Mir-199A-3p Inhibits Aurora Kinase A and Attenuates Prostate Cancer Growth
Yi Qu,Xiang Huang,Zhiqing Li,Junyan Liu,Jinlin Wu,Dapeng Chen,Fengyan Zhao,Dezhi Mu
DOI: https://doi.org/10.1016/j.ajpath.2014.01.017
IF: 5.77
2014-01-01
American Journal Of Pathology
Abstract:Prostate cancer (PCa) is the most common solid tumor malignancy in men that severely influences quality of life. Surgery is most often the recommended treatment for PCa, but radical prostatectomy can cause significant urinary adverse effects. Therefore, finding effective biochemical treatments for PCa remains a necessity. Aurora kinase A has been shown to be involved in PCa progression, thus making it a good target for PCa therapy. miRNAs are important regulators of gene expression, with some miRNAs specifically involved in carcinogenesis. Therefore, herein, we identified miRNAs targeted to aurora kinase A and examined their effects on the growth of PCa. We used primary samples from PCa patients and PCa cell lines as research subjects. We demonstrate that miR-199a-3p is down-regulated in PCa tissues compared with normal prostate tissues, with the expression pattern inversely correlated with the expression pattern of aurora kinase A. We find that miR-199a-3p agomir inhibits aurora kinase A and attenuates xenograft tumor growth of PCa. Moreover, we demonstrate that down-regulation of miR-199a-3p results from enhancement of the methylation of miR-199a gene in PCa. Furthermore, we find that the expression level of miR-199a-3p is inversely correlated to tumor stage and Gleason score of PCa. Revealing novel mechanisms for oncogene inhibition by miRNA-mediated pathways offers new avenues for PCa treatment. Prostate cancer (PCa) is the most common solid tumor malignancy in men that severely influences quality of life. Surgery is most often the recommended treatment for PCa, but radical prostatectomy can cause significant urinary adverse effects. Therefore, finding effective biochemical treatments for PCa remains a necessity. Aurora kinase A has been shown to be involved in PCa progression, thus making it a good target for PCa therapy. miRNAs are important regulators of gene expression, with some miRNAs specifically involved in carcinogenesis. Therefore, herein, we identified miRNAs targeted to aurora kinase A and examined their effects on the growth of PCa. We used primary samples from PCa patients and PCa cell lines as research subjects. We demonstrate that miR-199a-3p is down-regulated in PCa tissues compared with normal prostate tissues, with the expression pattern inversely correlated with the expression pattern of aurora kinase A. We find that miR-199a-3p agomir inhibits aurora kinase A and attenuates xenograft tumor growth of PCa. Moreover, we demonstrate that down-regulation of miR-199a-3p results from enhancement of the methylation of miR-199a gene in PCa. Furthermore, we find that the expression level of miR-199a-3p is inversely correlated to tumor stage and Gleason score of PCa. Revealing novel mechanisms for oncogene inhibition by miRNA-mediated pathways offers new avenues for PCa treatment. Prostate cancer (PCa) is the most common solid tumor malignancy in men that severely influences quality of life. However, the treatment for PCa remains challenging. Surgery is most often the recommended treatment for young men, but radical prostatectomy can cause significant urinary adverse effects. Therefore, finding effective biochemical treatments for PCa remains a necessity. Recently, aurora kinase A has been demonstrated to be involved in PCa progression and enhance cancer growth and migration.1Bar-Shira A. Pinthus J.H. Rozovsky U. Goldstein M. Sellers W.R. Yaron Y. Eshhar Z. Orr-Urtreger A. Multiple genes in human 20q13 chromosomal region are involved in an advanced prostate cancer xenograft.Cancer Res. 2002; 62: 6803-6807PubMed Google Scholar, 2Lee E.C. Frolov A. Li R. Ayala G. Greenberg N.M. Targeting Aurora kinases for the treatment of prostate cancer.Cancer Res. 2006; 66: 4996-5002Crossref PubMed Scopus (148) Google Scholar Furthermore, our previous work demonstrated that aurora kinase A up-regulation was closely associated with malignant phenotypes in PCa, suggesting that aurora kinase A may be a good target for PCa therapy.3Qu Y. Zhang L. Mao M. Zhao F. Huang X. Yang C. Xiong Y. Mu D. Effects of DNAzymes targeting Aurora kinase A on the growth of human prostate cancer.Cancer Gene Ther. 2008; 15: 517-525Crossref PubMed Scopus (23) Google Scholar To inhibit aurora kinase A and the subsequently induced malignant phenotypes, we need to clarify the mechanism by which aurora kinase A is up-regulated in PCa. In recent years, a new class of molecules has been found to regulate gene expression, known as miRNA.4Battel D.P. MicroRNAs: genomics, biogenesis, mechanism, and function.Cell. 2004; 116: 281-297Abstract Full Text Full Text PDF PubMed Scopus (29541) Google Scholar miRNAs are a family of small (approximately 22 nucleotides), endogenous, noncoding RNAs that, by binding complementary sequences in the 3′ untranslated region (3′-UTR) of mRNAs, either mediate translational repression or direct mRNA cleavage. Thus, the translation of these mRNAs is inhibited, or they are destabilized, resulting in down-regulation of the encoded protein.5Chua J.H. Armugam A. Jeyaseelan K. MicroRNAs: biogenesis, function and applications.Curr Opin Mol Ther. 2009; 11: 189-199PubMed Google Scholar A wide variety of miRNAs have been proved to contribute to malignant phenotypes in cancer.6Farazi T.A. Hoell J.I. Morozov P. Tuschl T. MicroRNAs in human cancer.Adv Exp Med Biol. 2013; 774: 1-20Crossref PubMed Scopus (93) Google Scholar Therefore, we hypothesize that regulation of some specific miRNAs could play a crucial role in the up-regulation of aurora kinase A in PCa. In the present study, we try to select and identify miRNAs targeted to aurora kinase A, and prove that these miRNAs regulate aurora kinase A in PCa. Meanwhile, because miRNAs are highly stable and compatible with tissues,7Sethupathy P. Collins F.S. MicroRNA target site polymorphisms and human disease.Trends Genet. 2008; 24: 489-497Abstract Full Text Full Text PDF PubMed Scopus (285) Google Scholar we explore the feasibility of using miRNAs to inhibit PCa growth both in vitro and in vivo. Surgical specimens used in this study were collected at The People's Hospital of Sichuan Province (Sichuan, China) from 2007 to 2011. Procedures were in accordance with the Declaration of Helsinki of 1975 and approved by the Ethics Committee of Human Experimentation of Sichuan University (Sichuan). Specimens were selected from the prostate tissue bank. Blocks were manually trimmed to further enrich the histological features of interest. Sections (8 μm thick) were prepared from each block. The first and last sections for each block were subjected to H&E staining and used to calculate epithelium percentage. The tumor content in the tumor specimens was >80%, whereas normal samples had at least 60% epithelium content and no evidence of tumor present. We acquired 12 cases of paired benign and tumor prostate tissues from patients with a Gleason score of ≥7. In these cases, matched normal and tumor samples from the same patients were available. We also acquired unpaired 50 PCa tissues and 20 normal prostate tissues. Human PCa cell lines PC-3, DU145, and LNCaP were purchased from ATCC (Manassas, VA), and cultured in RPMI 1640 medium, supplemented with 10% fetal bovine serum, 1% l-glutamine, and 1% penicillin-streptomycin. All cell cultures were incubated at 37°C in 5% CO2 and 95% air. For cell transfection, DU145 cells were seeded at a density of 1.0 × 105 (24-well plates). The dosage of miRNA agomir or antagomir (RiboBio, Guangzhou, China) used was determined according to the manufacturer's protocol. miR-199a-3p agomir (4 nmol/L, final concentration) or miR-199a-3p antagomir (20 nmol/L, final concentration) in 50 μL of Opti-MEM was complexed with 1 μL of transfection reagent (Lipofectamine) in 50 μL of Opti-MEM (Ambion, Austin, TX). The cells were transfected with these complexes and maintained for 8 hours, then fed with original medium. In all experiments, an equal concentration of nontargeting controls (agomir- or antagomir-negative controls) was used as controls for non–sequence-specific effects, whereas Lipofectamine was used as the vehicle control. At 24 and 48 hours after transfection, the cells were collected for detection. Total RNA was extracted from both PCa cells cultured in vitro and prostate tissues using an miRNeasy Mini Kit (Qiagen, Hilden, Germany), and from formalin-fixed, paraffin-embedded (FFPE) tissues using an miRNeasy FFPE Kit (Qiagen), according to the manufacturer's protocol. The cDNA was synthesized from 2 μg of total RNA with Moloney Murine Leukemia Virus (M-MLV) Reverse Transcriptase (Promega, Madison, WI) in a 25-μL volume [2 μg total RNA, 400 mmol/L reverse transcription primer (random primers for U6 rRNA- and miRNA-specific primers for miRNA), 4 U/μL M-MLV, and 0.4 mmol/L dNTP mix]. Real-time PCR was performed with SYBR Green I mix (Takara, Tokyo, Japan) in a 20-μL reaction volume (10 μL SYBR Green I mix, 200 mmol/L forward and reverse primer, and 2 μL cDNA template) on an MJ Opticon Monitor chromo4 instrument (Bio-Rad, Hercules, CA) using the following protocol: 95°C for 20 seconds and 40 cycles of 95°C for 10 seconds, 60°C for 20 seconds, and 70°C for 1 second. Data analysis was performed using the 2−ΔΔCT method.8Livak K.J. Schmittgen T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method.Methods. 2001; 25: 402-408Crossref PubMed Scopus (124098) Google Scholar The primers for miRNA were designed according to an miRNA primer designer (http://www.leonxie.com/miRNAprimerDesigner.php, last accessed March 11, 2014). The primers for hsa-miR-199a-3p were 5′-GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACTAACCA-3′ (reverse transcription), 5′-GCGGCGGACAGTAGTCTGCAC-3′ (forward), and 5′-ATCCAGTGCAGGGTCCGAGG-3′ (reverse). The primers for U6 rRNA were purchased from RiboBio. Proteins of cultured cells or prostate tissues were prepared by suspending cell pellets or grinding tissues in lysis buffer (50 mmol/L Tris-HCl, 150 mmol/L NaCl, 1% NP40, 0.5% deoxycholate, and 0.1% SDS), including proteinase inhibitors (1 mg/mL aprotinin, 1 mg/mL leupeptin, and 1 mmol/L phenylmethylsulphonyl fluoride). Proteins of FFPE tissues were extracted using a Qproteome FFPE Tissue Kit (Qiagen), according to the manufacturer's protocol. Protein concentration was measured using a DcProtein Assay Kit (Bio-Rad). Equal amounts of protein (40 mg) were loaded onto an SDS–polyacrylamide gel for electrophoresis and then transferred to a polyvinylidene fluoride membrane (Amersham, Uppsala, Sweden). The membrane was then incubated with primary antibody for 1 hour at room temperature. After washing with Tris-buffered saline with Tween 20, the membrane was incubated with corresponding horseradish peroxide–conjugated secondary antibody (Amersham) and the signals were visualized by electrochemiluminescence (Amersham). The primary antibodies used included anti-aurora A antibody to detect aurora kinase A (1:800; Santa Cruz Biotechnology, Santa Cruz, CA) and anti-active caspase-3 to detect phosphorylated caspase-3 (1:1000; Chemicon, Bedford, MA). An NIH image was used to measure the densities of the protein signals, and the protein levels were normalized to β-actin as an internal control. The miR-199a-3p binding site of aurora kinase A was amplified by PCR from genomic DNA of the human DU145 cell line, which was extracted with a QIAamp DNA Mini Kit (Qiagen). The PCR products were gel purified, digested, and inserted into the pMIR-RB-REPORT vector (Ribobio), and the resulting plasmid was labeled pWT. The miR-199a-3p binding site mutations (pMUT) were introduced using Multisite-Quickchange (Stratagene, San Diego, CA), according to the manufacturer's protocol, and cloned into the pMIR-RB-REPORT vector (Ribobio). All inserted or mutated sequences were confirmed by sequencing. HeLa cells were transfected with miRNA-mimics and pWT using Lipofectamine. Meanwhile, miRNA-mimics and pMUT, miRNA-control, and pWT, or miRNA-control and pMUT were also transfected in Hela cells. The cells were lysed 48 hours after the transfection for measurement of luciferase activity. A dual-luciferase assay was used to quantitate the effects of miR-199a-3p interaction with the 3′-UTR of aurora kinase A. In all experiments, transfection efficiencies were normalized to hLuc luciferase, which was constitutively expressed by the pMIR-RB-REPORT vector. To detect cell cycle distribution, cells were collected and washed with PBS, and then suspended in Krishan buffer (0.1% sodium citrate, 0.2 mg/mL RNAase, and 0.3% NP40) containing 0.05 mg/mL propidium iodide and then incubated at 4°C for 30 minutes. Cells were subjected to Coulter flow cytometry for analysis. Data were analyzed using ModFitLT 2.0 (Verity Software House, Topsham, ME). These studies were approved by the Animal Care and Ethics Committee of Sichuan University. In total, 30 five-week–old male BALB/C nude mice with an average weight of 20 g were housed in separate cages. DU145 cells were s.c. transplanted into each mouse. After 5 weeks, the tumor tissues had increased to a volume of approximately 65 mm3 and were ready to use for the following experiments. Nude mice were assigned to three groups, each consisting of 10 mice. The first group, serving as a control, received an equal volume (30 μL) of PBS injection, but no miRNA treatment. Each mouse in the second group was injected with 10 nmol/L of agomirs (dissolved in 30 μL PBS) into the tumor tissue every 2 days by multipoint intratumoral injection. Each mouse in the third group received 10 nmol/L of negative agomir control (dissolved in 30 μL PBS) in the same way as the other two groups. Tumor volumes were assessed using digital Vernier calipers every 2 days in a blinded way (Y.Q., D.M.), and each tumor volume (in mm3) was calculated by the following formula:V=0.4×D×d2(1) where V is volume, D is longitudinal diameter, and d is latitudinal diameter. Fifteen days later, mice in each group were sacrificed. The body weight of the mice and the wet weight of the tumors were measured. The tumor tissues were then collected to determine the expressions of aurora kinase A and active caspase-3 using Western blot analysis. DU145 cells were seeded at a density of 5 × 106/mL, and the DNA methylation inhibitor, 5-azacytidine (AZA; Merck, Bedford, MA), was added at a concentration of 1 μmol/L and incubated for 72 hours. Fresh medium was added every 24 hours, with or without AZA. Genomic DNA from culture cells was extracted with the QIAamp DNA mini kit (Qiagen) and from FFPE tissues with the QIAamp DNA FFPE Tissue Kit (Qiagen). The DNA (4 μg in a volume of 100 μL) was sheared 60 times with a 1-mL syringe, then 1 μg in a 50-μL volume was denatured by NaOH (final concentration, 0.27 mol/L) for 25 minutes at 37°C. Then, 30 μL of freshly prepared 10 mmol/L hydroquinone and 520 μL of 3 mol/L sodium bisulfite (containing 0.21 mol/L NaOH) were added, and the mixture was incubated under mineral oil at 50°C for 16 hours. Modified DNA was purified using the Axygen gel recovery kit, according to the manufacturer's instructions, and eluted into 50 μL. Modification was completed by NaOH (final concentration, 0.27 mol/L) treatment for 20 minutes at 37°C, followed by ethanol precipitation. The bisulfite-treated DNA was resuspended in 20 μL of water and used immediately or stored at −20°C. According to Cheung et al,9Cheung H.H. Davis A.J. Lee T.L. Pang A.L. Nagrani S. Rennert O.M. Chan W.Y. Methylation of an intronic region regulates miR-199a in testicular tumor malignancy.Oncogene. 2011; 30: 3404-3415Crossref PubMed Scopus (120) Google Scholar a hypermethylated region was mapped to the miR-199a gene and its upstream promoter (234 to 471), which leads to the down-regulation of miR-199a in testicular tumors. Therefore, we searched for CpG sites 400 bp upstream and 200 bp downstream from the miR-199a gene, and designed primers for methylation-specific PCR (MSP) using Methprimer (http://www.urogene.org/methprimer, last accessed March 11, 2014) (Table 1). The methylation status of this region was determined using an SYBR Green–based quantitative methylation-specific PCR (qMSP).Table 1Primers for MSP of hsa-miR-199aPrimer namePrimer sequencesProduct size (bp)Left M primer5′-AGGGAGGTTTTTTTTGAGGATC-3′134Right M primer5′-CCAATATACAAACTACTATACAACGAC-3′Left U primer5′-GGAGGTTTTTTTTGAGGATTG-3′132Right U primer5′-CCAATATACAAACTACTATACAACAAC-3′M, methylated; U, unmethylated. Open table in a new tab M, methylated; U, unmethylated. In brief, genomic DNA was extracted from cells or tissues, and modified by bisulfite, as previously described. For PCR (final volume, 20 μL), 0.5 μL of bisulfite-treated DNA template was mixed with 10 μL of 2× Power SYBR Green PCR Master Mix (Applied Biosystems, Foster, CA) plus primers to a final concentration of 200 nmol/L. The PCR conditions included an initial incubation at 50°C for 2 minutes, denaturing at 95°C for 10 minutes, and 40 cycles of denaturing at 95°C for 15 seconds and annealing at 60°C for 1 minute. After PCR amplification, a dissociation curve was generated to confirm the size of the PCR product. MSP reactions were optimized using methylated (methylated positive control) and unmethylated (unmethylated positive control) bisulfite-converted control DNA (Epitect control DNA set; Qiagen). Amplification products were quantified by counting CT values and visualized by UV illumination on a 2% agarose gel. The methylation percentage was estimated using the following formula:methylated(%)=1/1+2(−ΔCt)ΔCt=CtU−CtM,(2) where M is the copy number of methylated miR-199a, and U is the copy number of unmethylated miR-199a. Data are presented as means ± SEM from three independent experiments. A Student's t-test was used when comparing between two groups. An analysis of variance with Fisher's post hoc test was used when comparing more than two groups. The correlation coefficient was analyzed by Spearman's rank correlation. P < 0.05 was defined as the threshold for significance. To assess the miRNAs that can target aurora kinase A, a bioinformatic search was performed in the miRanda\miRDB\miRWalk\RNA22\targetscan databases (http://www.targetscan.org, last accessed March 11, 2014). The search yielded 407 miRNAs against aurora kinase A, as predicted by at least two databases. Among these miRNAs, let-7b/d/g, miR-23a/b, miR-92a/b, miR-199a-3p, and miR-24 were reported to be differentially expressed in miRNA profiles of PCa compared with noncancer prostate tissue10Porkka K.P. Pfeiffer M.J. Waltering K.K. Vessella R.L. Tammela T.L. Visakorpi T. MicroRNA expression profiling in prostate cancer.Cancer Res. 2007; 67: 6130-6135Crossref PubMed Scopus (795) Google Scholar, 11Szczyrba J. Löprich E. Wach S. Jung V. Unteregger G. Barth S. Grobholz R. Wieland W. Stöhr R. Hartmann A. Wullich B. Grässer F. The microRNA profile of prostate carcinoma obtained by deep sequencing.Mol Cancer Res. 2010; 8: 529-538Crossref PubMed Scopus (198) Google Scholar; therefore, we next examined the expression of these miRNAs in human PCa. To determine whether the levels of let-7b/d/g, miR-23a/b, miR-92a/b, miR-199a-3p, and miR-24 are down-regulated in clinical PCa, we used quantitative RT-PCR (RT-qPCR) to analyze RNAs from 12 paired benign and tumor human PCa specimens with a Gleason score of >7. RNAs were isolated from human tissues, reverse transcribed, and subjected to RT-qPCR. The levels of miR-199a-3p were significantly decreased in 9 of 12 tumors compared with their matched benign prostate tissues (Figure 1A), showing a much greater decrease than the other miRNAs (Table 2). Next, we examined the protein levels of aurora kinase A, the potential target of these miRNAs, via Western blot analysis. We found that aurora kinase A proteins were significantly up-regulated in most of the cancer tissues compared with their matched benign tissues (Figure 1B). Furthermore, the aurora kinase A protein levels were correlated inversely with the miR-199a-3p levels, with a correlation coefficient of 0.938 (Figure 1C). This was much higher than other miRNAs (Table 2), suggesting that aurora kinase A expression is regulated primarily by miR-199a-3p in human PCa. Thus, in this study, we chose to focus on miR-199a-3p.Table 2Expression of Aurora Kinase A and miRNAsGeneRelative expression of each caseR2RD123456789101112AuroraA2.21.81.01.91.92.41.11.61.91.02.11.9––let-7b0.71.10.61.00.50.50.71.20.50.61.20.50.0058/12let-7d0.71.00.61.10.50.60.71.10.60.61.20.50.0268/12let-7g0.71.10.71.10.60.50.71.10.50.71.10.60.0008/12miR-23a1.10.81.20.70.70.91.30.81.21.10.91.00.2437/12miR-23b1.00.91.20.90.81.01.10.90.91.00.90.80.3207/12miR-92a0.71.31.10.60.70.91.31.21.01.00.80.70.3196/12miR-92b0.71.21.10.80.70.91.21.21.10.80.90.80.1627/12miR-199a-3p0.30.51.00.50.40.21.20.70.41.00.30.40.9389/12miR-240.60.81.10.91.30.30.70.70.51.10.50.40.3339/12Relative expression of aurora kinase A protein and miRNAs was detected in the paired benign and malignant prostate tissues. The data of aurora kinase A protein and miRNA expression in benign or tumor prostate tissues from three independent experiments were averaged, the data for benign tissues were arbitrarily designated as one, and the relative expression was obtained when data for tumor tissues were divided by data for benign tissues.RD, rate of decrease; –, not applicable. Open table in a new tab Relative expression of aurora kinase A protein and miRNAs was detected in the paired benign and malignant prostate tissues. The data of aurora kinase A protein and miRNA expression in benign or tumor prostate tissues from three independent experiments were averaged, the data for benign tissues were arbitrarily designated as one, and the relative expression was obtained when data for tumor tissues were divided by data for benign tissues. RD, rate of decrease; –, not applicable. To validate direct interaction of miR-199a-3p with aurora kinase A mRNAs, their 3′-UTR target site was cloned into luciferase reporter plasmid. This construct was cotransfected with miR-199a-3p mimics into HeLa cells. Cells transfected with miR-199a-3p mimics caused a reduction in luciferase expression (Figure 2). Site-directed mutagenesis of the miR-199a-3p recognition site, located on the aurora kinase A 3′-UTR, abolished interactions between the miRNA and its aurora kinase A targets. This result indicates that miR-199a-3p can directly target aurora kinase A. We examined the expression of miR-199a-3p and aurora kinase A in PCa cell lines PC-3, DU145, and LNCaP, and found that aurora kinase A protein was the highest and miR-199a-3p was the lowest in DU145 cells (Figure 3A). To study the regulation of aurora kinase A expression by miR-199a-3p, we transfected the agomir or antagomir of miR-199a-3p into DU145 cells. We found that miR-199a-3p agomir significantly decreased aurora kinase A protein, whereas miR-199a-3p antagomir significantly increased aurora kinase A protein. Cleaved caspase 3 (CC3) was nearly undetectable in control groups, whereas miR-199a-3p agomir obviously increased CC3 protein (Figure 3B). We then examined the effect of miR-199a-3p agomir on cell cycle distribution of DU145 through flow cytometry. In agomir-treated cells, the percentage of cells in G2-M was increased to approximately 13.98% at 12 hours, 23.21% at 24 hours, and 29.45% at 48 hours. However, in agomir control-treated cells, the percentage of cells in G2-M was approximately 9.23% at each time point. In addition, we found that a fraction of cells in the sub-G1 phase of the cell cycle (indicating apoptotic cells) in the agomir-treated cells was significantly increased compared with the control-treated cells (Figure 3C). To further detect the function of miR-199a-3p agomir in vivo, we set up a tumor model by s.c. transplanting DU145 cells into nude mice. After tumors reached approximately 65 mm3, 10 nmol/L agomir and agomir control in 30 μL PBS or 30 μL PBS alone was injected into the tumor tissue every 2 days by multipoint intratumoral injection, lasting for 15 days. We found that agomir significantly inhibited the growth of human PCa cells as solid tumors in athymic nude mice, whereas control had no effect on tumor growth (Figure 4, A and B). Throughout the study, animals did not exhibit signs of discomfort as the result of tumor growth, and the body weights of the mice were not adversely affected by agomir administration (Figure 4C). We also found that agomir obviously inhibited aurora kinase A expression in tumor tissues (Figure 4D). Conversely, active caspase-3 in tumor tissues was increased (Figure 4D). After treatment of DU145 cells with the demethyltransferase inhibitor, AZA, methylation was decreased in the miR-199a gene (Figure 5A), and miR-199a-3p expression was up-regulated as determined by RT-PCR (Figure 5B). Aurora kinase A was down-regulated, as detected by Western blot analysis (Figure 5C), in these same DU145 cells, suggesting expression of miR-199a-3p in DU145 cells is methylation dependent. To analyze the relevance of promoter methylation on miR-199a-3p expression in PCa, MSP was performed for the 12 cases previously described. The average methylation percentage was 30% in normal surrounding tissue and 83% in paired cancer tissue (Figure 6A). This difference leads us to envision a relationship between miR-199a-3p expression, its promoter methylation, and clinical parameters. We then enlarged the sample size to examine this potential relationship. The FFPE prostatic tissues were obtained from The People's Hospital of Sichuan Province. All patients had undergone surgery at the same hospital between 2007 and 2011. A total of 50 PCa samples and 20 normal prostate tissues were acquired. RNA or DNA was extracted from FFPE tissues. miRNA was detected, and bisulfite conversion and MSP reactions were performed as previously described. Comparison of the noncancerous versus malignant groups indicated miR-199a-3p was significantly down-regulated in malignant tumors (Figure 6B). The relative expression of miR-199a-3p and the methylation of the miR-199a gene were correlated to tumor stage and Gleason score of PCa (Figure 6, B and C). Herein, we found that miR-199a-3p was down-regulated in PCa tissues compared with normal prostate tissues, and the expression pattern of miR-199a-3p was inversely correlated with the expression pattern of aurora kinase A, an oncogene that contributes to the malignant phenotypes of PCa. Furthermore, miR-199a-3p agomir attenuated PCa growth, which was concurrent with down-regulation of protein expression of aurora kinase A. Moreover, we demonstrated that miR-199a-3p targeted the aurora kinase A 3′-UTR. Thus, the present study indicates that miR-199a-3p plays a critical role in inhibition of PCa growth by targeting aurora kinase A. We also focused on the mechanism by which miR-199a-3p is down-regulated in PCa. miR-199a is regulated by its promoter methylation in testicular cancer,9Cheung H.H. Davis A.J. Lee T.L. Pang A.L. Nagrani S. Rennert O.M. Chan W.Y. Methylation of an intronic region regulates miR-199a in testicular tumor malignancy.Oncogene. 2011; 30: 3404-3415Crossref PubMed Scopus (120) Google Scholar and we hypothesized that this mechanism also played a role in PCa. We then treated DU145 cells with the demethyltransferase inhibitor, AZA, showing that the unmethylated fraction was increased in the miR-199a promoter after AZA treatment, and that miR-199a-3p expression was up-regulated and aurora kinase A expression was down-regulated. These results suggest miR-199a-3p expression in DU145 cells is methylation dependent. Next, when we examined the relevance of promoter methylation with miR-199a-3p expression in PCa patients, it showed more methylation in PCa tissues than in normal tissues, suggesting miR-199a promoter methylation might contribute to cancer malignancy. We then measured miR-199a promoter methylation status and miR-199a-3p expression in an enlarged PCa cohort and evaluated the relationship between miR-199a-3p expression, its promoter methylation, and clinical parameters of PCa. We found that the relative expression of miR-199a-3p and the percentage of methylation were correlated to tumor stage and Gleason score of the PCa, suggesting clinical significance for miR-199a-3p expression and miR-199a promoter methylation in PCa. Aberrant miR-199a-3p expression has been reported in several cancers; it can be either up-regulated or down-regulated in distinct cancer cell types.12Sakurai K. Furukawa C. Haraguchi T. Inada K. Shiogama K. Tagawa T. Fujita S. Ueno Y. Ogata A. Ito M. Tsutsumi Y. Iba H. MicroRNAs miR-199a-5p and -3p target the Brm subunit of SWI/SNF to generate a double-negative feedback loop in a variety of human cancers.Cancer Res. 2011; 71: 1680-1689Crossref PubMed Scopus (95) Google Scholar Duan et al13Duan Z. Choy E. Harmon D. Liu X. Susa M. Mankin H. Hornicek F. MicroRNA-199a-3p is downregulated in human osteosarcoma and regulates cell proliferation and migration.Mol Cancer Ther. 2011; 10: 1337-1345Crossref PubMed Scopus (214) Google Scholar have reported that miR-199a-3p was down-regulated in human osteosarcoma, and that restoring miR-199a-3p's function may provide therapeutic benefits in osteosarcoma. However, Wan et al14Wan D. He S. Xie B. Xu G. Gu W. Shen C. Hu Y. Wang X. Zhi Q. Wang L. Aberrant expression of miR-199a-3p and its clinical significance in colorectal cancers.Med Oncol. 2013; 30: 378Crossref PubMed Scopus (36) Google Scholar reported that miR-199a-3p expression was significantly up-regulated in human colorectal cancer. The different expression patterns of miR-199a-3p found in different cancers suggests a complicated role for miR-199a-3p in regulating cancer phenotype. Herein, we show, for the first time to our knowledge, that miR-199a-3p is a potent inhibitor of aurora kinase A and it inhibits PCa growth through targeting aurora kinase A. However, we still realize that, although aurora kinase A may be the primary gene regulated by miR-199a-3p in PCa, we cannot exclude the possibility that other targeted genes of miR-199a-3p might also exert some functions in PCa growth. Up until now, many inhibitors against aurora kinase A have been developed, such as VX-680,15Harrington E.A. Bebbington D. Moore J. Rasmussen R.K. Ajose-Adeogun A.O. Nakayama T. Graham J.A. Demur C. Hercend T. Diu-Hercend A. Su M. Golec J.M. Miller K.M. VX-680, a potent and selective small-molecule inhibitor of the Aurora kinases, suppresses tumor growth in vivo.Nat Med. 2004; 10: 262-267Crossref PubMed Scopus (898) Google Scholar MLN8054,16Manfredi M.G. Ecsedy J.A. Meetze K.A. Balani S.K. Burenkova O. Chen W. Galvin K.M. Hoar K.M. Huck J.J. LeRoy P.J. Ray E.T. Sells T.B. Stringer B. Stroud S.G. Vos T.J. Weatherhead G.S. Wysong D.R. Zhang M. Bolen J.B. Claiborne C.F. Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase.Proc Natl Acad Sci U S A. 2007; 104: 4106-4111Crossref PubMed Scopus (377) Google Scholar and aurora kinase A RNA interference.17Hata T. Furukawa T. Sunamura M. Egawa S. Motoi F. Ohmura N. Marumoto T. Saya H. Horii A. RNA interference targeting aurora kinase a suppresses tumor growth and enhances the taxane chemosensitivity in human pancreatic cancer cells.Cancer Res. 2005; 65: 2899-2905Crossref PubMed Scopus (213) Google Scholar However, to our knowledge, this is the first report to demonstrate that miRNA-mediated knockdown of aurora kinase A can be used as a specific gene-targeting therapy to suppress progression of PCa. As we know, miRNAs are endogenously produced molecules in the body and have higher stability and better tissue compatibility compared with other agents7Sethupathy P. Collins F.S. MicroRNA target site polymorphisms and human disease.Trends Genet. 2008; 24: 489-497Abstract Full Text Full Text PDF PubMed Scopus (285) Google Scholar; thus, they might be better treatments than currently available therapeutics. Throughout the study, we found that animals did not exhibit signs of discomfort, and the body weights of the mice were not adversely affected by ribonucleotide administration, proving the safety of miRNA administration in mice. Further work should be performed to enhance the delivery ratio of miRNAs/solid tumors to improve their therapeutic effects. In summary, our findings show that miR-199a-3p is a potent inhibitor of aurora kinase A in PCa. miR-199a-3p agomir effectively attenuates malignant progression of PCa both in vitro and in vivo. Revealing novel mechanisms for oncogene inhibition by miRNA-mediated pathways offers new avenues for PCa treatment. We thank Stephanie Cambier for proofreading the manuscript.
What problem does this paper attempt to address?
-
Nonconserved Mir‐608 Suppresses Prostate Cancer Progression Through RAC2/PAK4/LIMK1 and BCL2L1/caspase‐3 Pathways by Targeting the 3′‐utrs of RAC2/BCL2L1 and the Coding Region of PAK4
Xu Zhang,Jiajie Fang,Shiming Chen,Weiyu Wang,Shuai Meng,Ben Liu
DOI: https://doi.org/10.1002/cam4.2455
2019-01-01
Abstract:The aim of this study is to investigate the functions and mechanisms of miR-608 in prostate cancer (PCa). CISH and qRT-PCR analysis demonstrated that miR-608 was low expressed in PCa tissues and cells, which was partly attributed to the methylation of CpG island adjacent to the transcription start site (TSS) of miR-608 gene. Intracellular miR-608 overexpression inhibited in vivo PCa tumor growth, and suppressed PCa cell proliferation, G2/M transition, and migration in vitro, which was independent of EMT-associated mechanisms. Then RAC2, a GTPase previously deemed hematopoiesis-specific but now discovered to exist and play important roles in PCa, was verified by western blot and dual-luciferase reporter assays to mediate the effects of miR-608 through RAC2/PAK4/LIMK1/cofilin pathway. MiR-608 also promoted the apoptosis of PCa cells through BCL2L1/caspase-3 pathway by targeting the 3'-UTR of BCL2L1. Moreover, PAK4, the downstream effector of RAC2, was found to be targeted by miR-608 at the mRNA coding sequence (CDS) instead of the canonical 3'-UTR. Knocking down RAC2, PAK4, or BCL2L1 with siRNAs reproduced the antiproliferative, mitosis-obstructive, antimigratory and proapoptotic effects of miR-608 in PCa cells, which could be attenuated by downregulating miR-608. In conclusion, miR-608 suppresses PCa progression, and its activation provides a new therapeutic option for PCa.
-
Micro-RNA-186-5p inhibition attenuates proliferation, anchorage independent growth and invasion in metastatic prostate cancer cells
Dominique Z. Jones,M. Lee Schmidt,Suman Suman,Katharine R. Hobbing,Shirish S. Barve,Leila Gobejishvili,Guy Brock,Carolyn M. Klinge,Shesh N. Rai,Jong Park,Geoffrey J. Clark,Rajesh Agarwal,LaCreis R. Kidd
DOI: https://doi.org/10.1186/s12885-018-4258-0
IF: 4.638
2018-04-13
BMC Cancer
Abstract:BackgroundDysregulation of microRNA (miRNA) expression is associated with hallmarks of aggressive tumor phenotypes, e.g., enhanced cell growth, proliferation, invasion, and anchorage independent growth in prostate cancer (PCa).MethodsSerum-based miRNA profiling involved 15 men diagnosed with non-metastatic (stage I, III) and metastatic (stage IV) PCa and five age-matched disease-free men using miRNA arrays with select targets confirmed by quantitative real-time PCR (qRT-PCR). The effect of miR-186-5p inhibition or ectopic expression on cellular behavior of PCa cells (i.e., PC-3, MDA-PCa-2b, and LNCaP) involved the use bromodeoxyuridine (BrdU) incorporation, invasion, and colony formation assays. Assessment of the impact of miR-186-5p inhibition or overexpression on selected targets entailed microarray analysis, qRT-PCR, and/or western blots. Statistical evaluation used the modified t-test and ANOVA analysis.ResultsMiR-186-5p was upregulated in serum from PCa patients and metastatic PCa cell lines (i.e., PC-3, MDA-PCa-2b, LNCaP) compared to serum from disease-free individuals or a normal prostate epithelial cell line (RWPE1), respectively. Inhibition of miR-186-5p reduced cell proliferation, invasion, and anchorage-independent growth of PC-3 and/or MDA-PCa-2b PCa cells. AKAP12, a tumor suppressor target of miR-186-5p, was upregulated in PC-3 and MDA-PCa-2b cells transfected with a miR-186-5p inhibitor. Conversely, ectopic miR-186-5p expression in HEK 293 T cells decreased AKAP12 expression by 30%. Both pAKT and β-catenin levels were down-regulated in miR-186-5p inhibited PCa cells.ConclusionsOur findings suggest miR-186-5p plays an oncogenic role in PCa. Inhibition of miR-186-5p reduced PCa cell proliferation and invasion as well as increased AKAP12 expression. Future studies should explore whether miR-186-5p may serve as a candidate prognostic indicator and a therapeutic target for the treatment of aggressive prostate cancer.
oncology
-
Androgen receptor-regulated miRNA-193a-3p targets AJUBA to promote prostate cancer cell migration.
Li Jia,Bin Gui,Dali Zheng,Keith F Decker,Ilker Tinay,Mingyue Tan,Xiaowei Wang,Adam S Kibel
DOI: https://doi.org/10.1002/pros.23356
2017-01-01
Abstract:Background Dysregulation of microRNA (miRNA) expression is implicated in cancer development and progression by modulating oncogenes or tumor suppressors at the post-transcriptional level. Methods To investigate the role of miRNAs in prostate cancer (PCa) progression, we performed small RNA-sequencing (RNA-seq) analysis in androgen-dependent LNCaP cells and LNCaP-derived castration-resistant prostate cancer (CRPC) C4-2B cells. For functional validation, we specifically investigated miR-193a-3p, which is highly upregulated in C4-2B cells and modulated by the androgen receptor (AR). We elucidated the role of miR-193a-3p and its downstream target gene in PCa cell migration using biochemical approaches. Results We identified a subset of differentially expressed miRNAs in C4-2B cells compared to LNCaP cells. Computational analysis shows that the targets of upregulated miRNAs are significantly associated with downregulated protein-coding mRNAs in C4-2B cells. Gene Ontology analysis further reveals that these downregulated mRNAs are significantly enriched in cell-cell adhesion functions. Downregulation of these miRNA-targeted genes may change PCa cell motility resulting in the acquisition of metastatic potential. We then focus on miR-193a-3p and demonstrate overexpression of miR-193a-3p increases cell migration through downregulating its target AJUBA. AJUBA is a LIM domain protein and contributes to the formation and stability of cadherin-mediated cell-cell adhesion. Loss of AJUBA enhances PCa migration and downregulation of AJUBA expression is observed in metastatic PCa tumors. Conclusions Our results suggest a novel AR/miR-193a-3p/AJUBA pathway implicated in PCa progression. MiR-193a-3p is a potential therapeutic target for metastatic PCa.
-
Abstract 462: Developing the miR-888 cluster as a therapeutic target for prostate cancer
Katherine Routon,Trevor Fachko,Vishal Kasina,Raman Bahal,Aurora Esquela Kerscher
DOI: https://doi.org/10.1158/1538-7445.am2024-462
IF: 11.2
2024-03-22
Cancer Research
Abstract:Abstract Prostate Cancer (PCa) is a serious health crisis in the United States. 20% of men diagnosed with PCa will progress to fast-growing, advanced disease. There are no curative treatment options for PCa that has spread to distant sites and the 5-year survival rate for metastatic disease remains 30%. Recognizing the need for better PCa therapeutics, we are developing novel anti-microRNA delivery reagents targeted against the noncoding RNA miR-888 cluster to block prostate tumor progression. Our lab identified these oncomiRs in a biomarker discovery screen as elevated in human metastatic PCa cell lines and prostatic fluids from patients with high-grade PCa. We noted that over-expression of miR-888 cluster members (miR-888, miR-891a) promoted prostate cell growth and induced invasiveness in vitro. miR-888 and miR-891a also accelerated xenograft tumor growth in mice. Treatment of aggressive PCa cells in culture with synthetic antisense inhibitors against individual cluster members reversed these growth and invasion phenotypes, highlighting their clinical potential. We are testing pH Low Insertion Peptide (pHLIP)-Peptide Nucleic Acid (PNA)-based antimiR conjugates against miR-888 and miR-891a. The delivery vehicle pHLIP is a 36-amino acid peptide that at low pH (< pH 6.5) adopts an alpha-helical conformational change and facilitates the insertion of its C-terminus across lipid bilayers to transport conjugated anti-miRNA cargo into cells. When administered systemically, pHLIP effectively targets solid tumors with acidic microenvironments. We found that these reagents are readily internalized by human PCa cells cultured at pH 6.0 and in prostate xenograft tumors when delivered systemically in mice. Treatment significantly repressed miR-888 and miR-891a and acted to slow prostate cell growth in vitro. We are currently moving these studies into mouse prostate models. Furthermore, we are investigating the downstream signaling pathways for the miR-888 cluster. Our work indicates that the miR-888 cluster network coordinates the repression of the Tissue Inhibitors of Metalloproteinase (TIMP) and SMAD4 anti-metastatic pathways to drive extracellular matrix degradation and prostate cell invasion, and as a consequence, induces matrix metalloproteinase (MMP) activity. Expression profiling arrays and luciferase reporter assays indicated that multiple members of the miR-888 cluster co-suppress TIMPs in the prostate and similarly elevate multiple MMPs as well as cadherins, fibronectin and heparinase, and co-modulated several immune response genes. We are currently studying the consequence of various CRISPR-mediated miR-888 cluster deletions to understand how this non-coding RNA cluster functionally overlaps to promote aggressive PCa. This work will establish new clinical tools for aggressive PCa and optimize patient treatment management. Citation Format: Katherine Routon, Trevor Fachko, Vishal Kasina, Raman Bahal, Aurora Esquela Kerscher. Developing the miR-888 cluster as a therapeutic target for prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 462.
oncology
-
In silico analysis of prognostic and diagnostic significance of target genes from prostate cancer cell lines derived exomicroRNAs
Antonio Altuna-Coy,Xavier Ruiz-Plazas,Verónica Arreaza-Gil,José Segarra-Tomás,Matilde R Chacón
DOI: https://doi.org/10.1186/s12935-023-03123-1
2023-11-17
Abstract:Background: Cancer-secreted exovesicles are important for cell-to-cell communication by altering cancer-related signalling pathways. Exovesicles-derived miRNAs (exomiRNAs)-target genes can be useful for diagnostic and prognostic purposes. Methods: ExomiRNA from prostate cancer (PCa) cells (PC-3 and LNCaP) were quantified by qRT-PCR and compared to the healthy cell line RWPE-1 by using miRNome PCR 752 miRNAs Panel. MiRNet database was used to predict exomiRNA-target genes. ExomiRNA-target genes pathway functional enrichment was performed by using Reactome database and Enrichr platform. Protein-protein interaction analysis was carried out by using the STRING database. RNA target-gene sequencing data from The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) database was screened out in 465 PCa patients for candidate gene expression in prostate tumour (PT) tissue and non-pathologic prostate (N-PP) tissue. Signature gene candidates were statistically analysed for diagnosis and prognosis usefulness. Results: A total of 36 exomiRNAs were found downregulated when comparing PCa cells vs a healthy cell line; and when comparing PC-3 vs LNCaP, 14 miRNAs were found downregulated and 52 upregulated. Reactome pathway database revealed altered pathways and genes related to miRNA biosynthesis, miRNA-mediated gene silencing (TNRC6B and AGO1), and cell proliferation (CDK6), among others. Results showed that TNRC6B gene expression was up-regulated in PT tissue compared to N-PP (n = 52 paired samples) and could be useful for diagnostic purposes. Likewise, gene expression levels of CDK6, TNRC6B, and AGO1 were down-regulated in high-risk PT (n = 293) compared to low-risk PCa tissue counterparts (n = 172). When gene expression levels of CDK6, TNRC6B, and AGO1 were tested as a prognostic panel, the results showed that these improve the prognostic power of classical biomarkers. Conclusion: ExomiRNAs-targets genes, TNRC6B, CDK6, and AGO1, showed a deregulated expression profile in PCa tissue and could be useful for PCa diagnosis and prognosis.
-
MicroRNA-302a Suppresses Tumor Cell Proliferation by Inhibiting AKT in Prostate Cancer.
Gui-Ming Zhang,Chun-Yang Bao,Fang-Ning Wan,Da-Long Cao,Xiao-Jian Qin,Hai-Liang Zhang,Yao Zhu,Bo Dai,Guo-Hai Shi,Ding-Wei Ye
DOI: https://doi.org/10.1371/journal.pone.0124410
IF: 3.7
2015-01-01
PLoS ONE
Abstract:Micro (mi) RNAs are important regulators involved in various physical and pathological processes, including cancer. The miRNA-302 family has been documented as playing a critical role in carcinogenesis. In this study, we investigated the role of miRNA-302a in prostate cancer (PCa). MiRNA-302a expression was detected in 44 PCa tissues and 10 normal prostate tissues, and their clinicopathological significance was analyzed. Cell proliferation and cell cycle analysis were performed on PCa cells that stably expressed miRNA-302a. The target gene of miRNA-302a and the downstream pathway were further investigated. Compared with normal prostate tissues, miRNA-302a expression was downregulated in PCa tissues, and was even lower in PCa tissues with a Gleason score ≥8. Overexpression of miRNA-302a induced G1/S cell cycle arrest in PCa cells, and suppressed PCa cell proliferation both in vitro and in vivo. Furthermore, miRNA-302a inhibits AKT expression by directly binding to its 3΄ untranslated region, resulting in subsequent alterations of the AKT-GSK3β-cyclin D1 and AKT-p27Kip1 pathway. These results reveal miRNA-302a as a tumor suppressor in PCa, suggesting that miRNA-302a may be used as a potential target for therapeutic intervention in PCa.
-
Abstract A077: MicroRNA drivers of resistance to androgen deprivation therapy in prostate cancer
Philippa C. Saunders,Claire Fletcher
DOI: https://doi.org/10.1158/1538-7445.prca2023-a077
IF: 11.2
2023-06-02
Cancer Research
Abstract:Abstract INTRODUCTION: Prostate cancer is the most prevalent malignancy affecting Western males. It is initially an androgen-dependent disease: androgens bind to the androgen receptor and drive expression of genes that promote proliferation and evasion of apoptosis. Despite reduced androgen dependence in advanced prostate cancer, androgen receptor signalling remains a key driver of growth. Androgen deprivation therapy (ADT) is therefore a first line treatment approach and works well initially, but resistance inevitably develops. Abiraterone and Enzalutamide are drugs widely used in ADT and are androgen synthesis and androgen receptor signalling inhibitors respectively. The shortage of other treatment options means acquired resistance to these drugs is a major clinical problem. MicroRNAs (miRs) are important mediators of post-transcriptional gene regulation and show altered expression in cancer. Several have been linked to the development of resistance to ADT. Manipulation of such miRs may be a pathway to breakthrough treatments for advanced prostate cancer. This study aimed to validate ADT resistance-implicated miRs and their clinically relevant targets. MATERIALS AND METHODS: Small RNA-sequencing of Abiraterone- and Enzalutamide-resistant C42 prostate cancer cells identified subsets of miRs dysregulated as compared to parental cells. Real-Time Quantitative Reverse Transcription PCR (qRT-PCR) was used to validate altered expression of candidate ADT resistance-implicated miRs 195-5p, 497-5p and 29a-5p in ADT-resistant and -responsive prostate cancer cell lines, patient-derived xenografts (PDXs) and primary prostate cancer explants. RESULTS AND DISCUSSION: This study suggests a possible role for miR-497-5p in the development of ADT resistance in prostate cancer. MiR-497-5p expression was increased in ADT-resistant versus ADT-responsive prostate cancer cells. Importantly, miR-497-5p expression was also increased in Enzalutamide-treated, castrated (ADT-mimicking) PDXs versus intact PDXs. MiR-195-5p was also elevated in ADT-resistant versus -responsive prostate cancer cells, while there was a drop in miR-29a-5p expression. Candidate clinically relevant targets of miR-497-5p in prostate cancer were identified by mining AGO-PAR-CLIP-seq data sets and may include AVL9 and FZD6. CONCLUSION: In summary, this study identified microRNAs that are implicated in prostate cancer resistance to androgen deprivation therapy and could represent novel therapeutic targets for advanced disease. Citation Format: Philippa C. Saunders, Claire Fletcher. MicroRNA drivers of resistance to androgen deprivation therapy in prostate cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Prostate Cancer Research; 2023 Mar 15-18; Denver, Colorado. Philadelphia (PA): AACR; Cancer Res 2023;83(11 Suppl):Abstract nr A077.
oncology
-
MAGI2 antisense RNA 3 (MAGI2-AS3) knockdown contributes to prostate cancer by downregulating proline-rich membrane anchor 1 (PRIMA1) via miR-142-3p
Weiqi Lai,Xiaoke Huang,Shadan Li,Caiyong Song,Chengwan Liu,Qingfeng Tang
DOI: https://doi.org/10.5114/aoms/167431
2023-07-01
Archives of Medical Science
Abstract:Introduction Long non-coding RNA Membrane-Associated Guanylate Kinase Inverted 2 Antisense RNA 3 (MAGI2-AS3) has been identified as a predictive characteristic for prostate cancer (PC). However, the underlying mechanism of MAGI2-AS3 in PC is yet unclear. Material and methods The gene expression was detected via reverse transcription quantitative PCR. Cell viability, apoptosis and invasion were detected via cell counting kit-8, caspase-3 activity, flow cytometry, and transwell assays. Tumor xenograft experiment was performed to measure the tumor growth in vivo. Luciferase and radioimmunoprecipitation assays were utilized to determine the association between microRNA (miR)-142-3p and MAGI2-AS3 or proline-rich membrane anchor 1 (PRIMA1). Results MAGI2-AS3 and PRIMA1 were established to be downregulated in PC, whereas miR-142-3p exhibited the upregulation in PC. In vitro and in vivo loss-of-function assays demonstrated that MAGI2-AS3 silencing increased the viability and invasiveness, enhanced the tumor growth of PC cells, while reducing apoptosis. PRIMA1 silencing in PC cells showed the similar effect as MAGI2-AS3 silencing. Moreover, the miR-142-3p inhibitor reversed the impacts of the downregulation of MAGI2-AS3 or PRIMA1 on the malignant behavior of PC cells. Conclusions MAGI2-AS3 knockdown enhanced the malignant behavior of PC cells by targeting miR-142-3p to suppress PRIMA1 expression. Our findings reveal that MAGI2-AS3 can be a promising therapeutic target for the treatment of PC.
medicine, general & internal
-
rAAV-based and intraprostatically delivered miR-34a therapeutics for efficient inhibition of prostate cancer progression
Jianzhong Ai,Jia Li,Qin Su,Hong Ma,Ran He,Qiang Wei,Hong Li,Guangping Gao
DOI: https://doi.org/10.1038/s41434-021-00275-5
Abstract:At present, there is no effective treatment for prostate cancer (PCa). Previous studies have reported that miR-34a is significantly downregulated in PCa cells; therefore, modulation of miR-34a expression might be a promising therapeutic approach for PCa treatment. To this end, we first verified the downregulation of miR-34a in prostate tumors from a transgenic adenocarcinoma mouse prostate (TRAMP) model. We found that miR-34a overexpression significantly inhibited the cell cycle, viability, and migration of PCa cells by targeting its downstream genes. Next, we tested the concept of intraprostatic injection of rAAV9·pri-miR-34a into 8-week-old TRAMP mice to inhibit PCa progression. We observed that the treatment lowered body weights significantly compared to the control treatment starting at 30 weeks after injection. rAAV9·pri-miR-34a treatment also obviously extended the lifespan of TRAMP mice. Moreover, we confirmed that the neoplasia in the treated prostates was significantly diminished compared to that in the control group. In addition, overexpressed miR-34a downregulated the expression of its target genes. Taken together, our results demonstrated, for the first time, the potential of rAAV-mediated efficient modulation of miR-34a expression in the prostate to inhibit PCa progression by regulating its downstream gene expression.
-
The potential of miR-153 as aggressive prostate cancer biomarker
Irina Gilyazova,Elizaveta Ivanova,Mikhail Sinelnikov,Valentin Pavlov,Elza Khusnutdinova,Ilgiz Gareev,Aferin Beilerli,Ludmila Mikhaleva,Yanchao Liang
DOI: https://doi.org/10.1016/j.ncrna.2022.10.002
2022-10-13
Abstract:Introduction: Prostate cancer (PC) is one of the most frequently diagnosed cancers in males. MiR-153, as a member of the microRNA (miRNA) family, plays an important role in PC. This study aims to explore the expression and possible molecular mechanisms of the miR-153 action. Methods: Formalin-fixed paraffin-embedded (FFPE) tissues were collected from prostatectomy specimens of 29 metastatic and 32 initial stage PC patients. Expression levels of miR-153 were measured using real-time reverse transcription polymerase chain reaction (qRT-PCR). 2-ΔΔCT method was used for quantitative gene expression assessment. The candidate target genes for miR-153 were predicted by TargetScan. Mutations in target genes of miR-153 were identified using exome sequencing. Protein-protein interaction (PPI) networks, Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to investigate the potential molecular mechanisms of miR-153 in PC. Results: MiR-153 was significantly up-regulated in PC tissues compared to non-cancerous tissues. The analysis of correlation between the expression level of miR-153 and clinicopathological factors revealed a statistically significant correlation with the stage of the tumor process according to tumor, node, metastasis (TNM) staging system (p = 0.0256). ROC curve analysis was used to evaluate the predictive ability of miR-153 for metastasis development and it revealed miR-153 as a potential prognostic marker (AUC = 0.85; 95%CI 0.75-0.95; sensitivity = 0.72, specificity = 0.86)). According to logistic regression model the high expression of miR-153 increased the risk of metastasis development (odds ratios = 3.14, 95% CI 1.62-8.49; p-value = 0.006). Whole exome sequencing revealed nonsynonymous somatic mutations in collagen type IV alpha 1 (COL4A1), collagen type IV alpha 3 (COL4A3), forkhead box protein O1 (FOXO1), 2-hydroxyacyl-CoA lyase 1 (HACL1), hypoxia-inducible factor 1-alpha (HIF-1A), and nidogen 2 (NID2) genes. Moreover, KEGG analysis revealed that the extracellular matrix-receptor (ECM-receptor) interaction pathway is mainly involved in PC. Conclusion: MiR-153 is up-regulated in PC tissues and may play an important role in aggressive PC by targeting potential target genes.
-
Oncogene Mir-106A Promotes Proliferation and Metastasis of Prostate Cancer Cells by Directly Targeting PTEN in Vivo and in Vitro
Bo Luo,Na Kang,Yan Chen,Lihong Liu,Yongmei Zhang
DOI: https://doi.org/10.23736/s0026-4806.17.05342-3
2017-01-01
Minerva Medica
Abstract:BACKGROUND: Prostate cancer (PCa) is the second leading contributor to male malignancy-associated death in developed countries. The study aimed to evaluate the effects of miR-106a on prostate cancer cells and the underlying molecular mechanism. METHODS: The cell proliferation was detected using MTT assay and colony formation, and cells migration and invasion were also analyzed. Luciferase reporter assay was carried out to explore the correlation between miR-106a and PTEN. Besides, reverse transcriptase-polymerase chain reaction and western blot were carried out to measure the mRNA and protein levels in prostate tissues and PCa cells. Immunocytochemical staining was performed to detect the PTEN level in PCa tissues. RESULTS: MiR-106a level was dramatically increased in the PCa tissues and PCa cell lines. The overexpression of miR-106a significantly promoted the proliferation, migration and invasion of PCa cells. Besides, we confirmed that PTEN was regulated by miR-106a expression and was a direct target of miR-106a. Most importantly, we further found that miR-106a could promote the PCa cells xenograft growth in the nude mice by targeting PTEN, which was significantly downregulated in PCa tissues and cell lines and was closely related with PCa metastasis. CONCLUSIONS: MiR-106a as an oncogene could promote proliferation and metastasis of prostate cancer cells by directly targeting PTEN in vivo and in vitro.
-
MiR-197 Inhibitor Loaded AbCD133@MSNs@GNR Affects the Development of Prostate Cancer Through Targeting ITGAV
Guanqun Ju,Yingjian Zhu,Tao Du,Wanli Cao,Jianhai Lin,Chun Li,Dongliang Xu,Zhijun Wang
DOI: https://doi.org/10.3389/fcell.2021.646884
2021-06-14
Abstract:Prostate cancer is one of the most severe male malignant tumors, which ranks second in mortality rate among all tumors. Traditional methods of treatment for prostate cancer produce obvious side effects and a high recurrence rate. Cancer stem cells are considered to be a group of cells that determine the proliferation, metastasis, and drug resistance of tumor. Prostate cancer therapy based on microRNAs and prostate cancer stem cells (PCSCs) has been a research hot spot in this field. Previous studies have reported that miR-197 plays an important role in the occurrence and development of prostate cancer, but the molecular mechanism of miR-197 on the development of prostate cancer has not been reported yet. In this study, we verified that miR-197 is significantly overexpressed in prostate cancer tissues and prostate cancer cells. Then, we verified that miR-197 expression affects the proliferation, invasion, and metastasis of prostate cancer cells by regulating integrin subunit alpha V (ITGAV) expression through STAT5 pathway, and the results indicated that the miR-197 inhibitor can be a prostate cancer suppressor. Then we synthesized the AbCD133@GNR@MSNs@miR-197 inhibitor drug carrier, in which 35.42 μg of the miR-197 inhibitor could be loaded in 1 mg of AbCD133@GNR@MSNs. The AbCD133@GNR@MSNs@miR-197 inhibitor demonstrated good photothermal properties and photothermal controlled-release properties. The modified CD133 antibodies on the surface of the nano drug carrier helped more drug carriers to enter the PCSCs. The pharmacodynamic effects of the AbCD133@GNR@MSNs@miR-197 inhibitor on PCSCs in vivo and in vitro were studied under near-infrared radiation. The results showed that the AbCD133@GNR@MSNs@miR-197 inhibitor prepared in this study could not only significantly suppress the development of PCSCs through ITGAV/STAT5 pathway but also significantly suppress the growth of PCSC solid tumors. In short, our study verified that miR-197 regulates the development of PCSCs through STAT5 pathway by targeting ITGAV, and the AbCD133@MSNs@GNR@miR-197 inhibitor could be a potential suppressor used in prostate cancer treatment. In short, our study found that miR-197 affected the development of prostate cancer by regulating ITGAV. The AbCD133@GNR@MSNs@miR-197 inhibitor prepared in this study could suppress the development and growth of PCSCs in vitro and in solid tumors not only by targeting the ITGAV but also through photothermal therapy. Our study not only provides a theoretical basis for the clinical treatment of prostate cancer but also provides a research scheme of drug loading and microRNA-based photothermal controlled therapy for prostate cancer.
-
Abstract PO-129: Using the castration resistant prostate cancer patient-derived xenograft (PDX) tumor model to identify mRNA/miRNA biomarkers which distinguish abiraterone treatment responders and non-responders
Bin Ouyang,Dan Song,Jacek Biesiada,Yan Ren,Mario Medvedovic,Pheruza Tarapore
DOI: https://doi.org/10.1158/1538-7445.tumhet2020-po-129
2020-11-01
Abstract:Abstract Prostate cancer (PCa) is the most frequent cancer occurring in men in the United States and is the second leading cause of cancer deaths in men. Median survival for patients with metastatic hormone refractory PCa (HRPC/CRPC) is around 18 months. Abiraterone (Abi) is a next-generation androgen receptor targeting agent which has significant effects on PCa patients' survival, and has been approved for treatment of metastatic and CRPC. However, it is clear that there is development of Abi resistance (Abi-R). Abi inhibits the steroidogenic enzyme CYP17A1, a critical enzyme in androgen biosynthesis in the adrenals, testis as well as tumor tissues. To screen and identify biomarkers (genes and microRNA (miRNA)) for Abi-R in a heterogenous tumor, we used patient derived PCa xenograft animal model (PCa-PDX mice). Recent advances in the development of PDX models and their increasing sophistication have led to their escalating use for anticancer drug research and development, and as predictive clinical models. Hence, for the correct identification of Abi-R markers, we proposed using PCa-PDX animal model (PDX mice). We hypothesized that changes in miRNA expression underlie Abi resistance (Abi-R) mechanisms. Each tumor bearing mouse was castrated resulting in tumor regression followed by regrowth/relapse phase. Mice were then treated with Abi. The tumors were collected either once they regressed (called CRABS), or reinoculated into castrated host if they showed continued growth. The new host was treated with Abi once tumors reached 50-300 mm3. Tumors were harvested when they reached a volume of ~600-800 mm3 (CRABR2). The RNA was extracted from tumors using the miRvana kits, and RNA/miRNA-seq performed. We compared the Abi sensitive vs the Abi resistant (CRABR2) groups, to identify 316 genes and 55 miRNA whose expression was significantly changed between these two groups. We used Venn diagrams to overlap the validated gene targets for the miRNA (5,557 genes) with our 316 gene dataset. 85 common genes were identified, of which 63 genes were validated targets of hsa-miR-335 (1.7 fold in CRABS vs CRABR2), and 8 genes were validated targets of hsa-miR-574 (0.4 fold in CRABS vs CRABR2). Hsa-miR-335 has been previously identified to function as a candidate tumor suppressor in PCa. Reduced expression of miR-335 was found in human PCa tissues comparing with paired adjacent benign prostate tissues (P < 0.05). Moreover, the increased expression of miR-335 suppressed cell proliferation, invasion and migration of PCa cell lines in vitro. It has been found to suppress bone metastasis in PCa via targeting endothelial nitric oxide synthase. MicroRNA-574-5p promotes metastasis of non-small cell lung cancer, triple negative breast cancer and colorectal cancer to name a few. However, their function in Abi resistance has not been explored. In conclusion, we have identified two promising miRNA for distinguishing Abi sensitive from resistant tumors whose targeting should have profound effects on sensitizing castration resistant tumors to Abi. Citation Format: Bin Ouyang, Dan Song, Jacek Biesiada, Yan Ren, Mario Medvedovic, Pheruza Tarapore. Using the castration resistant prostate cancer patient-derived xenograft (PDX) tumor model to identify mRNA/miRNA biomarkers which distinguish abiraterone treatment responders and non-responders [abstract]. In: Proceedings of the AACR Virtual Special Conference on Tumor Heterogeneity: From Single Cells to Clinical Impact; 2020 Sep 17-18. Philadelphia (PA): AACR; Cancer Res 2020;80(21 Suppl):Abstract nr PO-129.
-
Hsa-miR-1-2/miR-133a-1 Cluster Expression Silencing by DNA Hypermethylation Triggers Oncogenes Contributing to Prostate Cancer Aggressiveness
Cintia Massillo,Rocío Belén Duca,Paula Lucía Farré,Kevin Gardner,Ezequiel Lacunza,Adriana De Siervi
DOI: https://doi.org/10.21203/rs.3.rs-800913/v1
2021-08-31
Abstract:Abstract Background: Prostate cancer (PCa) is currently the most commonly diagnosed type of cancer. The incidence and mortality of PCa worldwide correlate with increasing age and bad dietary habits. Previously, we investigated the mRNA/miRNA role on PCa development and progression using high fat diet (HFD) chronically fed mice models. Here our main goal was to investigate the effect of HFD on the expression of prostate cancer-related miRNAs and their relevance in PCa patients. Methods: Using microarray data from mice prostate tumors generated by TRAMP-C1 cell inoculation, we focused on the role of three candidate miRNAs (miR-133a-3p/133b/1-3p) and their target genes. Based on data from public databases, we examined the expression levels of hsa-miR-133a-3p/133b/1-3p and their correlation with clinicopathological features in PCa patients. The biological roles of hsa-miR-133a-3p/133b/1-3p and their relevant target genes were investigated by bioinformatics approaches. The promoter methylation of hsa-miR-133a-3p/133b/1-3p host genes and their correlation with mature miRNA expression was further evaluated. Results: We identified 6 up- and 18 down-regulated miRNAs in TRAMP-C1 mice prostate tumors under HFD conditions using miRNA microarrays. Target genes (1,278) of down-regulated miRNAs involved in cancer-related biological processes were identified using DIANA-TarBase and STRING databases. Three down-regulated miRNAs: hsa-miR-133a-3p, 133b and 1a-3p showed nine common target genes that negatively correlated with miRNA expression in prostate tumors from patients. Hsa-miR-133a-3p/133b/1-3p expression levels were significantly decreased in PCa compared to normal tissues and their low expression correlated with bad clinicopathological features in patients. We also examined the promoter region of hsa-miR-133a-3p/133b/1-3p encoding genes in PCa patients and then compared methylation at these loci with mature miRNA expression. This analysis revealed that hsa-miR-1-2/miR-133a-1 cluster promoter hypermethylation decreased hsa-miR-133a-3p/1-3p expression in prostate tumors. Furthermore, CENPF and WHSC1 , two common hsa-miR-133a-3p/133b/1-3p target genes, were found significantly up-regulated in PCa and positively correlated with advanced clinicopathological characteristics in PCa patients. Conclusion: Our results provide an explanation for the aggressiveness of PCa and link it mechanistically to the attenuation of hsa-miR-133a-3p/133b/1-3p expression by promoter hypermethylation. Hsa-miR-133a-3p/133b/1-3p downregulation may enhance PCa aggressiveness in part by targeting CENPF and WHSC1 . Therefore, hsa-miR-133a-3p/133b/1-3p might be potential therapeutic targets for lethal PCa.
-
MiR-199a-3p Suppresses Proliferation and Invasion of Prostate Cancer Cells by Targeting Smad1
Feng Qu,Jinyu Zheng,Weidong Gan,Huibo Lian,Hua He,Wuping Li,Tian Yuan,Yaling Yang,Xiaogong Li,Changwei Ji,Xiang Yan,Linfeng Xu,Hongqian Guo
DOI: https://doi.org/10.18632/oncotarget.17191
2017-01-01
Oncotarget
Abstract:Objectives This study was intended to analyze effects of miR-199a-3p and Smad1 on proliferation, migration and invasion of prostate cancer (PCa) cells. Results MiR-199a-3p was significantly decreased in PCa tissues in comparison to that in adjacent normal tissues (P < 0.05). Over-expressed miR-199a-3p markedly suppressed proliferation and invasion of PCa cells (P < 0.05). MiR-199a-3p was negatively correlated with Smad1 expression, and overexpression of Smad1 could antagonize the effects of miR-199a-3p on PCa cells. Materials and methods The PCa tissues and their adjacent normal tissues were collected from 54 PCa patients. Expressions of miR-199a-3p and Smad1 mRNA in tissues and cells were evaluated with real-time quantitative polymerase chain reaction (RT-qPCR), and immunohistochemistry assay was used to detect Smad1 protein expressions. The target relationship between miR-199a-3p and Smad1 was assessed by luciferase reporter assay. The PCa cell lines (i.e. PC-3 cells) were transfected with miR-199a-3p mimics and Smad1-cDNA. MTT and Transwell assays were applied to detect proliferative, migratory and invasive abilities of PCa cells. Conclusions MiR-199a-3p suppressed proliferation and invasion of PCa cells by targeting Smad1.
-
Abstract 5682: Micro RNA level changes in prostate cancer cells treated with MDMX and MDM2 inhibitors
Rojin Fatirkhorani,Ahmed Saleh Alsarrani,Shyam Sundar Jaganathan,Umamaheswari Natarajan,Appu Rathinavelu
DOI: https://doi.org/10.1158/1538-7445.am2024-5682
IF: 11.2
2024-03-22
Cancer Research
Abstract:Abstract INTRODUCTION: Prostate Cancer (PCa) is the most common non-cutaneous cancer and the second leading cause of cancer death among men in the United States. Most of the PCa cases are diagnosed in men between the ages of 65-74. Due to the recent push for frequent prostate screenings, these cancers are being detected early and treated much faster, leading to a 5-year survival of a localized or regional PCa to >99%. However, the 5-year survival of PCa with distant metastasis is significantly reduced to 32%, especially with involvement of osteoblastic bone lesions, lung lesions, and brain lesions. Therefore, there is a clinical need to identify prognostic markers and new targets for developing therapies and effectively treating advanced PCa. Previous studies have shown that levels of MDMX and MDM2, oncogenic inhibitors of p53 tumor suppressor protein, are significantly elevated in PCa cells. Inhibition of MDM2 and MDMX using specific inhibitors has been shown to elevate the levels of p21, p27, and p53 in LNCaP (Lymph Node Carcinoma of the Prostate) cells leading to cell death. Recent studies have demonstrated that micro RNAs (miRNAs) play an important role in supporting the tumor suppressor function of p53 through regulating the balance between p53 and MDM2. In this regard, several miRNAs were found to be significantly altered to modulate the level and intracellular mechanisms of p53 and MDM2 in various cancers. OBJECTIVES: To determine the relative levels of miRNA31, miRNA-34a and miRNA-128 in LNCaP cells that were treated with NSC-207895 (MDMX inhibitor), SJ-172550 (MDMX inhibitor), and RG-7388 (MDM2 inhibitor). METHODS: LNCaP cells were treated with NSC-207895 (20 uM), SJ-172550 (20 uM) or RG-7388 (2 uM) for 24 hrs. After completing the treatments, the miRNAs were extracted from the LNCaP cells using the miRNeasy Tissue/Cells Advanced Kit from Qiagen. We analyzed the levels of miRNA-31, miRNA-34a, and miRNA-128 in the treated samples using qRT-PCR analysis method and compared the levels to the control. During the qRT-PCR analysis the cDNA was amplified using Meridian Biosciences qPCR Master Mix and specific primers for miRNAs from Origene were used according to the manufacturer's protocol. RESULTS: Our experimental results indicate that inhibition of MDMX using NSC-207895 and SJ-172550 can elevate the levels of p21, p27, and p53 leading to cell death. Determination of cell death markers in the past revealed induction of necroptosis following drug treatments. Our current results indicate measurable changes in the levels of miRNA-31 and miRNA-34a following inhibition of MDMX using SJ-172550. We are in the process of determining the possible link between the changes in the levels of miRNAs and cell death. ACKNOWLEDGEMENT: This research was supported by the Royal Dames of Cancer Research, Ft. Lauderdale, Florida. Citation Format: Rojin Fatirkhorani, Ahmed Saleh Alsarrani, Shyam Sundar Jaganathan, Umamaheswari Natarajan, Appu Rathinavelu. Micro RNA level changes in prostate cancer cells treated with MDMX and MDM2 inhibitors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 5682.
oncology
-
Downregulation of miR‑224‑5p in prostate cancer and its relevant molecular mechanism via TCGA, GEO database and in silico analyses
Bin-Liang Gan,Li-Jie Zhang,Li Gao,Fu-Chao Ma,Rong-Quan He,Gang Chen,Jie Ma,Jin-Cai Zhong,Xiao-Hua Hu
DOI: https://doi.org/10.3892/or.2018.6766
Abstract:The function of the expression of microRNA (miR)‑224‑5p in prostate adenocarcinoma (PCa) remains to be elucidated, therefore, the present study aimed to investigate the clinical significance and potential molecular mechanism of miR‑224‑5p in PCa. Data on the expression of miR‑224‑5p in PCa were extracted from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), ArrayExpress and previous literature, and meta‑analyses with standardized mean difference (SMD) and summary receiver operating characteristic (sROC) methods were performed for statistical analyses. The prospective target genes of miR‑224‑5p were collected by overlapping the differentially expressed mRNAs in TCGA and GEO, and target genes predicted by miRWalk2.0. Subsequently, in silico analysis was performed to examine the associated pathways of miR‑224‑5p in PCa. The expression of miR‑224‑5p was markedly lower in PCa; the overall SMD was ‑0.562, and overall sROC area under the curve was 0.80. In addition, Kyoto Encyclopedia of Genes and Genomes analysis revealed that the prospective target genes of miR‑224‑5p were largely enriched in the amino sugar and nucleotide sugar metabolism signaling pathway, and three genes [UDP‑N‑acetylglucosamine pyrophosphorylase 1 (UAP1), hexokinase 2 (HK2) and chitinase 1 (CHIT1)] enriched in this pathway showed higher expression (P<0.05). In addition, key genes in the protein‑protein interaction network analysis [DNA topoisomerase 2‑α (TOP2A), ATP citrate lyase (ACLY) and ribonucleotide reductase regulatory subunit M2 (RRM2)] exhibited significantly increased expression (P<0.05). The results suggested that the downregulated expression of miR‑224‑5p may be associated with the clinical progression and prognosis of PCa. Furthermore, miR‑224‑5p likely exerts its effects by targeting genes, including UAP1, HK2, CHIT1, TOP2A, ACLY and RRM2. However, in vivo and in vitro experiments are required to confirm these findings.
-
Identification of Novel AR-Targeted MicroRNAs Mediating Androgen Signalling Through Critical Pathways to Regulate Cell Viability in Prostate Cancer
Wenjuan Mo,Jiyuan Zhang,Xia Li,Delong Meng,Yun Gao,Shu Yang,Xuechao Wan,Caihong Zhou,Fenghua Guo,Yan Huang,Stefano Amente,Enrico V. Avvedimento,Yi Xie,Yao Li
DOI: https://doi.org/10.1371/journal.pone.0056592
IF: 3.7
2013-01-01
PLoS ONE
Abstract:MicroRNAs (miRNAs) have been recognized as significantly involved in prostate cancer (PCa). Since androgen receptor (AR) plays a central role in PCa carcinogenesis and progression, it is imperative to systematically elucidate the causal association between AR and miRNAs, focusing on the molecular mechanisms by which miRNAs mediate AR signalling. In this study, we performed a series of time-course microarrays to observe the dynamic genome-wide expressions of mRNAs and miRNAs in parallel in hormone-sensitive prostate cancer LNCaP cells stimulated by androgen. Accordingly, we introduced Response Score to identify AR target miRNAs, as well as Modulation Score to identify miRNA target mRNAs. Based on theoretical identification and experimental validation, novel mechanisms addressing cell viability in PCa were unravelled for 3 miRNAs newly recognized as AR targets. (1) miR-19a is directly up-regulated by AR, and represses SUZ12, RAB13, SC4MOL, PSAP and ABCA1, respectively. (2) miR-27a is directly up-regulated by AR, and represses ABCA1 and PDS5B. (3) miR-133b is directly up-regulated by AR, and represses CDC2L5, PTPRK, RB1CC1, and CPNE3, respectively. Moreover, we found miR-133b is essential to PCa cell survival. Our study gives certain clues on miRNAs mediated AR signalling to cell viability by influencing critical pathways, especially by breaking through androgen's growth restriction effect on normal prostate tissue.
-
Methylation‐associated miR‐193b silencing activates master drivers of aggressive prostate cancer
Ying Z. Mazzu,Yuki Yoshikawa,Subhiksha Nandakumar,Goutam Chakraborty,Joshua Armenia,Lina E. Jehane,Gwo‐Shu Mary Lee,Philip W. Kantoff
DOI: https://doi.org/10.1002/1878-0261.12536
2019-07-19
Molecular Oncology
Abstract:<p>Epigenetic silencing of miRNAs is a primary mechanism of aberrant miRNA expression in cancer, and hypermethylation of miRNA promoters has been reported to contribute to prostate cancer initiation and progression. Recent data have shown that the miR‐193b promoter is hypermethylated in prostate cancer compared to normal tissue, but studies assessing its functional significance have not been performed. We aimed to elucidate the function of miR‐193b and identify its critical targets in prostate cancer. We observed an inverse correlation between miR‐193b level and methylation of its promoter in The Cancer Genome Atlas (TCGA) cohort. Overexpression of miR‐193b in prostate cancer cell lines inhibited invasion and induced apoptosis. We found that a majority of the top 150 genes downregulated when miR‐193b was overexpressed in liposarcoma are overexpressed in metastatic prostate cancer and that 41 miR‐193b target genes overlapped with the 86 genes in the aggressive prostate cancer subtype 1 (PCS1) signature. Overexpression of miR‐193b led to the inhibition of the majority of the 41 genes in prostate cancer cell lines. High expression of the 41 genes was correlated with recurrence of prostate cancer. Knockdown of miR‐193b targets <i>FOXM1</i> and <i>RRM2</i> in prostate cancer cells phenocopied overexpression of miR‐193b. Dual treatment with DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors decreased miR‐193b promoter methylation and restored inhibition of <i>FOXM1</i> and <i>RRM2</i>. Our data suggest that silencing of miR‐193b through promoter methylation may release the inhibition of PCS1 genes, contributing to prostate cancer progression and suggesting a possible therapeutic strategy for aggressive prostate cancer.</p><p>This article is protected by copyright. All rights reserved.</p>
oncology
-
miR-100-5p Downregulates mTOR to Suppress the Proliferation, Migration, and Invasion of Prostate Cancer Cells
Yun Ye,Su-Liang Li,Jian-Jun Wang
DOI: https://doi.org/10.3389/fonc.2020.578948
IF: 4.7
2020-12-01
Frontiers in Oncology
Abstract:Background Previous studies have shown that miR-100-5p expression is abnormal in prostate cancer. However, the role and regulatory mechanism of miR-100-5p requires further investigation. Thus, the aim of this study was to observe the effects of miR-100-5p on the proliferation, migration and invasion of prostate cancer (PCa) cells and to explore the potential related regulatory mechanism. Materials and Methods Differential miRNA expression analysis was performed using next-generation sequencing (NGS) in the patients with PCa and benign prostatic hyperplasia (BPH). The expression levels of miR-100-5p were detected using real-time fluorescence quantitative PCR (qRT-PCR). PCa cells were transfected with NC-mimics or miR-100-5p mimics, inhibitor by using liposome transfection. Moreover, the CCK-8 proliferation assay, colony formation assay, cell scratch assay and Transwell assay were used to detect the effects of miR-100-5p on cell proliferation, migration, and invasion. In addition, the target gene of miR-100-5p was verified by luciferase reporter gene assay, and the influence of miR-100-5p on the expression of mTOR mRNA by qRT-PCR and the expression of mammalian target of rapamycin (mTOR) protein was detected by western blot and immunohistochemical staining. Results Differential expression analysis of high-throughput sequencing data showed low expression of miR-100-5p in the patients of PCa. It was further confirmed by qRT-PCR that the expression of miR-100-5p in PCa cells was significantly lower than that in RWPE-1 cells ( P <0.01). miR-100-5p expression in lymph node carcinoma of prostate(LNCaP) cells was markedly upregulated after transfection with miR-100-5p mimics ( P <0.01), while cell proliferation, migration and invasion capacities were clearly reduced ( P <0.01). mTOR mRNA and protein expression was also substantially lowered ( P <0.01) and mTOR adjusted the expression of NOX4. Finally, we further confirmed by immunohistochemical staining that miR-100-5p regulated the expression of mTOR and NOX4. Conclusion miR-100-5p is expressed at low levels in PCa cells, and it can suppress PCa cell proliferation, migration and invasion, the mechanism of which is related to downregulating the expression of mTOR.
oncology