have overlap in a small number of benign and primary tumor tissues, the average miR-124 level was 2.7-fold less in primary CaPs than in BPHs (39 in CaPs vs 146 in BPHs, P<0.01) (Figure 2b). Interestingly, 10 CR tumors and 6 metastases expressed extremely low levels of miR-124. Compared with the primary CaP samples, the average miR-124 level decreases by 77% in these advanced tumors (39 in CaPs vs 9 in CR and metastatic tumors, P<0.05) (Figure 2b), suggesting a potential involvement of miR-124 in CaP progression. To confirm the downregulation of miR-124, 18 CaP samples and matched benign adjacent tissues were analyzed using qPCR. In comparison with matched benign tissues, 15 of 18 CaP samples (83%) expressed significantly reduced miR-124 (Figure 2c). In addition, we performed northern blot analysis in five matched prostate tissues in which sufficient quantities of RNA were available. As expected, the three CaPs (8, 11 and 16) having low miR-124 levels detected by qPCR exhibit lower signal intensity in northern blots (Supplementary Figure 2C). Taken together, these data provide strong evidence that miR-124 is significantly reduced in CaP cell lines and in a majority of clinical CaP samples, and also suggest that decreased expression of miR-124 is common in human CaP tissues.
MiR-124 and AR are negatively correlated in human CaP tissues
Having demonstrated that miR-124 targets the AR and is significantly downregulated in CaP cells, we therefore asked whether clinical CaP samples having a low level of miR-124 overexpressed the AR. To address the issue, eight additional matched pairs of CaP and BPH were examined for expression of AR protein using immunohistochemical analysis. In these samples, miR-124 level detected by qPCR was significantly decreased in CaP samples compared with that in BPH. Conversely, immunohistochemical results demonstrated that AR immunostaining was more intense in seven of eight CaP samples than that in BPH matches. These results reveal an inverse relationship between miR-124 and AR expression levels in prostatic tissues. Figure 3 shows the representative results from four matches. In CaP samples, AR protein staining is much higher than that in matched BPH tissues (Figure 3a), while the expression of miR-124 is significantly decreased in CaP tissue compared with that in BPH tissues (Figure 3b). Therefore, our data suggest that downregulation of miR-124 results in increased expression of the AR in clinical CaP tissues.
MiR-124 is methylated in CaP cells
The question remained: why is miR-124 downregulated in CaP cells? We first confirmed that androgens do not regulate miR-124, based on our observation that miR-124 levels were similar in androgen R1881-treated and untreated CaP cell lines (data not shown). A previous study has shown that DNA methylation regulates the expression of miRNAs.17 Moreover, computer analysis demonstrated that the miR-124–1 at 8p23.1 and the miR-124–3 at 20q13.33 are embedded within CpG islands and the miR-124–2 at 8q12.3 is 760 bp downstream of a CpG island.13 In order to determine that DNA methylation downregulates miR-124 in CaP cells, we performed a demethylation experiment in three androgen-independent cell lines (22Rv1, C4–2B and cds2). Cells were treated with 50 μM of 5′-Aza-2'-deoxycytidine (Aza) for 4 days and miR-124 was detected by qPCR. It was found that Aza treatment induced a 2- to 5-fold increase in the miR-124 level compared with that in the untreated cells (Figure 4a). As miR-124 targets the AR as shown in Figure 1, we tested whether Aza downregulates expression of the AR. It was found that treatment with Aza reduced the AR level by ∼40% in LNCaP and∼60% in C4–2B cells (Figure 4b). We then assessed the methylation status of two benign prostate cell lines (pRNS-1–1 and RWPE-1) and five CaP cell lines using methylation-specific PCR (MSP). The 5′-DNA fragments of three miR-124 genes were strongly amplified by MSP primers in all cell lines except RWPE-1 whose miR-124-2 fragment was not amplified by the MSP primers (Figure 4c). The fragments of miR-124-1 and miR-124-2 were also amplified weakly by unmethylated DNA primers in some of these cell lines (Figure 4c). MSP data suggest different extents of DNA methylation at the 5′ regions of miR-124 genes. In order to validate DNA methylation, the 5′-DNA fragments of three miR-124 genes in six cell lines were amplified using COBRA (combined bisulfite restriction analysis) primers, cloned and sequenced. It was found that 41–82% of CpG sites were methylated in miR-124-1, 17–57% in miR-124-2 and 15–31% in miR-124-3, in five CaP cell lines, and the CpG methylation are 18%, 13% and 12%, respectively, in benign RWPE-1 cells (Figure 4d).
We next determined the methylation status in 9 BPH tissues and 14 CaP tissues using COBRA. The 5′-DNA fragments of three miR-124 genes that contain two, five and six CGCG sites, respectively, were amplified with COBRA primers and digested with the restriction enzyme BstU1, which only cleaves methylated CGCG sites. It was found that BstU1 was able to cut the miR-124–1 PCR products in 13 of 14 (93%) CaP samples, miR-124–2 in 11 of 14 (79%) samples and miR-124–3 in 12 of 14 (86%) samples, while the BstU1-digested BPH DNA fragments of miR-124 genes are 11% (1/9), 22% (2/9) and 44% (4/9), respectively (Figure 4e), indicating that significant miR-124 methylation occurs in clinical CaP specimens. Taken together, our data suggest that DNA methylation occurring at the 5′ regions of miR-124 genes causes, at least in part, the reduced expression of miR-124 in CaP cells.
MiR-124 induces apoptosis
Previous studies demonstrated that the AR upregulates miR-125b, while miR-125b represses the expression of p5318 and facilitates the proliferation of CaP cells.16, 19 As miR-124 targets the AR and downregulates miR-125b, we asked whether increased miR-124 level increases apoptotic cell death. To address the issue, LNCaP and C4–2B cells were transfected with miR-124 mimic and then cultured for 4 days. Apoptotic dead cells were quantitatively detected by Annexin V binding assay. We observed that miR-124 mimic induced 10.4% of LNCaP cells and 9.7% of C4–2B cells to undergo apoptotic cell death (Figure 5a). The comparative figures for the miR-NC cells was 0.7% and 1.1%, respectively (P<0.01). A representative Annexin V assay is shown in Supplementary Figure 3, in which miR-124 mimic-induced apoptosis (upper-right and lower-right squares in quadrant gates) was 11.4% in LNCaP cells and 8.4% of C4–2B cells. To provide biochemical evidence for the occurrence of apoptosis, we determined whether miR-124 mimic increases the expression of p53 and caspase 3. As expected, treatment of C4–2B cells with miR-124 mimic induced an approximately ninefold-enhanced expression of p53 (Figure 5b) and an approximately twofold increase in caspase 3 (Figure 5c). These data indicate that miR-124, at least in part, induces apoptosis by activating the p53 signaling pathway.
MiR-124 inhibits xenograft growth of CaP cells
Having demonstrated that miR-124 inhibits proliferation of AR-positive CaP cells and induces apoptosis, we tested whether this miRNA exerts an inhibitory effect on xenograft growth of CaP cells in vivo. We used 22Rv1 CaP cells to address this question owing to their expression of low (Figure 2a) or undetectable15 level of miR-124, as well as their ability to efficiently form tumors in nude mice. Overexpression of miR-124 in 22Rv1 CaP cell lines was established using a lentiviral vector system that expresses 28-fold elevation of miR-124 relative to controls (data not shown). Xenograft tumors were generated by subcutaneously inoculating intact male nude mice with 22Rv1-lenti-miR-124 cells or 22Rv1-lenti-vector cells as a control. Tumors in both the miR-124 and control groups became palpable 18 days after inoculation, suggesting that miR-124 did not affect the onset of 22Rv1 tumors. However, overexpression of miR-124 significantly inhibited the growth of 22Rv1 tumors after 3 weeks when compared with controls (Figure 6a). At the end of the experiments, qPCR analysis of miR-124 levels was performed in three lenti-miR-124 tumors. It was found that miR-124 levels were increased 23-fold in the miR-124 tumors compared with the control tumors (Figure 6b), similar to that in cultured 22Rv1-lenti-miR-124 cells, revealing a stable expression of miR-124 in vivo. In addition, one lenti-vector tumor and two lenti-miR-124 tumors were examined by western blot analysis for their AR expression. As shown in Figure 6c, lenti-miR-124 tumors express moderate reduction of the full-length AR. Interestingly, overexpression of miR-124 in 22Rv1 cells induced a downregulation of the truncated AR that may contribute to the aggressive features of 22Rv1 cells.20 Collectively, our results suggest that miR-124 inhibits growth of 22Rv1 xenograft tumors by repressing AR expression.
Discussion
Although miR-124 has been reported to be involved in several other cancer types,9, 10, 11 its role in CaP is largely unknown. To date, only one study has reported that miR-124 was undetectable in 22Rv1 CaP cells.15 The present study explored the role of miR-124 in CaP. We found that miR-124 directly targets the AR, suggesting that it is a potential TSG in CaP. In addition, our expression analysis revealed that miR-124 was significantly downregulated in a majority of clinical CaP specimens tested, which suggests that dysregulation of miR-124 may be a common event in patients with CaP, and loss of miR-124 may contribute to the progression of CaP.
In this study, we address the potential mechanisms underlying miR-124 downregulation in CaP cells. As methylation of TSGs is very common in both early and advanced stages of CaP,21, 22, 23, 24 miR-124 acting as a potential TSG in CaP may be susceptible to epigenetic modification. Indeed, we observed DNA methylation of three miR-124 genes in CaP cell lines and clinical CaP samples, which provides an explanation for the reduction of miR-124 levels. This is supported by the fact that demethylation treatment with Aza increased the abundance of miR-124 in CaP cells. These data provide strong evidence that miR-124 is downregulated in CaP in part as a result of DNA methylation. Our results are consistent with those reported in other cancer types in which miR-124 loci are aberrantly methylated.9, 11, 13 However, methylation of miR-124 loci was not detected in a recent report.25 In that study, Rauhala et al.25 treated CaP cells with 1 μM of Aza for 2 days followed by 300 nM of trichostatin A (a histone deacetylase inhibitor) for 24 h They identified 38 methylated miRNAs but not miR-124. Mazar et al.26 reported that Aza upregulates miR-34b expression in a dose-dependent manner, and 1 μM of Aza did not induce elevation of miR-34b in melanoma cells. As up to 100 μM of Aza failed to induce cytotoxicity in CaP cell lines,27 we used 50 μM of Aza to treat CaP cells for 4 days, which induced significant elevation of miR-124. It is thus likely that our ability to detect the increased level of miR-124 is due to the higher concentration of Aza used. Besides the epigenetic regulation, the expression level of miR-124 can be affected by genetic factors, such as mutation and heterozygosity (LOH). Indeed, we detected a G to C point mutation (or single-nucleotide polymorphism) in the 5′ region of miR-124–2 (Supplementary Figure 4). Mutation (or single-nucleotide polymorphism) in the 5′ region can reduce expression of mature miRNA by altered miRNA processing.28, 29 In addition, a previous study revealed that tumor suppressive miRNAs are frequently located in LOH regions.30 We know that miR-124–1 is located at 8p23.1 and miR-124–2 at 8q12.3. LOH in these two regions were reported to occur in an ∼ 50% of CaPs.31, 32, 33 Thus, LOH may affect the levels of miR-124–1 and miR-124–2. In order to elucidate the mechanism underlying the downregulation of miR-124, further study is needed to detect the frequencies of DNA mutation (or single-nucleotide polymorphism) and LOH in the three miR-124 loci.
An interesting and clinically relevant discovery is that miR-124 directly targets the AR. The AR contributes to the initiation and progression of CaP. A study has demonstrated that a moderately altered AR expression is sufficient to convert androgen dependent CaP cells to a CR form.34 The AR has been reported to be upregulated in clinical CaPs, particularly those that are CR. Why AR is upregulated in CaP remains poorly understood. AR gene amplification appears to account for the AR upregulation in an ∼30% CR CaPs;35 however, it is unknown how the AR is upregulated in the other CR tumors. As the 3′ UTR of the AR mRNA contains a conserved miR-124 binding site, we performed several experiments to validate the regulation of the AR by miR-124. We also observed an inverse correlation between miR-124 abundance and the AR expression level in clinical CaP samples tested. Therefore, our study demonstrates that dysregulation of miR-124 may elevate the expression of the AR, which is a new mechanistic explanation for the overexpression of the AR in CaP. In this present study, identification of miR-124 as an AR-targeting miRNA was based on the TargetScan and miRanda prediction programs. We noted that miR-124, as well as the other three predicted miRNAs as shown in Figure 1a, were not selected as AR-targeting miRNAs in a recent study.36 In that study, the authors transfected five AR-positive CaP cell lines with 20 nM of human miRNA library, and selected 71 miRNA candidates that affect the level of AR in all the five cell lines. However, miRNA repression of target molecular expression is dose dependent.37 The dose of miR-124 mimic used in this study is 100 nM. In addition, miRNA functions in a cell type- and context-dependent manner,38 and some AR-targeting miRNAs may not equally affect the AR level in all the five CaP cell lines. Therefore, different treatment conditions and selection criteria may account for the differential results.
Although we focus on the AR in this study, we realize that miR-124 targets hundreds of human genes, and other potential targets may also be relevant for the tumor suppressive function of miR-124 in CaP. We have identified a putative miR-124 binding site in the 3′ UTR of high-mobility group AT-hook 1 gene (HMGA1). In a pilot study, transfection of C4–2B cells with miR-124 mimic resulted in the reduction of HMGA1 expression by 60%; furthermore, two clinical CaP specimens having low level of miR-124 did overexpress cytoplasmic HMGA1 (see Supplementary Figure 5), suggesting a link between low miR-124 and overexpression of HMGA1 in CaP. Previous studies revealed that HMGA1 was highly expressed in advanced CaPs,39, 40 and cytoplasmic HMGA1 directly interacts with p53, leading to p53 inactivation.41, 42 Therefore, functional characterization of this miR-124 target will provide new insight into elucidating the molecular alteration occurring in advanced CaP. In addition, cyclin-dependent kinase 6 and forkhead box A2 have been identified as promising targets of miR-124.13, 14 Both cyclin-dependent kinase 6 and Foxa2 were previously reported to be elevated in CaP cells and correlate with tumor grade.43, 44, 45, 46 Moreover, these two proteins interact with the AR and enhance the AR activity.47, 48 Further studies are required to understand whether upregulation of these two molecules in CaP cells are the result of decreased expression of miR-124.
Consistent with a previous report in nerve cells,49 restoration of miR-124 level by transfecting CaP cells with synthetic miR-124 induces increased apoptosis. We also found that treatment of CaP cells with miR-124 upregulated the expression of p53, which is due at least partially to miR-124-mediated inhibition of AR/miR-125b signaling.16 Thus, upregulation of p53 signaling may have a key role in miR-124-induced apoptosis. As miR-124 induces apoptosis, we evaluated its effect on proliferation of CaP cells. Restoration of miR-124 level significantly inhibited the growth of CaP cells. More interestingly, when lentiviral-transduced 22Rv1 CaP cells that stably express miR-124 were injected subcutaneously into intact male nude mice, xenograft tumor growth was also significantly inhibited. Therefore, further testing of miR-124 in pre-clinical models of CaP will help define its ultimate therapeutic potential for the treatment of CaP.
In conclusion, this study has shown that loss of miR-124 expression may be a common event in CaP. As miR-124 negatively regulates the AR, downregulation of miR-124, as found in CaP, increases expression of the receptor. Therefore, deregulation of miR-124 as well as other AR-targeting miRNA candidates36 contributes to high levels of AR expression in clinical CaP. In addition, our data suggest that downregulation of miR-124 may be involved in the pathogenesis of CaP. Our evidence supporting the ability of miR-124 to induce apoptosis in CaP cells encourages the hope that restoring miR-124 function in CaP cells, either by itself or in conjunction with other therapies, will offer improved survival, which will be through delayed development or treatment of CR CaP.
Materials and methods
Cell lines
Two immortalized benign prostatic epithelial cell lines (RWPE-1 and pRNS-1–1) and five CaP cell lines (LNCaP, C4–2B, cds2, 22Rv1 and LAPC-4) were used in this study. RWPE-1 (provided by Dr Mukta Webber, Michigan State University, East Lansing, MI, USA) and pRNS-1–1 (provided by Dr Johng Rhim, University of the Health Sciences, Bethesda, MD, USA) were maintained in keratinocyte serum-free medium supplemented with 50 mg/ml bovine pituitary extract and 5 ng/ml epidermal growth factor. CaP cell lines were maintained in RPMI 1640 medium containing 10% fetal bovine serum. Both C4–2B and cds2 cell lines are derivatives of LNCaP.50, 51
Clinical samples
Collection and use of CaP patient specimens were approved by the Institutional Review Board (UCD IRB#: 200312072–6). Primary CaP samples from radical prostatectomy, metastatic pelvic lymph nodes, CR tumor specimens and BPH tissues were obtained fresh from surgical excision by the Department of Urology, University of California, Davis, Sacramento, USA. These CR tumors were described in our previous publication:52 six from stage D patients and four from patients receiving combined androgen blockade therapy before radical prostatectomy. To ensure that portions of CaP tissues were enriched for tumor cells, a surgeon and a pathologist together sectioned the excised tissue, and a fresh piece was taken from the palpated cancer and rapidly frozen to −80 °C until use. A permanent section was then cut from the immediately adjacent area for hematoxylin and eosin staining. Based upon a histological review of hematoxylin and eosin section, it was confirmed that these CaP samples used in this study were tumor cell enriched.
Reporter plasmid construction and luciferase assay
To construct reporter plasmids, a 0.45-kb DNA fragment of the AR 3′ UTR containing the putative miR-124 binding site was prepared by PCR. The AR 3′ UTR fragment lacking the miR-124 binding site was used as negative control. DNA fragments were cloned into the pMIR-REPORT luciferase vector (Ambion) downstream of the reporter gene. The sequences and cloning direction of these PCR products were validated by DNA sequencing. For luciferase assay, cells (4 × 104 per well) were seeded into 24-well plates and cultured for 24 h. The cells were then cotransfected with reporter plasmids and 100 nM chemically-modified miR-124 mimic or miRNA-negative control (miR-NC). The pRL-SV40 Renilla luciferase plasmid (Promega, Madison, WI, USA) was used as an internal control. Two days later, cells were harvested and lysed with passive lysis buffer (Promega). Luciferase activity was measured using a dual luciferase reporter assay (Promega). Luciferase activity was normalized by Renilla luciferase activity.
Western blot analysis
Cells were grown to 70–80% confluence and lysed in lysis buffer consisting of 25 mmol/l Tris–HCl (pH 7.5), 150 mmol/l NaCl, 1 mmol/l EDTA, 1% Triton X-100, 0.1% SDS and 1 × proteinase inhibitor cocktail, and equal concentrations of denatured protein samples were loaded on a 10% SDS–polyacrylamide gel. After electrophoresis, proteins were transferred to an Immobilon PVDF membrane (Millipore, Bedford, MA, USA) and immunoblot analysis was conducted by the standard method.
Quantitative–PCR
For quantitative expression analysis of miRNA, total RNA was isolated from fresh-frozen prostatic tissues, cultured cells or xenograft tumors using TRIzol reagent (Life Technologies, Inc., Carlsbad, CA, USA). The miRNA level was measured using a TaqMan miRNA assay kit (Applied Biosystems, Foster City, CA, USA) following the manufacturer’s instruction and PCR amplification was carried out in a 7900HT Fast Real-time PCR system (Applied Biosystems). Real-time PCR was performed in triplicate for each sample. The abundance of miRNA was measured using Ct (threshold cycle) following the approach as described previously.53 ΔCt was calculated by subtracting the Ct of U6 RNA from the Ct of the miRNA tested. ΔΔCt was calculated by subtracting the ΔCt of a reference sample from the ΔCt of each sample. For clinical tissues, the reference sample is one CaP specimen tested that expresses the lowest level of miRNA. Fold change was generated using the equation .
Methylation assay
Genomic DNA was isolated from prostate cell lines using standard procedures and from CaP cell-enriched tumor tissues following our previous method.54 Bisulfite modification of DNA (1.0 μg) was carried out by using an EZ DNA methylation-direct kit (Zymo Research, Irvine, CA, USA). For the prostate cell lines, the methylation of 5′-CpG island regions was detected by MSP using the primers specific for either methylated or unmethylated DNA. The methylated CpG dinucleotides were validated using bisulfite DNA sequencing. Amplified CpG islands were separately cloned into pCR 2.1 vector using TOPO TA Cloning Kit (Invitrogen, Carlsbad, CA, USA), and at least three clones per PCR product were picked up for DNA sequencing. In clinical prostate tissues, the methylation status was assessed by COBRA assay. COBRA primer-amplified PCR products were purified using Amicon 30 filter (Millipore, Billerica, MA, USA), digested by BstUI and separated on a 3% agarose gel (1% agarose plus 2% NuSieve GTG low-melting temperature agarose). The primers used for MSP and COBRA were designed based on “The Li Lab” program (http://www.urogene.org/methprimer/index1.html). The primer sequences and amplified size are listed in Supplementary Table 1.
Annexin V binding assay
MiR-124-induced apoptosis was analyzed using a FACS Annexin V assay kit (Trevigen, Inc, Gaithersburg, MD, USA) following the protocol provided by the manufacturer. Briefly, cells were transfected with miR-124 mimic and incubated for 4 days. The cells were harvested, washed once with PBS, re-suspended in 100 μl of 1 × Annexin V binding buffer containing 1 μl of Annexin V-FITC conjugate and 10 μl of propidium iodide solution, and then incubated for 30 min at room temperature in the dark. Subsequently, 400 μl 1 × Annexin V binding buffer was added and samples were analyzed on a FACScan flow cytometer. Data analysis was performed using FACScan software (Becton Dickinson, Franklin Lakes, NJ, USA).
Animal experiment
A lentiviral miR-124 expression vector that expresses an ∼500-base human pre-miR-124–1 and the empty lentiviral vector were purchased from System Biosciences (SBI, Mountain View, CA, USA). Pseudovirus production and cell transduction were performed following the manufacturer's protocol. The resulting 22Rv1-miR-124 and 22Rv1-vector cells were selected through fluorescence-activated cell sorting (FACS) and were maintained in the medium described above. Overexpression of miR-124 in infected 22Rv1 cells was confirmed by qPCR. Animal studies were performed in male athymic nu/nu mice (4- to 6-week-old; Harlan Laboratories, Indianapolis, IN, USA). Mice were injected subcutaneously with suspensions of 2 × 106 22Rv1-miR–124 cells or 22Rv1-vector cells in a mixture (1:1 vol/vol) of culture medium and Matrigel (Becton Dickinson). Tumor growth was monitored and tumor dimensions were measured twice per week. Tumor volumes were calculated according to the following formula: ½ (length × width × height). Animal studies were performed according to the protocols approved by the Animal Care and Use Committee at University of California, Davis, Sacremento, CA, USA.
Statistical analysis
Statistical analysis was performed using GraphPad Prism software 5.0 (GraphPad Software, La Jolla, CA, USA). Student's t-test was used to analyze the difference in miR-124 levels between different groups. Statistical significance was set up to P<0.05 in each test.
References
Siegel R, Ward E, Brawley O, Jemal A . Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 2012; 61: 212–236.
Sadar MD . Small molecule inhibitors targeting the "achilles' heel" of androgen receptor activity. Cancer Res 2011; 71: 1208–1213.
Lewis BP, Burge CB, Bartel DP . Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005; 120: 15–20.
Friedman RC, Farh KK, Burge CB, Bartel DP . Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 2009; 19: 92–105.
Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA 2006; 103: 2257–2261.
Shi XB, Tepper CG, White RW . MicroRNAs and prostate cancer. J Cell Mol Med 2008; 12: 1456–1465.
Shenouda SK, Alahari SK . MicroRNA function in cancer: oncogene or a tumor suppressor? Cancer Metastasis Rev 2009; 28: 369–378.
Makeyev EV, Zhang J, Carrasco MA, Maniatis T . The MicroRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Mol Cell 2007; 27: 435–448.
Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I, Inazawa J . miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 2010; 31: 766–776.
Ando T, Yoshida T, Enomoto S, Asada K, Tatematsu M, Ichinose M et al. DNA methylation of microRNA genes in gastric mucosae of gastric cancer patients: its possible involvement in the formation of epigenetic field defect. Int J Cancer 2009; 124: 2367–2374.
Lujambio A, Ropero S, Ballestar E, Fraga MF, Cerrato C, Setien F et al. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res 2007; 67: 1424–1429.
Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I, Inazawa J . miR-124 and miR-203 are epigenetically silenced tumor-suppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 31: 766–776.
Agirre X, Vilas-Zornoza A, Jimenez-Velasco A, Martin-Subero JI, Cordeu L, Garate L et al. Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia. Cancer Res 2009; 69: 4443–4453.
Baroukh N, Ravier MA, Loder MK, Hill EV, Bounacer A, Scharfmann R et al. MicroRNA-124a regulates Foxa2 expression and intracellular signaling in pancreatic beta-cell lines. J Biol Chem 2007; 282: 19575–19588.
Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 2008; 105: 10513–10518.
Shi XB, Xue L, Yang J, Ma AH, Zhao J, Xu M et al. An androgen-regulated miRNA suppresses Bak1 expression and induces androgen-independent growth of prostate cancer cells. Proc Natl Acad Sci USA 2007; 104: 19983–19988.
Sinkkonen L, Hugenschmidt T, Berninger P, Gaidatzis D, Mohn F, Artus-Revel CG et al. MicroRNAs control de novo DNA methylation through regulation of transcriptional repressors in mouse embryonic stem cells. Nat Struct Mol Biol 2008; 15: 259–267.
Le MT, Teh C, Shyh-Chang N, Xie H, Zhou B, Korzh V et al. MicroRNA-125b is a novel negative regulator of p53. Genes Dev 2009; 23: 862–876.
Shi XB, Xue L, Ma AH, Tepper CG, Kung HJ, White RW . miR-125b promotes growth of prostate cancer xenograft tumor through targeting pro-apoptotic genes. Prostate 2011; 71: 538–549.
Tepper CG, Boucher DL, Ryan PE, Ma AH, Xia L, Lee LF et al. Characterization of a novel androgen receptor mutation in a relapsed CWR22 prostate cancer xenograft and cell line. Cancer Res 2002; 62: 6606–6614.
Graff JR, Herman JG, Lapidus RG, Chopra H, Xu R, Jarrard DF et al. E-cadherin expression is silenced by DNA hypermethylation in human breast and prostate carcinomas. Cancer Res 1995; 55: 5195–5199.
Lou W, Krill D, Dhir R, Becich MJ, Dong JT, Frierson HF et al. Methylation of the CD44 metastasis suppressor gene in human prostate cancer. Cancer Res 1999; 59: 2329–2331.
Kang GH, Lee S, Lee HJ, Hwang KS . Aberrant CpG island hypermethylation of multiple genes in prostate cancer and prostatic intraepithelial neoplasia. J Pathol 2004; 202: 233–240.
Woodson K, Hanson J, Tangrea J . A survey of gene-specific methylation in human prostate cancer among black and white men. Cancer Lett 2004; 205: 181–188.
Rauhala HE, Jalava SE, Isotalo J, Bracken H, Lehmusvaara S, Tammela TL et al. miR-193b is an epigenetically regulated putative tumor suppressor in prostate cancer. Int J Cancer 2010; 127: 1363–1372.
Mazar J, Khaitan D, DeBlasio D, Zhong C, Govindarajan SS, Kopanathi S et al. Epigenetic regulation of microRNA genes and the role of miR-34b in cell invasion and motility in human melanoma. PloS One 2011; 6: e24922.
Walton TJ, Li G, Seth R, McArdle SE, Bishop MC, Rees RC . DNA demethylation and histone deacetylation inhibition co-operate to re-express estrogen receptor beta and induce apoptosis in prostate cancer cell-lines. Prostate 2008; 68: 210–222.
Davis BN, Hata A . Regulation of MicroRNA biogenesis: a miRiad of mechanisms. Cell Commun Signal 2009; 7: 18.
Jazdzewski K, Murray EL, Franssila K, Jarzab B, Schoenberg DR, de la Chapelle A . Common SNP in pre-miR-146a decreases mature miR expression and predisposes to papillary thyroid carcinoma. Proc Natl Acad Sci USA 2008; 105: 7269–7274.
Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA 2004; 101: 2999–3004.
Matsuyama H, Pan Y, Yoshihiro S, Kudren D, Naito K, Bergerheim US et al. Clinical significance of chromosome 8p, 10q, and 16q deletions in prostate cancer. Prostate 2003; 54: 103–111.
Perinchery G, Bukurov N, Nakajima K, Chang J, Hooda M, Oh BR et al. Loss of two new loci on chromosome 8 (8p23 and 8q12-13) in human prostate cancer. Int J Oncol 1999; 14: 495–500.
Chang BL, Liu W, Sun J, Dimitrov L, Li T, Turner AR et al. Integration of somatic deletion analysis of prostate cancers and germline linkage analysis of prostate cancer families reveals two small consensus regions for prostate cancer genes at 8p. Cancer Res 2007; 67: 4098–4103.
Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med 2004; 10: 33–39.
Koivisto P, Kononen J, Palmberg C, Tammela T, Hyytinen E, Isola J et al. Androgen receptor gene amplification: a possible molecular mechanism for androgen deprivation therapy failure in prostate cancer. Cancer Res 1997; 57: 314–319.
Ostling P, Leivonen SK, Aakula A, Kohonen P, Makela R, Hagman Z et al. Systematic analysis of microRNAs targeting the androgen receptor in prostate cancer cells. Cancer Res 2011; 71: 1956–1967.
Yamakuchi M, Ferlito M, Lowenstein CJ . miR-34a repression of SIRT1 regulates apoptosis. Proc Natl Acad Sci USA 2008; 105: 13421–13426.
Olive V, Jiang I, He L . mir-17-92, a cluster of miRNAs in the midst of the cancer network. Int J Biochem Cell Biol 2010; 42: 1348–1354.
Tamimi Y, van der Poel HG, Denyn MM, Umbas R, Karthaus HF, Debruyne FM et al. Increased expression of high mobility group protein I(Y) in high grade prostatic cancer determined by in situ hybridization. Cancer Res 1993; 53: 5512–5516.
Takaha N, Hawkins AL, Griffin CA, Isaacs WB, Coffey DS . High mobility group protein I(Y): a candidate architectural protein for chromosomal rearrangements in prostate cancer cells. Cancer Res 2002; 62: 647–651.
Frasca F, Rustighi A, Malaguarnera R, Altamura S, Vigneri P, Del Sal G et al. HMGA1 inhibits the function of p53 family members in thyroid cancer cells. Cancer Res 2006; 66: 2980–2989.
Pierantoni GM, Rinaldo C, Esposito F, Mottolese M, Soddu S, Fusco A . High Mobility Group A1 (HMGA1) proteins interact with p53 and inhibit its apoptotic activity. Cell Death Differ 2006; 13: 1554–1563.
Qi J, Nakayama K, Cardiff RD, Borowsky AD, Kaul K, Williams R et al. Siah2-dependent concerted activity of HIF and FoxA2 regulates formation of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell 2010; 18: 23–38.
Chen Y, Robles AI, Martinez LA, Liu F, Gimenez-Conti IB, Conti CJ . Expression of G1 cyclins, cyclin-dependent kinases, and cyclin-dependent kinase inhibitors in androgen-induced prostate proliferation in castrated rats. Cell Growth Differ 1996; 7: 1571–1578.
Mirosevich J, Gao N, Gupta A, Shappell SB, Jove R, Matusik RJ . Expression and role of Foxa proteins in prostate cancer. Prostate 2006; 66: 1013–1028.
Qi J, Nakayama K, Cardiff RD, Borowsky AD, Kaul K, Williams R et al. Siah2-dependent concerted activity of HIF and FoxA2 regulates formation of neuroendocrine phenotype and neuroendocrine prostate tumors. Cancer Cell 18: 23–38.
Lim JT, Mansukhani M, Weinstein IB . Cyclin-dependent kinase 6 associates with the androgen receptor and enhances its transcriptional activity in prostate cancer cells. Proc Natl Acad Sci USA 2005; 102: 5156–5161.
Yu X, Gupta A, Wang Y, Suzuki K, Mirosevich J, Orgebin-Crist MC et al. Foxa1 and Foxa2 interact with the androgen receptor to regulate prostate and epididymal genes differentially. Ann NY Acad Sci 2005; 1061: 77–93.
Cao X, Pfaff SL, Gage FH . A functional study of miR-124 in the developing neural tube. Genes Dev 2007; 21: 531–536.
Lin DL, Tarnowski CP, Zhang J, Dai J, Rohn E, Patel AH et al. Bone metastatic LNCaP-derivative C4-2B prostate cancer cell line mineralizes in vitro. Prostate 2001; 47: 212–221.
Shi XB, Ma AH, Tepper CG, Xia L, Gregg JP, Gandour-Edwards R et al. Molecular alterations associated with LNCaP cell progression to androgen independence. Prostate 2004; 60: 257–271.
de Vere White R, Meyers F, Chi SG, Chamberlain S, Siders D, Lee F et al. Human androgen receptor expression in prostate cancer following androgen ablation. Eur Urol 1997; 31: 1–6.
Schmittgen TD, Livak KJ . Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 2008; 3: 1101–1108.
Shi XB, Bodner SM, deVere White RW, Gumerlock PH . Identification of p53 mutations in archival prostate tumors. Sensitivity of an optimized single-strand conformational polymorphism (SSCP) assay. Diagn Mol Pathol 1996; 5: 271–278.
Acknowledgements
This work was supported in part by NIH 1RO1CA136597 (R deVere White), Department of Defense PCRP PC080488 (R deVere White) and The LANIE Foundation. We thank Dr Mukta Webber for providing the RWPE-1 cell line and Dr Johng Rhim for the pRNS-1–1 cell line. We are also grateful to Dr Melanie C Bradnam for her editorial assistance and Stephanie Soares for critically reading the manuscript. This work was supported by NCI grant (no. CA136597) and DOD grant (no. PC080488).
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Competing interests
The authors declare no conflict of interest.
Additional information
Supplementary Information accompanies the paper on the Oncogene website
Rights and permissions
This work is licensed under the Creative Commons Attribution-NonCommercial-Share Alike 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/
About this article
Cite this article
Shi, XB., Xue, L., Ma, AH. et al. Tumor suppressive miR-124 targets androgen receptor and inhibits proliferation of prostate cancer cells. Oncogene 32, 4130–4138 (2013). https://doi.org/10.1038/onc.2012.425
Received:
Revised:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/onc.2012.425