Open Access

Investigation of the molecular mechanisms underlying metastasis in prostate cancer by gene expression profiling

  • Authors:
    • Xinghua Zhang
    • Xiaoli Yao
    • Cong Qin
    • Pengcheng Luo
    • Jie Zhang
  • View Affiliations

  • Published online on: May 20, 2016     https://doi.org/10.3892/etm.2016.3376
  • Pages: 925-932
  • Copyright: © Zhang 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

The present study aimed to screen potential genes associated with metastatic prostate cancer (PCa), in order to improve the understanding of the mechanisms underlying PCa metastasis. The GSE3325 microarray dataset, which was downloaded from the Gene Expression Omnibus database, consists of seven clinically localized PCa samples, six hormone-refractory metastatic PCa samples and six benign prostate tissue samples. The Linear Models for Microarray Data package was used to identify differentially-expressed genes (DEGs) and a hierarchical cluster analysis for DEGs was performed with the pheatmap package. Furthermore, potential functions for the DEGs were predicted by a functional enrichment analysis. Subsequently, microRNAs (miRNAs) potentially involved in the regulation of PCa metastasis were identified by WebGestalt software, and the miRNA‑DEG regulatory network was visualized using Cytoscape. In addition, a pathway enrichment analysis for DEGs in the regulatory network was performed. A total of 306 and 2,073 genes were differentially expressed in the clinically localized PCa and the metastatic PCa groups, respectively, as compared with the benign prostate group, of which 174 were differentially expressed in both groups. A number of the DEGs, including CAMK2D and SH3BP4, were significantly enriched in the cell cycle, and others, such as MAF, were associated with the regulation of cell proliferation. Furthermore, some DEGs (CAMK2D and PCDH17) were observed to be regulated by miR‑30, whereas others (ADCY2, MAF, SH3BP4 and PCDH17) were modulated by miR‑182. Additionally, ADCY2 and CAMK2D were distinctly enriched in the calcium signaling pathway. The present study identified novel DEGs, including ADCY2, CAMK2D, MAF, SH3BP4 and PCDH17, that may be involved in the metastasis of PCa.

Introduction

Prostate cancer (PCa) is the most common cancer among European and American men, and accounts for 27% (233,000) of cancer incidences in men in the USA (1). It has a high mortality rate as a result of its high propensity for metastasis (2,3). PCa has been shown to preferentially metastasize to the bone marrow stroma of the axial skeleton (4); however, the precise mechanism underlying PCa metastasis is currently unclear. Therefore, the identification of specific metastasis biomarkers and novel diagnostic targets is required in order to improve the prognosis and treatment of the disease.

Previous studies have made considerable progress in identifying the key regulators in the PCa metastatic process. E-cadherin, which is attached to the actin cytoskeleton via intracellular catenin, has been implicated in the process of PCa metastasis; in primary PCa, reduced E-cadherin expression was associated with bone metastasis and a poor prognosis (5). In addition, the expression of the DLC1 tumor-suppressor gene in metastatic PCa cells has been shown to upregulate the expression of E-cadherin, resulting in the suppression of highly metastatic PCa cell invasion by inhibiting the activity of RhoA-GTP and RhoC-GTP (6). The activation of Rho GTPases is dependent on the downstream Ras protein, which has a major influence on cell signaling (7). Members of the Rho GTPase family are involved in cancer cell motility by regulating actin dynamics and controlling morphological changes (8). A previous study demonstrated that the suppression of the farnesyl and geranyl-geranyl prenylation pathways markedly reduced the migration and motility of PCa cells by inhibiting Ras prenylation and concurrent Rho activation (9). Furthermore, activation of the phosphoinositide 3-kinase/protein kinase B (AKT) signaling pathway has been more frequently observed in resistant and metastatic PCa compared with primary PCa, and thus targeting this signaling pathway may improve the outcome of patients with aggressive PCa (10). Previous studies have reported various genes able to promote PCa tumorigenesis and metastasis, including CCL2 (11), SERPINB5 (12), SRC (13), TMPRSS2-ERG gene fusion and PCA3 (14). In addition, microRNAs (miRNAs), which are considered to be important regulators of gene expression, have been associated with the development of metastatic PCa. For instance, miR-203 (15), miR-16 (16), miR-205 (17), miR-24 (18), miR-29a (19) and miR-145 (16) have all been implicated in PCa metastasis.

Varambally et al (20) performed an integrative genomic and proteomic analysis of benign prostate and metastatic PCa; they reported 48–64% concordance between protein and transcript levels and demonstrated that proteomic alterations between metastatic and clinically localized PCa, which map concordantly to gene transcripts, can serve as predictors of clinical outcome in PCa as well as other solid tumors. However, to the best of our knowledge, the potential miRNAs involved in metastatic PCa, and the interactions of differentially-expressed genes (DEGs) targeted by miRNAs, have yet to be investigated. Therefore, the present study aimed to further elucidate the molecular mechanisms underlying the metastasis of PCa by analyzing the microarray data of benign prostate, clinically localized and metastatic PCa deposited by Varambally et al (20) in the Gene Expression Omnibus (GEO) database. Initially a hierarchical cluster analysis for DEGs was performed, followed by a Gene Ontology (GO) functional enrichment analysis. Furthermore, potential miRNAs in metastatic PCa were identified and a miRNA-DEG regulatory network was constructed. Finally, a pathway enrichment analysis for DEGs in the regulatory network was performed. The results of this bioinformatics analysis may shed light on the molecular mechanisms underlying the metastasis of PCa and provide novel diagnostic biomarkers.

Materials and methods

Affymetrix microarray data

The GSE3325 gene expression profile data (20) was downloaded from the GEO (http://www.ncbi.nlm.nih.gov/geo/) and was based on the GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array platform. A total of 19 human prostate tissue samples were available for further analysis, including seven clinically localized PCa samples, six hormone-refractory metastatic PCa samples and six benign prostate tissue samples.

CEL and probe annotation files were downloaded from GEO, and the gene expression data for all samples were preprocessed via Robust Multichip Averaging background correction, quantile normalization and probe summarization (21) in the affy software package (version 1.34.0; http://bioconductor.org/packages/release/bioc/html/affy.html), as described previously (22).

DEGs screening

The Linear Models for Microarray Data package of R (https://bioconductor.org/packages/release/bioc/html/limma.html) was used to identify genes that were differentially expressed in the primary PCa and metastatic PCa groups, as compared with the benign prostate group, as described previously (23). The raw P-value was adjusted according to the false discovery rate (FDR) using the Benjamin and Hochberg method (24). Only genes with a cut-off criteria of |log2fold change| >1 and FDR<0.01 were considered to be differentially expressed.

Hierarchical cluster analysis for DEGs

Hierarchical clustering is a common method used to determine clusters of similar data points in a multidimensional space (25). The pheatmap package (version 1.0.2; https://cran.r-project.org/web/packages/pheatmap/index.html) was used to perform hierarchical clustering of the DEGs via joint between-within distances, as described previously (26). Expression values from multiple clones or probe sets mapping to the same Unigene Cluster ID were averaged.

GO functional enrichment analysis for DEGs

The Database for Annotation, Visualization and Integrated Discovery (DAVID; https://david.ncifcrf.gov/) provides a comprehensive set of novel and powerful tools for assigning biological meaning to a set of genes (27). FDR<0.05 was used as the cut-off criterion for GO functional enrichment analysis by DAVID.

Integrated miRNA-DEG regulatory network construction

The common miRNAs in Gene set B, as predicted by the databases of miRecords (http://c1.accurascience.com/miRecords/), TarBase (http://diana.imis.athena-innovation.gr/DianaTools/index.php?r=tarbase/index) and TargetScan (http://www.targetscan.org/), were selected using WEB-based GEne SeT AnaLysis Toolkit software (update 2013; http://bioinfo.vanderbilt.edu/webgestalt/), and P<0.05 was used as the cut-off criterion. Subsequently, the Search Tool for the Retrieval of Interacting Genes (http://string-db.org/) was used to analyze the interactions between the DEGs targeted by miRNAs by calculating their combined score; a score of >0.4 was set as the cut-off criterion. Finally, the integrated miRNA-DEG regulatory network was visualized using Cytoscape (http://cytoscape.org/).

Pathway enrichment analysis for DEGs in the regulatory network

Pathway enrichment analysis was conducted as described previously (28) to identify significant metabolic pathways for the DEGs. P<0.05 was used as the cut-off criterion for the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis using DAVID.

Results

Identification of DEGs

Based on the cut-off criteria, 2,727 DEGs were identified for the clinically localized PCa and metastatic PCa groups, of which 306 were differentially expressed in the clinically localized PCa group only (Gene set A). A total of 2,073 genes were differentially expressed in the metastatic PCa group only (Gene set B) and 174 genes were differentially expressed in both groups (Gene set C; Fig. 1), as compared with the benign prostate group.

Hierarchical cluster analysis

An unsupervised hierarchical cluster analysis of the data revealed that the DEGs could be used to accurately classify prostate samples as benign, clinically localized prostate cancer or metastatic disease (Fig. 2).

GO functional enrichment analysis for Gene sets A, B and C

In Gene set A, DLX2, DLX1, HOXD10 and HOXD11 DEGs were associated with proximal/distal pattern formation (FDR=3.55E-04), whereas RBP4, PDE3B and PPARG were implicated in the response to insulin (FDR=7.8400), homeostatic processes (FDR=9.6200), chemical homeostasis (FDR=0.0019) and responses to peptide hormones (FDR=0.0023) and organic substances (FDR=0.0029) (Table I).

Table I.

Enriched terms for Gene sets A, B and C.

Table I.

Enriched terms for Gene sets A, B and C.

CategoryTermNo. of genesFDRGenes
Gene set A GO:0009954~proximal/distal pattern formation     53.5500DLX2, DLX1, GREM1, HOXD10, HOXD11
GO:0032868~response to insulin stimulus     87.8400RBP4, EIF4EBP1, FADS1, PPARG, PDE3B, STXBP4, GAL, VLDLR
GO:0042592~homeostatic process  249.6200RBP4, SLC12A2, PPARG, F2RL1, PRDX4, PDE3B, CACNG2, ITPR3, PPARGC1A, MUC6
GO:0001501~skeletal system development  140.0011RBP4, HOXD10, HOXD11, MSX2, DLX2, DLX1, COL9A2, BCL2, CLEC3A, NAB1
GO:0048878~chemical homeostasis  180.0019RBP4, F2RL1, NOX1, PPARG, PDE3B, PPARGC1A, PRKCB, CCL11, MALL, ATP7B
GO:0021877~forebrain neuron fate commitment     30.0022DLX2, DLX1, LHX6
GO:0043434~response to peptide hormone stimulus     90.0023RBP4, EIF4EBP1, FADS1, BCL2, PPARG, PDE3B, STXBP4, GAL, VLDLR
GO:0010033~response to organic substance  220.0029RBP4, ADCY1, GNRH1, FADS1, LOC646626, PPARG, PTGS1, PDE3B, COLEC12, STXBP4
GO:0009725~response to hormone stimulus  140.0038RBP4, ADCY1, GNRH1, FADS1, PTGS1, PPARG, PDE3B, STXBP4, GAL, EIF4EBP1
GO:0034637~cellular carbohydrate biosynthetic process     660.0038RBP4, ISYNA1, UAP1, GNE, PPARGC1A, ACN9
Gene set BGO:0022402~cell cycle process1025.2300PRC1, ZAK, AIF1, BTRC, CDCA8, CDC6, CENPF, PTTG1, AURKB, TGFB2
GO:0051726~regulation of cell cycle  681.1100E2F2, PTGS2, ZAK, FAM175A, PKMYT1, PDCD4, PTEN, GTSE1, TGFB2, MYC
GO:0007049~cell cycle1281.8200ZAK, PRC1, AIF1, BTRC, PKMYT1, RBM7, AURKA, AURKB, PTTG1, TGFB2
GO:0051301~cell division  614.6100PRC1, PTTG1, CCNE1, CDCA2, CDC6, CABLES2, CDCA5, CCNA2, ASPM, CDK1
GO:0022403~cell cycle phase  784.7700E2F1, PRC1, PKMYT1, RBM7, AURKA, AURKB, PTTG1, GTSE1, CCNE1, CDCA8
GO:0010035~response to inorganic substance  476.4600CAV1, GCLC, PTGS2, PDGFA, SNCA, TPM1, PTEN, KCNMB1, FOS, GSN
GO:0007346~regulation of mitotic cell cycle  380.0011CAV2, HOXA13, PML, PKMYT1, ASNS, ANLN, ZNF655, RCC1, SCRIB, MYC
GO:0042127~regulation of cell proliferation1260.0012HRAS, CD38, IL6ST, PDGFA, TP63, MAF, TGFB3, STRN, PNP, TGFB2
GO:0030030~cell projection organization  700.0015CAV2, HOXA13, PML, PKMYT1, ANLN, ZNF655, RCC1, SCRIB, GTSE1, MYC
GO:0000278~mitotic cell cycle  700.0018E2F1, PRC1, BTRC, PKMYT1, AURKA, AURKB, PTTG1, GTSE1, CCNE1, NDE1
Gene set CGO:0022402~cell cycle process  213.3000MKI67, DLGAP5, SGOL1, NUSAP1, BIRC5, PBK, CDKN3, CCNB1, CENPA, CAMK2D
GO:0022403~cell cycle phase  183.5700MKI67, DLGAP5, SGOL1, NUSAP1, TTK, BIRC5, PBK, CCNB1, CAMK2D, ID4
GO:0000278~mitotic cell cycle  174.0700DLGAP5, SGOL1, NUSAP1, TTK, BIRC5, PBK, CDKN3, CCNB1, CENPA, CAMK2D
GO:0000279~M phase  152.7400MKI67, DLGAP5, SGOL1, NUSAP1, TTK, BIRC5, PBK, UBE2C, CCNB1, KIF2C
GO:0007049~cell cycle  221.1800DLGAP5, SGOL1, NUSAP1, BIRC5, PBK, CCNB1, SH3BP4, KIF2C, CENPA, CAMK2D
GO:0000280~nuclear division  114.5000CCNB1, KIF2C, CCNB2, DLGAP5, SGOL1, NUSAP1, BIRC5, PBK, UBE2C, ERCC6L
GO:0007067~mitosis  114.5000CCNB1, KIF2C, CCNB2, DLGAP5, SGOL1, NUSAP1, BIRC5, PBK, UBE2C, ERCC6L
GO:0000087~M phase of mitotic cell cycle  115.2300CCNB1, KIF2C, CCNB2, DLGAP5, SGOL1, NUSAP1, BIRC5, PBK, UBE2C, ERCC6L
GO:0048285~organelle fission  116.3000CCNB1, KIF2C, CCNB2, DLGAP5, SGOL1, NUSAP1, BIRC5, PBK, UBE2C, ERCC6L
GO:0007346~regulation of mitotic cell cycle     99.6800DLGAP5, CAMK2D, NUSAP1, TTK, BIRC5, AFAP1L2, GAS1, UBE2C, MYC

[i] Gene set A represents the genes only differentially expressed in the clinically localized prostate cancer group; Gene set B represents the genes only differentially expressed in the metastatic prostate cancer group; Gene set C represents the genes differentially expressed in both groups. FDR, false discovery rate.

In Gene set B, the DEGs were predominantly associated with the cell cycle: PRC1, ZAK, PTTG1, TGFB2, CDCA8, CDC6 and CENPF were associated with the cell cycle process (FDR=5.2300); PRC1, PTTG1, CCNE1, CDCA2 and CDC6 were involved in cell division (FDR=4.6100); and HRAS, CD38, IL6ST, PDGFA, TP63, MAF and TGFB3 were associated with the regulation of cell proliferation (FDR=0.0012) (Table I).

In Gene set C, the DEGs were also predominantly associated with the cell cycle. DLGAP5, SGOL1, NUSAP1, PBK, BIRC5 and CCNB1 were associated with the cell cycle process (FDR=3.3000), M phase (FDR=2.7400), mitosis (FDR=4.5000) and organelle fission (FDR=6.3000), whereas SH3BP4, KIF2C, CCNB2, CENPA and CAMK2D were associated with the cell cycle only (FDR=1.1800) (Table I).

Analysis of the miRNA-DEG regulatory network

A total of 10 miRNAs were identified in Gene set B, including miR-374, miR-128, miR-182, miR-30, miR-302c and miR-524. Notably, miR-30 targeted the majority of the DEGs (11 DEGs, including CAMK2D, PCDH17, EDNRB, KCNJ3 and SOX4), and miR-182 targeted seven DEGs, including EDNRB, MAF, ADCY2, PCDH17, RET, SH3BP4 and BCL11A (Table II).

Table II.

Enriched microRNAs in Gene set B.

Table II.

Enriched microRNAs in Gene set B.

microRNAP-valueCountGenes targeted by microRNA
hsa_TATTATA, MIR-3742.110010RORB, HOMER1, KIF20A, SYBU, DACH1, GATA3, ARHGAP28, AFAP1L2, SOX4, SYT1
hsa_CACTGTG, MIR-1280.0003  9RORB, FBLN2, ADCY2, ACOT11, INSM1, SYT1, FOXQ1, MME, BCL11A
hsa_ATGCAGT, MIR-2170.0003  6STX1A, MAF, PCDH17, DACH1, EZH2, BCL11A
hsa_TGTTTAC, MIR-300.000511SOBP, CAMK2D, COL13A1, SLC36A1, PCDH17, AFAP1L2, EDNRB, KCNJ3, SOX4, MATR3……
hsa_ACAACTT, MIR-3820.0032  4NDRG2, SYT1, MATR3, DACH1
hsa_ACTGCCT, MIR-34B0.0032  6INSM1, SOX4, MYC, ADCY2, PIEZO2, JAKMIP1
hsa_TTGCCAA, MIR-1820.0036  7EDNRB, MAF, ADCY2, PCDH17, RET, SH3BP4, BCL11A
hsa_CTTTGTA, MIR-5240.0036  8SOBP, CTHRC1, PCDH17, ECT2, ID4, RCAN2, HOXD13, SOX4
hsa_ATGTTAA, MIR-302C0.0038  6EDNRB, SALL3, MAF, MATR3, DACH1, BCL11A
hsa_TGCACTT, MIR-5190.0038  8SOBP, RORB, SYBU, NETO2, SOX4, SYT1, APCDD1, JAKMIP1

[i] Count represents the number of differentially-expressed genes targeted by microRNA. Gene set B represents the genes only differentially expressed in the metastatic prostate cancer group.

The miRNA-DEG regulatory network in Fig. 3 contained 10 miRNAs and 43 corresponding DEGs. ADCY2 was regulated by miR-128, miR-34B and miR-182; EDNRB was regulated by miR-30, miR-182 and miR-302C; CAMK2D was regulated by miR-30; PCDH17 was modulated by miR-217, miR-30, miR-182 and miR-524; SH3BP4 was modulated by miR-182; and MAF interacted with miR-182, miR-302c and BCL11A.

Pathway enrichment analysis for the DEGs in the regulatory network

The DEGs in the regulatory network were enriched in two pathways, including the calcium signaling pathway (EDNRB, ADCY2 and CAMK2D), and thyroid cancer (RET and MYC; Table III).

Table III.

Enriched pathways for the differentially-expressed genes in the regulatory network.

Table III.

Enriched pathways for the differentially-expressed genes in the regulatory network.

TermDescriptionCountP-valueGenes
hsa04020Calcium signaling pathway30.02231EDNRB, ADCY2, CAMK2D
hsa05216Thyroid cancer20.03927RET, MYC

Discussion

The present study identified 306 and 2,073 genes that were differentially expressed in the clinically localized PCa group and the metastatic PCa group, respectively, as compared with the benign prostate group. Of these, 174 genes were differentially expressed in both the clinically localized PCa and metastatic PCa groups.

ADCY2, which encodes adenylate cyclase 2, and CAMK2D, which encodes calcium/calmodulin-dependent protein kinase II δ (29,30), were shown to be enriched in the calcium signaling pathway. Metastasis is the predominant cause of mortality in patients with PCa, and Ca2+ is a crucial regulator of cell migration (31). Elevated intracellular concentrations of Ca2+ may facilitate the metastasis of PCa by triggering the activation of the Akt signaling pathway and promoting PCa cell (PC3) attachment (32). CAMK2D encodes components of the Wnt/β-catenin-signaling pathway, the inhibition of which delays metastatic PCa cell cycle progression and proliferation (33). In the present study, CAMK2D was associated with the cell cycle, which is known to be a critical event in tumor growth and metastasis (34). Furthermore, CAMK2D was observed to be regulated by miR-30. As a tumor suppressor, miR-30 has been shown to be downregulated by oncogenic signals, such as hepatocyte growth factor and epidermal growth factor, in PCa samples (35), and overexpression of miR-30 in PCa cells was reported to suppress the epithelial-to-mesenchymal transition and inhibit cell migration and invasion (36).

ADCY2 was observed to be modulated by miR-182. A previous study demonstrated that ectopic expression of miR-182 in PC3 significantly reduced protein expression levels of GNA13, GNA13-3′-untranslated region (UTR)-reporter activity and extracorporeal invasion of these cells (37). In addition, aberrant overexpression of miR-182 was shown to promote the proliferation, increase the invasion, facilitate the G1/S cell cycle transition and reduce early apoptosis of PC3 cells; and, miR-182 was able to suppress the expression of the NDRG1 tumor suppressor gene by directly targeting the NDRG1 3′-UTR (38). Therefore, CAMK2D and ADCY2 may be involved in the metastasis of PCa via calcium signaling and regulation by miR-30 and miR-182, respectively.

MAF, which was also modulated by miR-182 in the present study, was associated with the regulation of cell proliferation. MAF acts as a macrophage-activating factor and is generated from a precursor protein termed the Gc protein (39). Deglycosylation of the Gc protein prevents its conversion to MAF, inhibiting macrophage activation and resulting in immunosuppression (40). In a previous study, patients with metastatic PCa were administered Gc protein with MAF precursor activity (100 ng/week), and were shown to have serum activity levels of Nagalase equivalent to those of healthy controls, thus suggesting that these patients were tumor-free (41). Furthermore, MAF expression has been associated with the receptor tyrosine kinase, platelet-derived growth factor receptor (PDGFR)-β status (42). In the miRNA-DEG regulatory network, MAF was also modulated by miR-302c, and it has been reported that miR-302c is downregulated in clinical PCa samples (43). In addition, MAF interacted with BCL11A, which was observed to be upregulated in PC3 holoclones (44). Therefore, MAF may have an important role in the metastasis of PCa by interacting with miR-182, miR-302c and BCL11A.

In the present study, the downregulated DEG SH3BP4 was shown to be associated with the cell cycle and was also regulated by miR-182. SH3BP4 encodes SH3-domain binding protein 4 (45). SH3 domains are found in a variety of proteins, including tyrosine kinases, such as Abl and Src, and are involved in endocytosis, intracellular sorting and the cell cycle (46). Another downregulated DEG PCDH17, which encodes protocadherin 17, was shown to interact with miR-182 and miR-30. PCDH17 methylation is a common tumor-specific event in PCa and has been associated with a shorter biochemical recurrence-free survival rate and a reduced overall survival rate of patients with PCa following a radical prostatectomy (47). Therefore, SH3BP4 and PCDH17 may be responsible for the metastasis of PCa via their interactions with miR-182 and/or miR-30. Furthermore, miR-374 was significantly enriched in Gene set B. Previous studies have reported that miR-374 is markedly downregulated in PCa (48,49). Furthermore, miR-374b, which is a subtype of miR-374, has been shown to be downregulated in prostate fluid or serum samples from prostate cancer patients, and thus may serve as a PCa biomarker in clinical diagnosis (50).

In conclusion, the present study identified numerous important DEGs, including ADCY2, CAMK2D, MAF, SH3BP4 and PCDH17, that may be involved in the metastasis of PCa. However, the results of the present study require validation by further experiments, and the molecular mechanisms underlying metastatic PCa require further investigation.

Acknowledgements

The present study was supported by the National Science Foundation of China (grant no. 81470923).

References

1 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Ferlay J, Parkin DM and Steliarova-Foucher E: Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 46:765–781. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Watahiki A and Wang Y, Morris J, Dennis K, O'Dwyer HM, Gleave M, Gout PW and Wang Y: MicroRNAs associated with metastatic prostate cancer. PLoS One. 6:e249502011. View Article : Google Scholar : PubMed/NCBI

4 

Sun YX, Schneider A, Jung Y, Wang J, Dai J, Wang J, Cook K, Osman NI, Koh-Paige AJ, Shim H, et al: Skeletal localization and neutralization of the SDF-1 (CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. J Bone Miner Res. 20:318–329. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Cheng L, Nagabhushan M, Pretlow TP, Amini SB and Pretlow TG: Expression of E-cadherin in primary and metastatic prostate cancer. Am J Pathol. 148:1375–1380. 1996.PubMed/NCBI

6 

Tripathi V, Popescu NC and Zimonjic DB: DLC1 induces expression of E-cadherin in prostate cancer cells through Rho pathway and suppresses invasion. Oncogene. 33:724–733. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Hu L, Shi Y, Hsu JH, Gera J, Van Ness B and Lichtenstein A: Downstream effectors of oncogenic ras in multiple myeloma cells. Blood. 101:3126–3135. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Hall A: Rho family GTPases. Biochem Soc Trans. 40:1378–1382. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Khafagy R, Stephens T, Hart C, Ramani V, Brown M and Clarke N: In vitro effects of the prenyl transferase inhibitor AZD3409 on prostate cancer epithelial cells. J Clin Oncol. 22:47442004.

10 

Toren P and Zoubeidi A: Targeting the PI3K/Akt pathway in prostate cancer: Challenges and opportunities (review). Int J Oncol. 45:1793–1801. 2014.PubMed/NCBI

11 

Zhang J, Patel L and Pienta KJ: CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis. Cytokine Growth Factor Rev. 21:41–48. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Luo JL, Tan W, Ricono JM, Korchynskyi O, Zhang M, Gonias SL, Cheresh DA and Karin M: Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature. 446:690–694. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Cai H, Smith DA, Memarzadeh S, Lowell CA, Cooper JA and Witte ON: Differential transformation capacity of Src family kinases during the initiation of prostate cancer. Proc Natl Acad Sci USA. 108:6579–6584. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Salagierski M and Schalken JA: Molecular diagnosis of prostate cancer: PCA3 and TMPRSS2: ERG gene fusion. J Urol. 187:795–801. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Saini S, Majid S, Yamamura S, Tabatabai L, Suh SO, Shahryari V, Chen Y, Deng G, Tanaka Y and Dahiya R: Regulatory role of miR-203 in prostate cancer progression and metastasis. Clin Cancer Res. 17:5287–5298. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Schaefer A, Jung M, Mollenkopf HJ, Wagner I, Stephan C, Jentzmik F, Miller K, Lein M, Kristiansen G and Jung K: Diagnostic and prognostic implications of microRNA profiling in prostate carcinoma. Int J Cancer. 126:1166–1176. 2010.PubMed/NCBI

17 

Gandellini P, Folini M and Zaffaroni N: Towards the definition of prostate cancer-related microRNAs: Where are we now? Trends Mol Med. 15:381–390. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Szczyrba J, Löprich E, Wach S, Jung V, Unteregger G, Barth S, Grobholz R, Wieland W, Stöhr R, Hartmann A, et al: The microRNA profile of prostate carcinoma obtained by deep sequencing. Mol Cancer Res. 8:529–538. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Varambally S, Yu J, Laxman B, Rhodes DR, Mehra R, Tomlins SA, Shah RB, Chandran U, Monzon FA, Becich MJ, et al: Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression. Cancer Cell. 8:393–406. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Gautier L, Cope L, Bolstad BM and Irizarry RA: affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:2004.PubMed/NCBI

24 

Verhoeven KJ, Simonsen KL and McIntyre LM: Implementing false discovery rate control: Increasing your power. Oikos. 108:643–647. 2005. View Article : Google Scholar

25 

Olson CF: Parallel algorithms for hierarchical clustering. Parallel Computing. 21:1313–1325. 1995. View Article : Google Scholar

26 

Kolde R: Pheatmap: Pretty Heatmaps. R package version 0.7. 7. Journal. 2012.

27 

Huang DW, Sherman BT, Tan Q, Collins JR, Alvord G, Roayaei J, Stephens R, Baseler MW, Lane HC and Lempicki RA: The DAVID Gene functional classification tool: A novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 8:R1832007. View Article : Google Scholar : PubMed/NCBI

28 

Huang DW, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Visel A, Alvarez-Bolado G, Thaller C and Eichele G: Comprehensive analysis of the expression patterns of the adenylate cyclase gene family in the developing and adult mouse brain. J Comp Neurol. 496:684–679. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Hagemann D, Bohlender J, Hoch B, Krause EG and Karczewski P: Expression of Ca2+/calmodulin-dependent protein kinase II delta-subunit isoforms in rats with hypertensive cardiac hypertrophy. Mol Cell Biochem. 220:69–76. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Prevarskaya N, Skryma R and Shuba Y: Calcium in tumour metastasis: New roles for known actors. Nat Rev Cancer. 11:609–618. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Liao J, Schneider A, Datta NS and McCauley LK: Extracellular calcium as a candidate mediator of prostate cancer skeletal metastasis. Cancer Res. 66:9065–9073. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Rajan P, Sudbery IM, Villasevil ME, Mui E, Fleming J, Davis M, Ahmad I, Edwards J, Sansom OJ, Sims D, et al: Next-generation sequencing of advanced prostate cancer treated with androgen-deprivation therapy. Eur Urol. 66:32–39. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Whitfield ML, Sherlock G, Saldanha AJ, Murray JI, Ball CA, Alexander KE, Matese JC, Perou CM, Hurt MM, Brown PO and Botstein D: Identification of genes periodically expressed in the human cell cycle and their expression in tumors. Mol Biol Cell. 13:1977–2000. 2002. View Article : Google Scholar PubMed/NCBI

35 

White R and Kung H: miR-30 as a tumor suppressor connects EGF/Src signal to ERG and EMT. Oncogene. 33:2495–2503. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Kao C, Martiniez A, Shi X, Yang J, Evans C, Dobi A, Devere White R and Kung H: miR-30 as a tumor suppressor connects EGF/Src signal to ERG and EMT. Oncogene. 33:2495–2503. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Rasheed SA, Teo CR, Beillard EJ, Voorhoeve PM and Casey PJ: MicroRNA-182 and microRNA-200a control G-protein subunit α-13 (GNA13) expression and cell invasion synergistically in prostate cancer cells. J Biol Chem. 288:7986–7995. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Liu R, Li J, Teng Z, Zhang Z and Xu Y: Overexpressed microRNA-182 promotes proliferation and invasion in prostate cancer PC-3 cells by down-regulating N-myc downstream regulated gene 1 (NDRG1). PLoS One. 8:e689822013. View Article : Google Scholar : PubMed/NCBI

39 

Nagasawa H, Uto Y, Sasaki H, Okamura N, Murakami A, Kubo S, Kirk KL and Hori H: Gc protein (vitamin D-binding protein): Gc genotyping and GcMAF precursor activity. Anticancer Res. 25:3689–3695. 2005.PubMed/NCBI

40 

Yamamoto N, Naraparaju VR and Asbell SO: Deglycosylation of serum vitamin D3-binding protein leads to immunosuppression in cancer patients. Cancer Res. 56:2827–2831. 1996.PubMed/NCBI

41 

Yamamoto N, Suyama H and Yamamoto N: Immunotherapy for prostate cancer with Gc protein-derived macrophage-activating factor, GcMAF. Transl Oncolo. 1:65–72. 2008. View Article : Google Scholar

42 

Sharad S, Srivastava A, Ravulapalli S, Parker P, Chen Y, Li H, Petrovics G and Dobi A: Prostate cancer gene expression signature of patients with high body mass index. Prostate Cancer Prostatic Dis. 14:22–29. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Coppola V, De Maria R and Bonci D: MicroRNAs and prostate cancer. Endocr Relat Cancer. 17:F1–F17. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Zhang K and Waxman DJ: PC3 prostate tumor-initiating cells with molecular profile FAM65Bhigh/MFI2low/LEF1low increase tumor angiogenesis. Mol Cancer. 9:3192010. View Article : Google Scholar : PubMed/NCBI

45 

Dunlevy JR, Koppelman ED and Kolberg JB: The expression of a SH3BP4-related protein in retinal cells. Invest Ophthalmol Vis Sci. 46:2996. 2005.

46 

Dunlevy JR, Berryhill BL, Vergnes JP, SundarRaj N and Hassell JR: Cloning, chromosomal localization and characterization of cDNA from a novel gene, SH3BP4, expressed by human corneal fibroblasts. Genomics. 62:519–524. 1999. View Article : Google Scholar : PubMed/NCBI

47 

Lin YL, Xie PG, Wang L and Ma JG: Aberrant methylation of protocadherin 17 and its clinical significance in patients with prostate cancer after radical prostatectomy. Med Sci Monit. 20:1376–1382. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Tang X, Tang X, Gal J, Kyprianou N, Zhu H and Tang G: Detection of microRNAs in prostate cancer cells by microRNA array. Methods Mol Biol. 732:69–88. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Ma S, Chan YP, Kwan PS, Lee TK, Yan M, Tang KH, Ling MT, Vielkind JR, Guan XY and Chan KW: MicroRNA-616 induces androgen-independent growth of prostate cancer cells by suppressing expression of tissue factor pathway inhibitor TFPI-2. Cancer Res. 71:583–592. 2011. View Article : Google Scholar : PubMed/NCBI

50 

He HC, Han ZD, Dai QS, Ling XH, Fu X, Lin ZY, Deng YH, Qin GQ, Cai C, Chen JH, et al: Global analysis of the differentially expressed miRNAs of prostate cancer in Chinese patients. BMC Genomics. 14:7572013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2016
Volume 12 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Yao X, Qin C, Luo P and Zhang J: Investigation of the molecular mechanisms underlying metastasis in prostate cancer by gene expression profiling. Exp Ther Med 12: 925-932, 2016
APA
Zhang, X., Yao, X., Qin, C., Luo, P., & Zhang, J. (2016). Investigation of the molecular mechanisms underlying metastasis in prostate cancer by gene expression profiling. Experimental and Therapeutic Medicine, 12, 925-932. https://doi.org/10.3892/etm.2016.3376
MLA
Zhang, X., Yao, X., Qin, C., Luo, P., Zhang, J."Investigation of the molecular mechanisms underlying metastasis in prostate cancer by gene expression profiling". Experimental and Therapeutic Medicine 12.2 (2016): 925-932.
Chicago
Zhang, X., Yao, X., Qin, C., Luo, P., Zhang, J."Investigation of the molecular mechanisms underlying metastasis in prostate cancer by gene expression profiling". Experimental and Therapeutic Medicine 12, no. 2 (2016): 925-932. https://doi.org/10.3892/etm.2016.3376