Open Access

Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics

  • Authors:
    • Zu‑Cheng Xie
    • Jia‑Cheng Huang
    • Li‑Jie Zhang
    • Bin‑Liang Gan
    • Dong‑Yue Wen
    • Gang Chen
    • Sheng‑Hua Li
    • Hai‑Biao Yan
  • View Affiliations

  • Published online on: October 25, 2018     https://doi.org/10.3892/mmr.2018.9598
  • Pages: 5630-5646
  • Copyright: © Xie et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer (PCa) remains a principal issue to be addressed in male cancer‑associated mortality. Therefore, the present study aimed to examine the clinical value and associated molecular mechanism of microRNA (miR)‑1 in PCa. A meta‑analysis was conducted to evaluate the diagnosis of miR‑1 in PCa via Gene Expression Omnibus and ArrayExpress datasets, The Cancer Genome Atlas miR‑1 expression data and published literature. It was identified that expression of miR‑1 was significantly downregulated in PCa. Decreased miR‑1 expression possessed moderate diagnostic value, with area under the curve, sensitivity, specificity and odds ratio values at 0.73, 0.77, 0.57 and 4.60, respectively. Using bioinformatics methods, it was revealed that a number of pathways, including the ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’, were important in PCa. A total of seven hub genes, including phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthase (PAICS), cadherin 1 (CDH1), SRC proto‑oncogene, non‑receptor tyrosine kinase, twist family bHLH transcription factor 1 (TWIST1), ZW10 interacting kinetochore protein (ZWINT), PCNA clamp associated factor (KIAA0101) and androgen receptor, among which, five (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) were significantly upregulated and negatively correlated with miR‑1, were identified as key miR‑1 target genes in PCa. Additionally, it was investigated whether miR‑1 and its hub genes were associated with clinical features, including age, tumor status, residual tumor, lymph node metastasis, pathological T stage and prostate specific antigen level. Collectively the results suggest that miR‑1 may be involved in the progression of PCa, and consequently be a promising diagnostic marker. The ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’ may be crucial interactive pathways in PCa. Furthermore, PAICS, CDH1, TWIST1, ZWINT and KIAA0101 may serve as crucial miR‑1 target genes in PCa.

Introduction

Prostate cancer (PCa) remains the cancer with the highest incidence and third highest cause of cancer-associated mortality in males, with an estimated 161,360 new cases and 26,730 mortalities, according to 2017 statistics (1). A number of factors contribute to the survival of patients with PCa, particularly the disease stage at the time of diagnosis. Therefore, the early detection of PCa may improve clinical treatment and patient prognosis. Various techniques for the diagnosis of PCa have been developed; these include pathological biopsy, the gold standard in diagnosis, and other detection methods, including the prostate-specific antigen (PSA) assay (24), positron emission tomography (5), multiparametric magnetic resonance imaging (6,7), and urinary long non-coding RNA and microRNA (miRNA/miR) detection (8,9), which have also been used in clinical diagnosis. Despite progress, PCa still requires further novel diagnostic biomarkers to aid in its diagnosis.

miRNAs are known to have a crucial role in the regulation of tumor suppressor genes and oncogenes, resulting in the regulation of cell proliferation and apoptosis (10). miR-1 has been previously reported to be implicated in various tumors (11). The diagnostic value of miR-1 has been reported in breast cancer (12), colorectal cancer (13) and prostate recurrence (14). Details of the molecular mechanisms of miR-1 in diverse cancer types have been reported over the years. For example, the HOX transcript antisense RNA-miR-1-cyclin D2 axis was reported to promote thyroid cancer cell growth, invasion and migration (15). miR-1 was revealed to suppress colorectal cancer cell proliferation by inhibiting SMAD3-mediated tumor glycolysis (16). In bladder cancer, miR-1 suppressed cell proliferation, invasion and migration by upregulating secreted frizzled-related protein 1 expression (17). miR-1 was also reported to inhibit breast cancer progression by downregulating K-ras and metastasis associated lung adenocarcinoma transcript 1 (18). Regarding PCa, Chang et al (19) identified that the epidermal growth factor receptor was able to boost PCa bone metastasis by downregulating miR-1 and activating twist-related protein 1 (TWIST1). Stope et al (20) reported that heat-shock protein β-1 reduced the expression of miR-1, which restored oncogenic pathways in PCa cells.

Despite all the studies regarding miR-1 in PCa, to the best of our knowledge, no study has examined its clinical diagnostic significance via systematic meta-analysis and identified the potential target genes and pathways using bioinformatics methods. Therefore, it was necessary for the present study to conduct systematic meta-analysis and bioinformatics analysis to comprehensively evaluate the diagnostic value of miR-1, and examine the potential molecular modulatory mechanisms in PCa. The aim of the present study was to elucidate the diagnostic value of miR-1 in PCa in a more comprehensive way and pose a perspective into the potential molecular mechanism, which may contribute to clinical diagnosis and treatment.

In the present study, a meta-analysis using Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), ArrayExpress and data from published literature was conducted, to assess the clinical diagnostic value of miR-1 in PCa. By collecting the potential target genes of miR-1 and using bioinformatics analysis, the important targets and signaling pathways of miR-1 in PCa were identified. The present study may aid validation of the diagnostic value of miR-1 in PCa and provide insight into the theoretical molecular modulatory mechanisms for future studies.

Materials and methods

Collection of miR-1 expression data from TCGA

miRNA expression matrix was downloaded from TCGA (https://cancergenome.nih.gov/) (21). The expression data of miR-1-1 and miR-1-2 were included in the investigation. Samples missing miR-1-1 or miR-1-2 expression data were removed from the present research. All the data was normalized using log2 conversion. Additionally, mRNA data was downloaded from the portal of prostate adenocarcinoma in TCGA. The DESeq package (22) in the R program (version 3.30) was applied to obtain the differentially expressed genes (DEGs).

Retrieval of GEO and ArrayExpress PCa datasets

To analyze the expression level of miR-1 in PCa and non-tumor tissue, the GEO and ArrayExpress databases were searched. The key words were as follows: Prostat* and (malignan* OR cancer OR tumor OR tumour OR tumor OR neoplas* OR carcinoma). Microarray datasets that contained the miR-1 values were included. All the data in the datasets were calculated in the log2 scale for normalization. To collect DEGs in PCa following miR-1 pretreatment, the Gene Expression Omnibus (GEO) and ArrayExpress databases were additionally searched with the following retrieval strategies: Prostat* and (malignan* OR cancer OR tumor OR tumor OR neoplas* OR carcinoma) and (miR-1 OR miRNA-1 OR microRNA-1 OR miR1 OR miRNA1 OR microRNA1 OR ‘miR 1’ OR ‘miRNA 1’ OR ‘microRNA 1’ OR miR-1-3p OR miRNA-1-3p OR microRNA-1-3p OR miR-1-1 OR miR-1-2 OR miR1-1 OR miR1-2). Microarrays containing mRNA expression following miR-1 silencing or overexpression were enrolled for further analysis.

Retrieval of published literatures

PubMed (https://www.ncbi.nlm.nih.gov/pubmed/), Science Direct (https://www.sciencedirect.com/), Google Scholar (https://scholar.google.com/), Ovid (http://www.ovid.com), Wiley Online Library (https://onlinelibrary.wiley.com/), EMBASE (https://www.embase.com), Web of Science (https://clarivate.com/products/web-of-science/), Chong Qing VIP (http://qikan.cqvip.com/), CNKI (http://cnki.net/), Wan Fang (www.wanfangdata.com.cn) and China Biology Medicine Disc (http://www.sinomed.ac.cn/) were used for literature retrieval. The searching strategies were as follows: Prostat* and (malignan* OR cancer OR tumor OR tumor OR neoplas* OR carcinoma) and (miR-1 OR miRNA-1 OR microRNA-1 OR miR1 OR miRNA1 OR microRNA1 OR ‘miR 1’ OR ‘miRNA 1’ OR ‘microRNA 1’ OR miR-1-3p OR miRNA-1-3p OR microRNA-1-3p OR miR-1-1 OR miR-1-2 OR miR1-1 OR miR1-2). Studies for which the mean, standard deviation and case number of miR-1 in prostate cancer and non-tumor groups were available were further analyzed. Additionally, studies that provided the true positive, false positive, false negative and true negative values were included in the diagnostic meta-analysis.

Identification of putative miR-1 target genes

For the microarray data with silenced or overexpressed miR-1, the mRNA expression was analyzed and log2 fold change (FC) was calculated to select the DEGs. Log2FC<0 or log2FC>0 was adopted to screen the downregulated or upregulated genes in the miR-1 overexpressed or silenced microarrays, respectively. Additionally, DEGs were obtained from the TCGA data through the standard log2FC>1. Furthermore, the miRWalk 2.0 tool (http://zmf.umm.uni-heidelberg.de/mirwalk2/) (23), which links to 11 other online prediction databases, was used to predict the putative target genes of miR-1. Targets predicted by more than two datasets were finally selected. Genes that were included as DEGs from the GEO microarrays and TCGA, and the predicted target genes, were filtered further to collect the more specific target genes of miR-1. Finally, the validated target genes reported in the published literature were included in the genes for further analysis.

Bioinformatics analysis

To uncover the potential crucial signal pathways and target genes of miR-1 in PCa, Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using DAVID 6.8 (david-d.ncifcrf.gov/). P<0.05 was considered to indicate a statistically significant difference. The false discovery rate was adopted to reflect the rate of type I errors in multiple comparisons. A protein-protein interaction (PPI) network was plotted using the plugin stringAPP 1.10 in Cytoscape 3.50 (http://www.cytoscape.org/) (24), which has the ability to import the network from the STRING (string-db.org) database. Connection degrees >3 among the nodes were used to select the hub genes.

Validation of the hub genes

GEPIA (gepia.cancer-pku.cn), a newly developed interactive web server, was designed to analyze the RNA sequencing expression data of 9,736 tumors and 8,587 normal samples from the TCGA and the GTEx projects using a standard processing pipeline (25). Thus, GEPIA was used to validate the expression of the hub genes that were selected in the PPI. As miR-1 exerts it functions by specifically binding to its target genes, a Spearman's correlation analysis was conducted to validate the correlation between miR-1 and hub genes. The expression data of miR-1 and hub genes was downloaded from TCGA database. The expression data of miR-1 in TCGA included miR-1-1 and miR-1-2; therefore, the average expression of miR-1-1 and miR-1-2 was calculated to represent the expression of miR-1. The expression data of miR-1 and hub genes were normalized with log2 (x+1).

Clinical value of miR-1 and hub genes in PCa

The data of miR-1, hub genes and clinical phenotype of PCa were obtained from TCGA database. The information of age, cancer status, residual tumor, Tumor, Node, Metastasis stage, survival time, final status, PSA level, hormone therapy and drug response were collected. The expression of miR-1, hub genes and PSA value were normalized with log2 (x+1). The expression difference analysis of miR-1, phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthase (PAICS), cadherin 1 (CDH1), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), twist family bHLH transcription factor 1 (TWIST1), ZW10 interacting kinetochore protein (ZW10), ZWINT, PCNA clamp associated factor (KIAA0101) and androgen receptor (AR) among AR-dependent, castrate-resistant (AR-independent) PCa and normal tissues were performed. The correlation among miR-1, upregulated hub genes (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) and PSA was investigated. The association between miR-1, upregulated hub genes (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) and clinicopathological parameters was investigated using SPSS 23.0 (IBM Corp., Armonk, NY, USA).

Statistical analysis

The standard mean deviation (SMD) was used to evaluate the miR-1 expression differences between PCa and adjacent normal tissues via continuous variable meta-analysis in Stata 14.0 (StataCorp LLC, College Station, TX, USA). The fixed effects model was adopted to analyze the SMD if there was no heterogeneity among the included studies (I2>50; P<0.05). Otherwise, the random effects model was used. Begg and Deeks funnel plots were applied to test the publication bias of the included studies. Additionally, a sensitivity analysis was performed to demonstrate the influence of individual studies on the whole dataset. Summary receiver operating characteristic (SROC) curves, Fagan plots and forest plots of sensitivity, specificity, positive likelihood ratios, negative likelihood ratios, diagnostic scores, in addition to odds ratios depicted through Stata 14.0 (StataCorp LLC), were performed to comprehensively evaluate the diagnostic value of miR-1 in PCa. Furthermore, bivariate boxplots and likelihood matrices were plotted to illustrate the sensitivity, specificity, positive likelihood ratio and negative likelihood ratio in a plane coordinate system. SPSS 23.0 (IBM Corp.) and GraphPad Prism 6 (GraphPad Software, Inc., La Jolla, CA, USA) were used for the clinicopathological parameter analysis and to display the results, respectively. Spearman's correlation analysis was used to analyze the correlation among miR-1, hub genes and PSA level. Student's t test was applied for comparison of two groups and the log-rank test was used for the survival analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression level of miR-1 in PCa

For all the included studies, the combined SMD was −0.31 [95% confidence interval (CI), −0.60 to 0.22] based on the random effects model (I2=87.8%; Fig. 1). Publication bias was not observed in the funnel plots, as it was basically symmetrical (Fig. 2). Significant heterogeneity was observed in the forest plot. As a result, a sensitivity analysis was performed, and it was considered that GSE54516 may cause heterogeneity (Fig. 3). Following removal of GSE54516, the combined SMD was −0.42 (95% CI, −0.52 to −0.32) with an I2=80.4%, which indicated downregulation (Fig. 4).

Diagnostic value of miR-1 in PCa

An SROC curve was used to evaluate the pooled diagnostic value of miR-1 in the included studies. The SROC curve demonstrated that the pooled area under the curve (AUC) was 0.73 (95% CI, 0.69 to 0.77; Fig. 5), which suggested a moderate diagnostic efficacy of miR-1 in PCa. The combined sensitivity and specificity were 0.77 (95% CI, 0.62 to 0.88) and 0.57 (95% CI, 0.41 to 0.72), respectively (Fig. 6). The positive diagnostic likelihood ratio (DLR) and negative DLR were 1.82 (95% CI, 1.36 to 2.43) and 0.40 (95% CI, 0.26 to 0.60), respectively (Fig. 7). The diagnostic score and odds ratios were also calculated and were 1.53 (95% CI, 0.97 to 2.08) and 4.60 (95% CI, 2.63 to 8.04), respectively (Fig. 8). A Fagan plot was used to demonstrate how much the result on the diagnostic test alters the probability that a patient has PCa. The positive and negative posterior probabilities were 31 and 9%, respectively, and the pre-test probability was 20%. (Fig. 9). Deek's funnel plot was created to check for the potential publication bias of included studies. As demonstrated in Fig. 10, the Deek's funnel plot obtained a P-value of 0.85, which suggested there was no publication bias in the diagnostic meta-analysis. A bivariate boxplot was additionally constructed to assess distributional properties of sensitivity vs. specificity and to identify potential outliers (Fig. 11). An index test was used to display informativeness of measured test, which displayed the distribution of positive and negative likelihood ratio in the diagnostic meta-analysis (Fig. 12). Collectively, no publication bias existed in the present study and downregulated miR-1 demonstrated moderate diagnostic accuracy in PCa.

Collection of possible target genes

In the GEO microarray datasets, GSE26032 and GSE31620, which included transfection of pre-miR oligos in prostate carcinoma cell lines to overexpress, were finally included. The expression data of the genes was extracted and the log2FC was calculated. With the standard log2FC<0, the downregulated genes were collected. A total of 1,239 genes were finally gathered following intersecting the downregulated genes in GSE26032 and GSE31620, which contained two and three paired samples, respectively. For TCGA DEGs, the log2FC>1 was restricted and 684 genes were obtained. Furthermore, 10,025 predicted target genes were gathered using the 12 online prediction tools. To improve the specificity of the putative target genes of miR-1, the GEO downregulated genes were intersected, TCGA DEGs, in addition to the predicted genes, and 45 putative significant genes were finally obtained for further bioinformatics analysis (Table I).

Table I.

54 putative potential targets of microRNA-1.

Table I.

54 putative potential targets of microRNA-1.

Gene symbolOfficial name
SEL1L3SEL1L family member 3
PPM1HProtein phosphatase, Mg2+/Mn2+ dependent 1H
LMNB1Lamin B1
NKX3-1NK3 homeobox 1
SLC4A4Solute carrier family 4 member 4
AP1S1Adaptor related protein complex 1 subunit sigma 1
PPIBPeptidylprolyl isomerase B
PSAT1Phosphoserine aminotransferase 1
PABPC1Poly(A) binding protein cytoplasmic 1
BCAMBasal cell adhesion molecule (Lutheran blood group)
AGA Aspartylglucosaminidase
TMEM97Transmembrane protein 97
SSR1Signal sequence receptor subunit 1
GALNT1Polypeptide N-acetylgalacto-saminyltransferase 1
SEC23BSec23 homolog B, coat complex II component
GOLM1Golgi membrane protein 1
DSG2Desmoglein 2
APLP2Amyloid β (A4) precursor-like protein 2
GCNT1Glucosaminyl (N-acetyl) transferase 1, core 2
EPB41L4BErythrocyte membrane protein band 4.1 like 4B
CLCN3Chloride voltage-gated channel 3
PDIA6Protein disulfide isomerase family A member 6
SLC27A2Solute carrier family 27 member 2
MYO6Myosin VI
DSC2Desmocollin 2
ABCC4ATP binding cassette subfamily C member 4
SLC45A2Solute carrier family 45 member 2
NUP210Nucleoporin 210
PAICS Phosphoribosylaminoimidazole carboxylase and phosphoribo-sylaminoimidazolesuccino-carboxamide synthase
CENPFCentromere protein F
ACSL3Acyl-CoA synthetase long chain family member 3
PPP3CAProtein phosphatase 3 catalytic subunit α
ABHD2Abhydrolase domain containing 2
NCAPD3Non-SMC condensin II complex subunit D3
SEL1L3SEL1L family member 3
MAP7Microtubule associated protein 7
ZWINTZW10 interacting kinetochore protein
NAAAN-acylethanolamine acid amidase
TUSC3Tumor suppressor candidate 3
PCLAFPCNA clamp associated factor
ETO2Neuropilin and tolloid like 2
TPD52Tumor protein D52
CDH1Cadherin 1
CADM1Cell adhesion molecule 1
EIF2AK1Eukaryotic translation initiation factor 2 α kinase 1
PMEPA1Prostate transmembrane protein, androgen induced 1
PNPPurine nucleoside phosphorylase
PTMAProthymosin α
ARAndrogen receptor
SRCSRC proto-oncogene, non-receptor tyrosine kinase
TWIST1Twist family bHLH transcription factor 1
SlugSnail family transcriptional repressor 2
KLF4Kruppel like factor 4
XPO6Exportin 6
TWF1Twinfilin actin binding protein 1
Bioinformatics analysis

A total of 45 promising overlapping target genes were collected for the bioinformatics analysis (Table II; Fig. 13). The GO annotation elucidated that the promising target genes of miR-1 were closely associated with ‘the response to the drug’, ‘adherens junction organization’ and ‘androgen receptor signaling pathway’ in the ‘biological process’ (BP) domain. For the ‘cellular component’ (CC) domain, the promising miR-1 targets were mostly associated with the ‘perinuclear region of cytoplasm’, ‘extracellular exosome’ and ‘membrane’. In the ‘molecular function’ (MF) domain, the putative miR-1 targets were mostly enriched in the ‘androgen receptor activity’, the ‘transcription factor binding’ and ‘cadherin binding involved in cell-cell adhesion’. In addition, the KEGG pathway analysis demonstrated that the ‘protein processing pathway in the endoplasmic reticulum’ was the most significant. Using the PPI network, the construction of seven hub genes (PAICS, CDH1, SRC, TWIST1, ZWINT, KIAA0101 and AR) were identified to act as the potential key targets of miR-1 in PCa (Fig. 14).

Table II.

Significant GO and KEGG pathways of the overlapping genes.

Table II.

Significant GO and KEGG pathways of the overlapping genes.

CategoryTermCountP-valueFDR
GOTERM_BP_DIRECTGO:0042493~response to drug  60.001872.677129
GOTERM_BP_DIRECTGO:0034332~adherens junction organization  30.0052057.287529
GOTERM_BP_DIRECTGO:0030521~androgen receptor signaling pathway  30.0063618.837711
GOTERM_BP_DIRECTGO:0010628~positive regulation of gene expression  50.0070789.786328
GOTERM_BP_DIRECT GO:0007156~homophilic cell adhesion via plasma membrane adhesion molecules  40.01097414.78529
GOTERM_BP_DIRECTGO:0086073~bundle of His cell-Purkinje myocyte adhesion involved in cell communication  20.01738422.45141
GOTERM_BP_DIRECT GO:0044539~long-chain fatty acid import  20.02025225.66973
GOTERM_BP_DIRECTGO:0014067~negative regulation of phosphatidylinositol 3-kinase signaling  20.02880834.54625
GOTERM_BP_DIRECT GO:0006886~intracellular protein transport  40.03144437.07541
GOTERM_BP_DIRECT GO:0098911~regulation of ventricular cardiac muscle cell action potential  20.03164437.26326
GOTERM_BP_DIRECTGO:0071456~cellular response to hypoxia  30.03196337.56294
GOTERM_BP_DIRECTGO:2000679~positive regulation of transcription regulatory region DNA binding  20.04290547.05066
GOTERM_CC_DIRECT GO:0048471~perinuclear region of cytoplasm129.07E-070.001083
GOTERM_CC_DIRECT GO:0070062~extracellular exosome213.29E-050.039319
GOTERM_CC_DIRECT GO:0016020~membrane170.0002180.25958
GOTERM_CC_DIRECT GO:0005913~cell-cell adherens junction  70.0002720.324715
GOTERM_CC_DIRECT GO:0005789~endoplasmic reticulum membrane  80.00974311.03435
GOTERM_CC_DIRECT GO:0016323~basolateral plasma membrane  40.01394715.44149
GOTERM_CC_DIRECTGO:0032587~ruffle membrane  30.02211423.43573
GOTERM_CC_DIRECTGO:0000139~Golgi membrane  60.02430725.46094
GOTERM_CC_DIRECTGO:0031965~nuclear membrane  40.0262127.17933
GOTERM_CC_DIRECT GO:0005783~endoplasmic reticulum  70.02734228.18388
GOTERM_CC_DIRECT GO:0034663~endoplasmic reticulum chaperone complex  20.03036530.80375
GOTERM_CC_DIRECTGO:0005794~Golgi apparatus  70.03258832.67453
GOTERM_CC_DIRECTGO:0016021~integral component of membrane220.03297232.99373
GOTERM_CC_DIRECT GO:0005737~cytoplasm220.03709336.3252
GOTERM_MF_DIRECTGO:0004882~androgen receptor activity  20.00903712.08615
GOTERM_MF_DIRECT GO:0008134~transcription factor binding  50.01054512.91881
GOTERM_MF_DIRECTGO:0098641~cadherin binding involved in cell-cell adhesion  50.0113216.85447
GOTERM_MF_DIRECTGO:0050839~cell adhesion molecule binding  30.01507619.38597
GOTERM_MF_DIRECTGO:0008022~protein C-terminus binding  40.01757919.79801
GOTERM_MF_DIRECTGO:0086083~cell adhesive protein binding involved in bundle of His cell-Purkinje myocyte communication  20.01799323.42536
GOTERM_MF_DIRECTGO:0005102~receptor binding  50.02172725.48576
GOTERM_MF_DIRECT GO:0102391~decanoate-CoA ligase activity  20.0239226.92366
GOTERM_MF_DIRECT GO:0008013~β-catenin binding  30.02548438.00481
GOTERM_MF_DIRECT GO:0004467~long-chain fatty acid-CoA ligase activity  20.03858457.75621
KEGG_PATHWAYhsa04141:Protein processing in endoplasmic reticulum  50.00446910.44355

[i] GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; FDR, false discovery rate; BP, biological process; CC, cellular component; MF, molecular function.

Validation of the hub genes

The expression of the seven hub genes (PAICS, CDH1, SRC, TWIST1, ZWINT, KIAA0101 and AR) was analyzed in the PCa and adjacent normal samples from TCGA and GTEx. A total of 492 PCa and 152 normal tissue samples were enrolled in the expression validation. Among the seven hub genes, it was observed that five of them (PAICS, CDH1, KIAA0101, TWIST1, and ZWINT) were significantly upregulated in PCa samples (Fig. 15A-E), which gave them more potential to be the key target genes of miR-1 in PCa. The other two hub genes (SRC and AR) did not demonstrate statistical significance between PCa and normal tissues based on the current data (Fig. 15F and G). Spearman's correlation analysis between miR-1 and the five up-regulated hub genes (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) was performed (Fig. 16). A significant negative correlation was identified between miR-1 and PAICS, ZWINT, in addition to KIAA0101 (P<0.001; Fig. 16A-C), which supported their specific binding. Regarding CDH1 and TWIST1, a trend of negative correlation with miR-1 was observed (Fig. 16D and E). However, no statistical significance was revealed.

Clinical significance of miR-1 and hub genes in PCa

The alterations of miR-1/hub genes among 31 AR-dependent, 9 castrate-resistant PCa and 52 normal tissues were compared. For AR-dependent and castrate-resistant PCa, miR-1 was significantly downregulated; whereas, PAICS, TWIST1, ZWINT and KIAA0101 were significantly upregulated compared with normal tissues (P<0.001; Fig. 17A and B). Regarding CDH1, SRC and AR, the expression of SRC and AR only demonstrated significant increase in AR-dependent PCa not castrate-resistant PCa, while CDH1 did not reveal any alterations in AR-dependent and castrate-resistant PCa compared with normal tissues (Fig. 17A and B). However, for comparison between AR-dependent and castrate-resistant PCa, no statistically significant expression difference of miR-1 or hub genes was observed (Fig. 17C). Further studies with larger samples are required to elucidate the alterations of miR-1 or hub genes among AR-dependent, castrate-resistant PCa and normal tissues. The correlation among miR-1, upregulated hub genes and PSA level was investigated. In Fig. 18, the expression of miR-1 was negatively correlated with PSA level. Regarding the five upregulated hub genes, statistically significant negative correlation among PAICS, ZWINT, KIAA0101 and PSA level was identified. However, for CDH1 and TWIST1, no significantly negative correlation with PSA level was revealed. A total of 491 PCa samples were included for the clinicopathological features analysis. As demonstrated in Table III, the association among miR-1, upregulated hub genes and clinicopathological parameters is presented. In summary, miR-1 was significantly associated with cancer status, distant metastasis and lymph node metastasis and CDH1 was identified to be associated with age, cancer status and residual tumor. TWIST1 was only associated with pathological T stage. Furthermore, ZWINT was associated with cancer status, residual tumor, lymph node metastasis and pathological T stage; whereas, KIAA0101 was associated with age, cancer status, residual tumor, lymph node metastasis and pathological T stage. However, regarding PAICS, no statistically significant association with clinicopathological features was identified, and survival analysis of miR-1 and hub genes did not demonstrate statistical significance (data not shown).

Table III.

Association between miR-1/hub genes and clinicopathological parameters in prostate cancer.

Table III.

Association between miR-1/hub genes and clinicopathological parameters in prostate cancer.

A, miR-1

Clinicopathological featurenExpression (mean ± SD)t-testP-value
Age (years) 1.6050.109
  <601998.759±1.229
  ≥602928.569±1.332
Tumor status −2.3170.021a
  With tumor938.391±1.328
  Tumor-free3108.746±1.286
Residual tumor 1.8360.067
  No3118.728±1.255
  Yes1508.495±1.318
Distant metastasis −4.4550.000a
  No4514.617±2.286
  Yes28.677±1.283
Lymph node metastasis 2.7640.006a
  No3428.713±1.251
  Yes778.281±1.172
Pathological T 1.8690.062
  T1 + T21868.782±1.183
  T3 + T42988.561±1.316

B, PAICS

Clinicopathological featurenExpression (mean ± SD)t-testP-value

Age (years) 0.480.631
  <601994.368±0.489
  ≥602924.347±0.488
Tumor status 1.1560.249
  With tumor934.409±0.576
  Tumor-free3104.340±0.473
Residual tumor −0.1530.878
  No3114.358±0.488
  Yes1504.365±0.493
Distant metastasis 1.7880.075
  No4514.350±0.486
  Yes24.966±0.697
Lymph node metastasis 0.2130.832
  No3424.356±0.509
  Yes774.343±0.406
Pathological T −1.1930.233
  T1 + T21864.323±0.485
  T3 + T42984.377±0.487

C, CDH1

Clinicopathological featurenExpression (mean ± SD)t-testP-value

Age (years) 2.40.017a
  <601996.593±0.710
  ≥602926.431±0.754
Tumor status −3.1350.002a
  With tumor936.306±1.018
  Tumor-free3106.586±0.658
Residual tumor 2.9670.003a
  No3116.583±0.636
  Yes1506.338±0.912
Distant metastasis −1.1920.234
  No4516.490±0.723
  Yes25.880±0.697
Lymph node metastasis 1.1850.237
  No3426.521±0.681
  Yes776.409±1.003
Pathological T 1.6310.104
  T1+T21866.567±0.646
  T3+T42986.454±0.793

D, TWIST1

Clinicopathological featurenExpression (mean ± SD)t-testP-value

Age (years) −0.5490.583
  <601992.307±1.131
  ≥602922.363±1.107
Tumor status 1.2740.203
  With tumor932.404±1.198
  Tumor-free3102.240±1.056
Residual tumor −1.6360.103
  No3112.271±1.067
  Yes1502.454±1.241
Distant metastasis 1.4440.150
  No4512.329±1.110
  Yes23.463±0.032
Lymph node metastasis −1.6290.104
  No3422.271±1.067
  Yes772.501±1.329
Pathological T −2.2440.025a
  T1+T21862.200±1.053
  T3+T42982.434±1.154

E, ZWINT

Clinicopathological featurenExpression (mean ± SD)t-testP-value

Age (years) −1.4820.139
  <601992.279±0.693
  ≥602922.375±0.715
Cancer status 3.4950.001a
  With tumor932.570±0.839
  Tumor-free3102.241±0.645
Residual tumor −3.4580.001a
  No3112.255±0.641
  Yes1502.505±0.764
Distant metastasis 0.6290.643
  No4512.339±0.693
  Yes23.629±2.902
Lymph node metastasis −3.1930.002a
  No3422.287±0.662
  Yes772.607±0.821
Pathological T −4.940.000a
  T1+T21862.157±0.526
  T3+T42982.445±0.758

F, KIAA0101

Clinicopathological featurenExpression (mean ± SD)t-testP-value

Age (years) −2.2840.023a
  <601991.067±0.506
  ≥602921.181±0.590
Cancer status 3.410.001a
  With tumor931.322±0.692
  Tumor-free3101.059±0.495
Residual tumor −3.1880.002a
  No3111.063±0.498
  Yes1501.249±0.624
Distant metastasis 0.9580.514
  No4511.135±0.553
  Yes22.353±1.799
Lymph node metastasis −2.8060.006a
  No3421.118±0.532
  Yes771.347±0.671
Pathological T −6.1850.000a
  T1+T21860.960±0.405
  T3+T42981.243±0.599

a Statistically significant. miRNA/miR, microRNA; SD, standard deviation; PAICS, phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthase; CDH1, cadherin 1; TWIST1, twist family bHLH transcription factor 1; ZWINT, ZW10 interacting kinetochore protein; KIAA0101, PCNA clamp associated factor.

Discussion

PCa is the cancer type that results in the highest worldwide morbidity in the male population (1). Searching for novel diagnostic biomarkers and examining the molecular mechanisms in PCa are of great importance for its clinical diagnosis and treatment. The present study aimed to evaluate the diagnostic value of miR-1 and identify the key target genes and signaling pathways in PCa.

Using comprehensive meta-analysis of expression data from the GEO, ArrayExpress, TCGA and published literature, it was identified that the expression of miR-1 was significantly downregulated in PCa compared with the adjacent normal tissues. It was demonstrated that miR-1 has a moderate diagnostic value in PCa (AUC, 0.73; sensitivity, 0.77; specificity, 0.57; odds ratio, 4.60). To the best of our knowledge, there is only one study, conducted by Pashaei et al (26), which has identified downregulated miR-1 in recurrent PCa using a meta-analysis. However, there were only six GEO datasets included in the meta-analysis (26). A larger sample may improve the integration and conviction of the study. In the present study, 27 GEO datasets, two published studies, one ArrayExpress microarray and two sets of TCGA data were analyzed.

Based on the online prediction databases, the potential target genes of miR-1 were collected to further examine the key enriched metabolic pathways in PCa. Subsequently, GO analysis revealed the enriched pathways in three categories. In ‘BP’, the ‘response to drug’, ‘adherens junction organization’, and ‘androgen receptor signaling’ pathways were the top three pathways. In ‘CC’, the top three items were the ‘perinuclear region of the cytoplasm’, the ‘extracellular exosome’, and the ‘membrane’. In the ‘MF’, the target genes were primarily enriched in ‘androgen receptor activity’, ‘transcription factor binding’ and ‘cadherin binding in cell-cell adhesion’. Furthermore, KEGG analysis identified a significant pathway, namely ‘protein processing in the endoplasmic reticulum’. Among the above pathways discovered, it was identified that the ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’ and ‘protein processing in the endoplasmic reticulum’ have been previously reported. The ‘androgen receptor signaling pathway’ has been implicated in the therapeutic strategies of PCa (2729). Regarding the ‘androgen receptor activity’, Ylitalo et al (30) documented that it was involved in castrate-resistant PCa bone metastases. The regulation of androgen receptor activity was additionally discovered to be associated with therapy resistance and disease progression in PCa (3133). Furthermore, transcription factor binding sites were additionally identified to be a crucial reaction domain in PCa, which may aid the explanation of the underlying regulatory mechanism (34,35). Regarding KEGG pathway analysis, Kojima et al (36) additionally revealed that ‘protein processing in the endoplasmic reticulum’ was a significant miR-143/145 regulated signal pathway in PCa, which may provide novel insights into the potential mechanism of PCa oncogenesis and metastasis. In summary, the enriched pathways, namely, the ‘androgen receptor signaling pathway’, ‘androgen receptor activity’, ‘transcription factor binding’, and ‘protein processing in the endoplasmic reticulum’, possess great potential to be involved the regulatory mechanisms of miR-1 in PCa. For the other pathways that have not been reported in PCa, further examination of their role in the regulatory mechanisms of miR-1 in PCa is worth pursuing.

Using the PPI network, seven hub genes (PAICS, CDH1, SRC, TWIST1, ZWINT, KIAA0101 and AR) that may be the key target genes of miR-1 in PCa were identified. As miR-1 was downregulated in PCa, according to the meta-analysis, it was hypothesized that its target genes are upregulated accordingly. Thus, the expression of five hub genes was validated based on TCGA and GTEx data. It was identified that the five hub genes (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) were significantly upregulated in PCa. Since miR-1 was able to specifically bind to its targets, it was hypothesized that there is a correlation between miR-1 and its targets. As expected, there was significant negative correlation between miR-1 expression, and PAICS, ZWINT and KIAA0101 expression. Regarding CDH1 and TWIST1, a there was no significant correlation with miR-1. AR-dependent and castrate-resistant PCa may have different miRNA/mRNA expression profiles. Therefore, the expression of miR-1 and hub genes among AR-dependent, castrate-resistant PCa and normal tissues was investigated. In summary, it was identified that there was a trend that miR-1 was downregulated, while hub genes were upregulated in AR-dependent and castrate-resistant PCa compared with normal tissues. However, the expression difference of miR-1 and hub genes between AR-dependent and castrate-resistant PCa remains unclear. Studies with a larger sample size are required to elucidate the expression profiling alterations of miR-1/hub genes in AR-dependent and castrate-resistant PCa. Considering that PSA level was a useful specific marker in the clinical diagnosis of PCa, the correlation between miR-1/hub genes and PSA level was investigated. Notably, it was identified that miR-1 was negatively correlated with PSA level, while hub genes, PAICS, ZWINT and KIAA0101, were positively correlated with PSA level. Furthermore, it was identified that miR-1 and its hub genes were significantly associated with specific clinical phenotypes including age, tumor status, residual tumor, lymph node metastasis and pathological T stage. These results may pose a perspective for the clinical application of miR-1 and its hub genes. Combining PSA with miR-1/hub genes may be useful for the clinical diagnosis of PCa, and the involved molecular mechanism of miR-1 and its hub genes may shed light on novel therapies for patients with PCa. Consequently, the five upregulated genes, PAICS, CDH1, TWIST1, ZWINT and KIAA0101 were focused on for a more detailed discussion.

PAICS encodes a bifunctional enzyme. The octameric structure of the bifunctional enzyme PAICS in purine biosynthesis may contribute to PAICS-specific inhibitor design, which may be used in cancer chemotherapy (37). PAICS has been reported as a therapeutic target in breast cancer (38). Furthermore, increased PAICS was additionally identified to be associated with poor prognosis in lung cancer, which made it a promising prognostic biomarker (39). Cifola et al (40) first identified that PAICS was a mutated gene in melanoma. In the study conducted by Chakravarthi et al (41), the role of PAICS in the proliferation and invasion of PCa cell was evaluated, implying that PAICS was a therapeutic target. Collectively, PAICS may be a promising target in PCa. However, its association with miR-1 still requires experimental validation.

CDH1, additionally termed uvomorulin and CDHE, encodes a classical cadherin of the cadherin superfamily. Loss of or reduced expression of CDH1 is thought to contribute to more invasive tumors (42). A previous study discovered that CDH1 polymorphisms may be a prognostic indicator in non-metastatic laryngeal cancer (43). Jiao et al (44) identified that aberrant CDH1 was able to predict a poor prognosis for patients with pancreatic cancer. In addition, CDH1 was additionally identified to be associated with the risk of lung cancer (45), breast cancer (46), and gastric cancer (47), and may be a potential drug target. In PCa, there was additionally a meta-analysis that evaluated CDH1-60 C/A polymorphism as a risk factor in the development of PCa (48). In summary, CDH1 was a promising key target in PCa; however, it requires further experimental evaluation.

TWIST1 was reported to be implicated in cell lineage determination and differentiation. TWIST1 has been revealed to serve as an effective target for cancer metastasis and chemoresistance (49). In pancreatic cancer, TWIST1 was identified as a therapeutic target and prognostic marker for tumor metastasis (50). TWIST1 was additionally a target in lung cancer, which was correlated with the inhibition of proliferation, epithelial-mesenchymal transition and metastasis (51). Furthermore, overexpression of TWIST1 was additionally identified to determine lung cancer chemoresistance and prognosis (52). TWIST1 was additionally documented to be associated with the invasion and metastasis of gastric cancer (53,54), and was a potential prognostic marker in colorectal cancer (5557). Regarding PCa, numerous studies have reported that TWIST1 was involved in progression, including cancer invasion, migration and migration (5863). As for the association between miR-1 and TWIST1, Chang et al (19) discovered that epidermal growth factor receptor translocation promotes the bone metastasis of PCa by downregulating miR-1, which directly increases the expression of TWIST1. Given the above information, TWIST1 may be considered a key target of miR-1 in PCa. This finding requires further experimental research to validate it.

ZWINT encodes a protein that is involved in kinetochore function. Xu et al (64) identified that ZWINT was increased in ovarian cancer and correlated with worse overall survival in patients with ovarian cancer. In bladder cancer, Ho et al (65) observed that ZWINT was associated with a cell proliferation marker. ZWINT was additionally identified to be significantly correlated with the clinical outcome of chronic lymphocytic leukemia (66). The association between ZWINT and PCa has not been reported in previously published literature. Further evidence is required to further elucidate the function of ZWINT in PCa.

KIAA0101 has been widely reported in the progression of various cancer types. For example, in hepatocellular carcinoma, overexpressed KIAA0101 was significantly associated with distant metastasis, advanced stage, early tumor recurrence and poor prognosis, which made it a potential therapeutic target (6769). In gastric cancer, KIAA0101 was identified to be involved in the proliferation and invasion of PCa cells (70,71). In addition, overexpression of KIAA0101 was indicated to predict poor survival in lung cancer (72), esophageal cancer (73) and renal cancer (74). As for its role in PCa, to the best of our knowledge, no study at present has elucidated the association between KIAA0101 and PCa. Therefore, further studies are required to determine the precise role of KIAA0101 in PCa.

Collectively, it was identified that miR-1 was downregulated in PCa and was negatively correlated with PSA level. miR-1 was additionally associated with the tumor status, metastasis and lymph node metastasis of PCa. A total of seven hub genes, PAICS, CDH1, SRC, TWIST1, ZWINT, KIAA0101 and AR, were identified. Five of them (PAICS, CDH1, TWIST1, ZWINT and KIAA0101) were significantly upregulated in PCa and were negatively correlated with miR-1, which made them potential key target genes of downregulated miR-1 in PCa. The hub genes were associated with certain clinical features, including age, tumor status, residual tumor, lymph node metastasis, pathological T stage and PSA level. Using a review of published studies, further evidence was provided to support PAICS, CDH1 and TWIST1 as potential target genes of miR-1 in PCa. For ZWINT and KIAA0101, there is a lack of sufficient studies to demonstrate their role in PCa, and as such, they require further exploration. All the analyses in the present study were based on online database sources at the bioinformatics level. Therefore, further experimental studies are required to validate these genes. Nevertheless, the results obtained in the present study may aid the clinical diagnosis of PCa and provide insight into the molecular mechanisms of PCa.

Acknowledgements

Not applicable.

Funding

The present study was funded by the Promoting Project of Basic Capacity for Young and Middle-aged University Teachers in Guangxi (grant no. KY2016LX034). The funders had no role in study design, data collection and analysis, preparation of the manuscript, or decision to publish.

Availability of data and materials

All the data and materials used in the study are currently available at Pubmed (https://www.ncbi.nlm.nih.gov/pubmed/), GEO (https://www.ncbi.nlm.nih.gov/geo/), ArrayExpress (https://www.ebi.ac.uk/arrayexpress/), TCGA (https://cancergenome.nih.gov/), miRWalk 2.0 (http://zmf.umm.uni-heidelberg.de/apps/zmf/mirwalk2/), DAVID (https://david.ncifcrf.gov/) and STRING (https://string-db.org/) databases.

Authors' contributions

HB-Y, SH-L and GC designed the study and revised the manuscript. ZC-X, JC-H, LJ-Z, BL-G and DY-W contributed to the collection and analysis of the data, in addition to the writing of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

PCa

prostate cancer

PSA

prostate-specific antigen

miRNA/miR

microRNA

GEO

Gene Expression Omnibus

TCGA

The Cancer Genome Atlas

SMD

standard mean deviation

SROC

summary receiver operating characteristic

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

PPI

protein-protein interaction

AUC

area under the curve

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Lavi A and Cohen M: Prostate cancer early detection using psa-current trends and recent updates. Harefuah. 156:185–188. 2017.(In Hebrew). PubMed/NCBI

3 

Filella X and Foj L: Prostate cancer detection and prognosis: From prostate specific antigen (PSA) to exosomal biomarkers. Int J Mol Sci. 17:pii: E1784. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Kirby R: The role of PSA in detection and management of prostate cancer. Practitioner. 260(17–21): 32016.

5 

Bednarova S, Lindenberg ML, Vinsensia M, Zuiani C, Choyke PL and Turkbey B: Positron emission tomography (PET) in primary prostate cancer staging and risk assessment. Transl Androl Urol. 6:413–423. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Wallis CJD, Haider MA and Nam RK: Role of mpMRI of the prostate in screening for prostate cancer. Transl Androl Urol. 6:464–471. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Johnson DC and Reiter RE: Multi-parametric magnetic resonance imaging as a management decision tool. Transl Androl Urol. 6:472–482. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Balacescu O, Petrut B, Tudoran O, Feflea D, Balacescu L, Anghel A, Sirbu IO, Seclaman E and Marian C: Urinary microRNAs for prostate cancer diagnosis, prognosis, and treatment response: Are we there yet? Wiley Interdiscip Rev RNA. 8:2017. View Article : Google Scholar : PubMed/NCBI

9 

Martignano F, Rossi L, Maugeri A, Gallà V, Conteduca V, De Giorgi U, Casadio V and Schepisi G: Urinary RNA-based biomarkers for prostate cancer detection. Clin Chim Acta. 473:96–105. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Mishra S, Yadav T and Rani V: Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit Rev Oncol Hematol. 98:12–23. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Han C, Shen JK, Hornicek FJ, Kan Q and Duan Z: Regulation of microRNA-1 (miR-1) expression in human cancer. Biochim Biophys Acta Gene Regul Mech. 1860:227–232. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Rigaud VO, Ferreira LR, Ayub-Ferreira SM, Ávila MS, Brandão SM, Cruz FD, Santos MH, Cruz CB, Alves MS, Issa VS, et al: Circulating miR-1 as a potential biomarker of doxorubicin-induced cardiotoxicity in breast cancer patients. Oncotarget. 8:6994–7002. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Wu X, Li S, Xu X, Wu S, Chen R, Jiang Q, Li Y and Xu Y: The potential value of miR-1 and miR-374b as biomarkers for colorectal cancer. Int J Clin Exp Pathol. 8:2840–2851. 2015.PubMed/NCBI

14 

Wei W, Leng J, Shao H and Wang W: MiR-1, a potential predictive biomarker for recurrence in prostate cancer after radical prostatectomy. Am J Med Sci. 353:315–319. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Di W, Li Q, Shen W, Guo H and Zhao S: The long non-coding RNA HOTAIR promotes thyroid cancer cell growth, invasion and migration through the miR-1-CCND2 axis. Am J Cancer Res. 7:1298–1309. 2017.PubMed/NCBI

16 

Xu W, Zhang Z, Zou K, Cheng Y, Yang M, Chen H, Wang H, Zhao J, Chen P, He L, et al: MiR-1 suppresses tumor cell proliferation in colorectal cancer by inhibition of Smad3-mediated tumor glycolysis. Cell Death Dis. 8:e27612017. View Article : Google Scholar : PubMed/NCBI

17 

Shang A, Yang M, Shen F, Wang J, Wei J, Wang W, Lu W and Wang C and Wang C: MiR-1-3p Suppresses the proliferation, invasion and migration of bladder cancer cells by up-regulating sfrp1 expression. Cell Physiol Biochem. 41:1179–1188. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Liu R, Li J, Lai Y, Liao Y, Liu R and Qiu W: Hsa-miR-1 suppresses breast cancer development by down-regulating K-ras and long non-coding RNA MALAT1. Int J Biol Macromol. 81:491–497. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Chang YS, Chen WY, Yin JJ, Sheppard-Tillman H, Huang J and Liu YN: EGF Receptor promotes prostate cancer bone metastasis by downregulating mir-1 and activating TWIST1. Cancer Res. 75:3077–3086. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Stope MB, Stender C, Schubert T, Peters S, Weiss M, Ziegler P, Zimmermann U, Walther R and Burchardt M: Heat-shock protein HSPB1 attenuates microRNA miR-1 expression thereby restoring oncogenic pathways in prostate cancer cells. Anticancer Res. 34:3475–3480. 2014.PubMed/NCBI

21 

Deng M, Bragelmann J, Schultze JL and Perner S: Web-TCGA: An online platform for integrated analysis of molecular cancer data sets. BMC Bioinformatics. 17:722016. View Article : Google Scholar : PubMed/NCBI

22 

Anders S and Huber W: Differential expression analysis for sequence count data. Genome Biol. 11:R1062010. View Article : Google Scholar : PubMed/NCBI

23 

Dweep H and Gretz N: miRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat Methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

24 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Pashaei E, Pashaei E, Ahmady M, Ozen M and Aydin N: Meta-analysis of miRNA expression profiles for prostate cancer recurrence following radical prostatectomy. PLoS One. 12:e01795432017. View Article : Google Scholar : PubMed/NCBI

27 

Gravina GL, Festuccia C, Bonfili P, Di Staso M, Franzese P, Ruggieri V, Popov VM, Tombolini V, Masciocchi C, Carosa E, et al: Strategies for imaging androgen receptor signaling pathway in prostate cancer: Implications for hormonal manipulation and radiation treatment. Biomed Res Int. 2013:4605462013.PubMed/NCBI

28 

Attard G, Richards J and de Bono JS: New strategies in metastatic prostate cancer: Targeting the androgen receptor signaling pathway. Clin Cancer Res. 17:1649–1657. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Wang L, Song G, Chang X, Tan W, Pan J, Zhu X, Liu Z, Qi M, Yu J and Han B: The role of TXNDC5 in castration-resistant prostate cancer-involvement of androgen receptor signaling pathway. Oncogene. 34:4735–4745. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Ylitalo EB, Thysell E, Jernberg E, Lundholm M, Crnalic S, Egevad L, Stattin P, Widmark A, Bergh A and Wikström P: Subgroups of castration-resistant prostate cancer bone metastases defined through an inverse relationship between androgen receptor activity and immune response. Eur Urol. 71:776–787. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Pinto F, Pértega-Gomes N, Vizcaino JR, Andrade RP, Cárcano FM and Reis RM: Brachyury as a potential modulator of androgen receptor activity and a key player in therapy resistance in prostate cancer. Oncotarget. 7:28891–28902. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Ware KE, Somarelli JA, Schaeffer D, Li J, Zhang T, Park S, Patierno SR, Freedman J, Foo WC, Garcia-Blanco MA and Armstrong AJ: Snail promotes resistance to enzalutamide through regulation of androgen receptor activity in prostate cancer. Oncotarget. 7:50507–50521. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Wang Y, Ledet RJ, Imberg-Kazdan K, Logan SK and Garabedian MJ: Dynein axonemal heavy chain 8 promotes androgen receptor activity and associates with prostate cancer progression. Oncotarget. 7:49268–49280. 2016.PubMed/NCBI

34 

Jiang J, Jia P, Shen B and Zhao Z: Top associated SNPs in prostate cancer are significantly enriched in cis-expression quantitative trait loci and at transcription factor binding sites. Oncotarget. 5:6168–6177. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Eisermann K, Tandon S, Bazarov A, Brett A, Fraizer G and Piontkivska H: Evolutionary conservation of zinc finger transcription factor binding sites in promoters of genes co-expressed with WT1 in prostate cancer. BMC Genomics. 9:3372008. View Article : Google Scholar : PubMed/NCBI

36 

Kojima S, Enokida H, Yoshino H, Itesako T, Chiyomaru T, Kinoshita T, Fuse M, Nishikawa R, Goto Y, Naya Y, et al: The tumor-suppressive microRNA-143/145 cluster inhibits cell migration and invasion by targeting GOLM1 in prostate cancer. J Hum Genet. 59:78–87. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Li SX, Tong YP, Xie XC, Wang QH, Zhou HN, Han Y, Zhang ZY, Gao W, Li SG, Zhang XC and Bi RC: Octameric structure of the human bifunctional enzyme PAICS in purine biosynthesis. J Mol Biol. 366:1603–1614. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Gallenne T, Ross KN, Visser NL, Salony, Desmet CJ, Wittner BS, Wessels LFA, Ramaswamy S and Peeper DS: Systematic functional perturbations uncover a prognostic genetic network driving human breast cancer. Oncotarget. 8:20572–20587. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Goswami MT, Chen G, Chakravarthi BV, Pathi SS, Anand SK, Carskadon SL, Giordano TJ, Chinnaiyan AM, Thomas DG, Palanisamy N, et al: Role and regulation of coordinately expressed de novo purine biosynthetic enzymes PPAT and PAICS in lung cancer. Oncotarget. 6:23445–23461. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Cifola I, Pietrelli A, Consolandi C, Severgnini M, Mangano E, Russo V, De Bellis G and Battaglia C: Comprehensive genomic characterization of cutaneous malignant melanoma cell lines derived from metastatic lesions by whole-exome sequencing and SNP array profiling. PLoS One. 8:e635972013. View Article : Google Scholar : PubMed/NCBI

41 

Chakravarthi BV, Goswami MT, Pathi SS, Dodson M, Chandrashekar DS, Agarwal S, Nepal S, Balasubramanya Hodigere SA, Siddiqui J, Lonigro RJ, et al: Expression and role of PAICS, a de novo purine biosynthetic gene in prostate cancer. Prostate. 77:10–21. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Gall TM and Frampton AE: Gene of the month: E-cadherin (CDH1). J Clin Pathol. 66:928–932. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Li D, Zhang R, Jin T, He N, Ren L, Zhang Z, Zhang Q, Xu R, Tao H, Zeng G and Gao J: ADH1B and CDH1 polymorphisms predict prognosis in male patients with non-metastatic laryngeal cancer. Oncotarget. 7:73216–73228. 2016.PubMed/NCBI

44 

Jiao F, Hu H, Han T, Zhuo M, Yuan C, Yang H and Wang L and Wang L: Aberrant expression of nuclear HDAC3 and cytoplasmic CDH1 predict a poor prognosis for patients with pancreatic cancer. Oncotarget. 7:16505–16516. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Yu Q, Guo Q, Chen L and Liu S: Clinicopathological significance and potential drug targeting of CDH1 in lung cancer: A meta-analysis and literature review. Drugb Des Devel Ther. 9:2171–2178. 2015.

46 

Huang R, Ding P and Yang F: Clinicopathological significance and potential drug target of CDH1 in breast cancer: A meta-analysis and literature review. Drug Des Devel Ther. 9:5277–5285. 2015.PubMed/NCBI

47 

Zeng W, Zhu J, Shan L, Han Z, Aerxiding P, Quhai A, Zeng F, Wang Z and Li H: The clinicopathological significance of CDH1 in gastric cancer: A meta-analysis and systematic review. Drug Des Devel Ther. 9:2149–2157. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Qiu LX, Li RT, Zhang JB, Zhong WZ, Bai JL, Liu BR, Zheng MH and Qian XP: The E-cadherin (CDH1)-160 C/A polymorphism and prostate cancer risk: A meta-analysis. Eur J Hum Genet. 17:244–249. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Ren H, Du P, Ge Z, Jin Y, Ding D, Liu X and Zou Q: TWIST1 and BMI1 in cancer metastasis and chemoresistance. J Cancer. 7:1074–1080. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Liu H, Wang H, Liu X and Yu T: miR-1271 inhibits migration, invasion and epithelial-mesenchymal transition by targeting ZEB1 and TWIST1 in pancreatic cancer cells. Biochem Biophys Res Commun. 472:346–352. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Li L and Wu D: miR-32 inhibits proliferation, epithelial-mesenchymal transition, and metastasis by targeting TWIST1 in non-small-cell lung cancer cells. Onco Targets Ther. 9:1489–1498. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Ávila-Moreno F, Armas-López L, Álvarez-Moran AM, López-Bujanda Z, Ortiz-Quintero B, Hidalgo-Miranda A, Urrea-Ramírez F, Rivera-Rosales RM, Vázquez-Manríquez E, Peña-Mirabal E, et al: Correction: Overexpression of MEOX2 and TWIST1 is associated with H3K27me3 levels and determines lung cancer chemoresistance and prognosis. PLoS One. 11:e01465692016. View Article : Google Scholar : PubMed/NCBI

53 

Wang T, Hou J, Li Z, Zheng Z, Wei J, Song D, Hu T, Wu Q, Yang JY and Cai JC: miR-15a-3p and miR-16-1-3p negatively regulate twist1 to repress gastric cancer cell invasion and metastasis. Int J Biol Sci. 13:122–134. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Cao C, Sun D, Zhang L and Song L: miR-186 affects the proliferation, invasion and migration of human gastric cancer by inhibition of Twist1. Oncotarget. 7:79956–79963. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Matsusaka S, Zhang W, Cao S, Hanna DL, Sunakawa Y, Sebio A, Ueno M, Yang D, Ning Y, Parekh A, et al: TWIST1 polymorphisms predict survival in patients with metastatic colorectal cancer receiving first-line bevacizumab plus oxaliplatin-based chemotherapy. Mol Cancer Ther. 15:1405–1411. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Zhu DJ, Chen XW, Zhang WJ, Wang JZ, Ouyang MZ, Zhong Q and Liu CC: Twist1 is a potential prognostic marker for colorectal cancer and associated with chemoresistance. Am J Cancer Res. 5:2000–2011. 2015.PubMed/NCBI

57 

Yusup A, Huji B, Fang C, Wang F, Dadihan T, Wang HJ and Upur H: Expression of trefoil factors and TWIST1 in colorectal cancer and their correlation with metastatic potential and prognosis. World J Gastroenterol. 23:110–120. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Malek R, Gajula RP, Williams RD, Nghiem B, Simons BW, Nugent K, Wang H, Taparra K, Lemtiri-Chlieh G, Yoon AR, et al: TWIST1-WDR5-hottip regulates hoxa9 chromatin to facilitate prostate cancer metastasis. Cancer Res. 77:3181–3193. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Zhao X, Deng R, Wang Y, Zhang H, Dou J, Li L, Du Y, Chen R, Cheng J and Yu J: Twist1/Dnmt3a and miR186 establish a regulatory circuit that controls inflammation-associated prostate cancer progression. Oncogenesis. 6:e3152017. View Article : Google Scholar : PubMed/NCBI

60 

Zhao X, Wang Y, Deng R, Zhang H, Dou J, Yuan H, Hou G, Du Y, Chen Q and Yu J: miR186 suppresses prostate cancer progression by targeting Twist1. Oncotarget. 7:33136–33151. 2016.PubMed/NCBI

61 

Cho KH, Choi MJ, Jeong KJ, Kim JJ, Hwang MH, Shin SC, Park CG and Lee HY: A ROS/STAT3/HIF-1α signaling cascade mediates EGF-induced TWIST1 expression and prostate cancer cell invasion. Prostate. 74:528–536. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Gajula RP, Chettiar ST, Williams RD, Thiyagarajan S, Kato Y, Aziz K, Wang R, Gandhi N, Wild AT, Vesuna F, et al: The twist box domain is required for Twist1-induced prostate cancer metastasis. Mol Cancer Res. 11:1387–1400. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Cho KH, Jeong KJ, Shin SC, Kang J, Park CG and Lee HY: STAT3 mediates TGF-β1-induced TWIST1 expression and prostate cancer invasion. Cancer Lett. 336:167–173. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Xu Z, Zhou Y, Cao Y, Dinh TL, Wan J and Zhao M: Identification of candidate biomarkers and analysis of prognostic values in ovarian cancer by integrated bioinformatics analysis. Med Oncol. 33:1302016. View Article : Google Scholar : PubMed/NCBI

65 

Ho JR, Chapeaublanc E, Kirkwood L, Nicolle R, Benhamou S, Lebret T, Allory Y, Southgate J, Radvanyi F and Goud B: Deregulation of Rab and Rab effector genes in bladder cancer. PLoS One. 7:e394692012. View Article : Google Scholar : PubMed/NCBI

66 

Gilling CE, Mittal AK, Chaturvedi NK, Iqbal J, Aoun P, Bierman PJ, Bociek RG, Weisenburger DD and Joshi SS: Lymph node-induced immune tolerance in chronic lymphocytic leukaemia: A role for caveolin-1. Br J Haematol. 158:216–231. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Abdelgawad IA, Radwan NH and Hassanein HR: KIAA0101 mRNA expression in the peripheral blood of hepatocellular carcinoma patients: Association with some clinicopathological features. Clin Biochem. 49:787–791. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Yuan RH, Jeng YM, Pan HW, Hu FC, Lai PL, Lee PH and Hsu HC: Overexpression of KIAA0101 predicts high stage, early tumor recurrence, and poor prognosis of hepatocellular carcinoma. Clin Cancer Res. 13:5368–5376. 2007. View Article : Google Scholar : PubMed/NCBI

69 

Liu L, Liu Y, Chen X, Wang M, Zhou Y, Zhou P, Li W and Zhu F: Variant 2 of KIAA0101, antagonizing its oncogenic variant 1, might be a potential therapeutic strategy in hepatocellular carcinoma. Oncotarget. 8:43990–44003. 2017.PubMed/NCBI

70 

Zhu K, Diao D, Dang C, Shi L, Wang J, Yan R, Yuan D and Li K: Elevated KIAA0101 expression is a marker of recurrence in human gastric cancer. Cancer Sci. 104:353–359. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Zhang CF, Xia YH, Zheng QF, Li ZJ, Guo XH, Zhou HC, Zhang LL, Dong LP and Han Y: Effects of KIAA0101 expression on proliferation and invasion of gastric carcinoma MKN-45 cells. Zhonghua Bing Li Xue Za Zhi. 41:553–557. 2012.(In Chinese). PubMed/NCBI

72 

Kato T, Daigo Y, Aragaki M, Ishikawa K, Sato M and Kaji M: Overexpression of KIAA0101 predicts poor prognosis in primary lung cancer patients. Lung Cancer. 75:110–118. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Cheng Y, Li K, Diao D, Zhu K, Shi L, Zhang H, Yuan D, Guo Q, Wu X, Liu D and Dang C: Expression of KIAA0101 protein is associated with poor survival of esophageal cancer patients and resistance to cisplatin treatment in vitro. Lab Invest. 93:1276–1287. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Fan S and Li X, Tie L, Pan Y and Li X: KIAA0101 is associated with human renal cell carcinoma proliferation and migration induced by erythropoietin. Oncotarget. 7:13520–13537. 2016.PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 18 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xie ZC, Huang JC, Zhang LJ, Gan BL, Wen DY, Chen G, Li SH and Yan HB: Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics. Mol Med Rep 18: 5630-5646, 2018
APA
Xie, Z., Huang, J., Zhang, L., Gan, B., Wen, D., Chen, G. ... Yan, H. (2018). Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics. Molecular Medicine Reports, 18, 5630-5646. https://doi.org/10.3892/mmr.2018.9598
MLA
Xie, Z., Huang, J., Zhang, L., Gan, B., Wen, D., Chen, G., Li, S., Yan, H."Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics". Molecular Medicine Reports 18.6 (2018): 5630-5646.
Chicago
Xie, Z., Huang, J., Zhang, L., Gan, B., Wen, D., Chen, G., Li, S., Yan, H."Exploration of the diagnostic value and molecular mechanism of miR‑1 in prostate cancer: A study based on meta‑analyses and bioinformatics". Molecular Medicine Reports 18, no. 6 (2018): 5630-5646. https://doi.org/10.3892/mmr.2018.9598