Open Access

Screening of potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles

  • Authors:
    • Rui Zhao
    • Yao Wang
    • Muchun Zhang
    • Xinquan Gu
    • Weihua Wang
    • Jiufeng Tan
    • Xin Wei
    • Ning Jin
  • View Affiliations

  • Published online on: September 4, 2017     https://doi.org/10.3892/ol.2017.6879
  • Pages: 5361-5369
  • Copyright: © Zhao 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 aim of the present study was to analyze potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles. First, gene expression profiles GSE38241 and GSE3933 were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between prostate cancer and normal control samples were identified using the Linear Models for Microarray Data package. Pathway enrichment analysis of DEGs was performed using Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes. Furthermore, protein‑protein interaction (PPI) networks of DEGs were constructed, on the basis of the Search Tool for the Retrieval of Interacting Genes/Proteins database. The Molecular Complex Detection was utilized to perform module analysis of the PPI networks. In addition, transcriptional regulatory networks were constructed on the basis of the associations between transcription factors (TFs) and target genes. A total of 529 DEGs were identified, including 129 upregulated genes that were primarily associated with to the cell cycle. Additionally, 400 downregulated genes were identified, which were principally enriched in the pathways associated with vascular smooth muscle contraction and focal adhesion. Cell Division Cycle Associated 8, Cell Division Cycle 45, Ubiquitin Conjugating Enzyme E2 C and Thymidine Kinase 1 were identified as hub genes in the upregulated sub‑network. Furthermore, the upregulated TF E2F, and the downregulated TF Early Growth Response 1, were identified to be critical in the transcriptional regulatory networks. The identified DEGs and TFs may have critical roles in the progression of prostate cancer, and may be used as target molecules for treating prostate cancer.

Introduction

Prostate cancer is a malignancy that occurs in the prostate epithelial cells and it is the most common type of reproductive system cancer in males worldwide (1,2). Cancer statistics in 2016 revealed that prostate cancer accounts for ~20% novel cancer cases in males in the USA (3). Radical prostatectomy is an effective treatment to improve patient survival time (4), but it is only suitable for ~10% of all cases (5). A number of other therapies, including radiotherapy, hormonal therapy, chemotherapy and immunotherapy, have been developed for prostate cancer treatment (6); however, there is limited information regarding the long-term survival rate, and the mortality rate of patients with prostate cancer remains high (7). Therefore, investigations into novel treatment strategies for patients with prostate cancer are required.

Gene therapy and small molecule drugs are novel strategies for cancer treatment, and have received increasing attention over the past few decades (8). Recently, a number of studies have been conducted to reveal the underlying molecular mechanisms and identify treatment targets for prostate cancer (920). Specific genes involved in the DNA damage response, including breast cancer 1, breast cancer 2 and tumor protein 53 genes, are mutated during the progression of prostate cancer (911). A number of activated carcinogenic signaling pathways, including c-Myc, protein kinase B and Ras, induce the replication and genomic instability of prostate cancer cells (1214). The histone-lysine N-methyltransferase gene is overexpressed in prostate cancer and may act as a therapeutic target (15). Previous studies have primarily focused on a certain gene or pathway; therefore, it is necessary to explore the underlying molecular mechanisms and therapy targets for prostate cancer using other methods.

Identification and analysis of differentially expressed genes (DEGs) is an effective method to acquire multiple novel targets for the treatment of diseases (16,17). An expression profiling analysis for prostate cancer has been studied previously (18). In addition, DNA methylation alterations in prostate cancer have been analyzed using the gene expression profile GSE38241 (19), and clinically relevant subtypes, including subgroups I (the clinically least aggressive subclass) and subgroups II/III (clinically aggressive tumor subclasses), of prostate cancer have been studied using the gene expression profile GSE3933 (20). However, DEGs and their regulatory factors between prostate cancer and normal samples were not analyzed as part of the present study.

In the present study, the expression-profiling data GSE38241 and GSE3933 were integrated to identify DEGs between prostate cancer samples and normal samples. The functions of DEGs were analyzed using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Furthermore, protein-protein interactions (PPIs) of DEGs were investigated and hub genes, genes identified to be key genes, in the PPI network were identified. In addition, transcriptional regulatory networks were constructed on the basis of the associations between transcription factors (TFs) and DEGs. The results of the present study may identify the underlying molecular mechanisms of prostate cancer and provide targets for the treatment of prostate cancer.

Materials and methods

mRNA expression profiles of prostate cancer

The datasets of prostate cancer gene expression profiling by array with large sample size and high data quality were searched in the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo). Prostate cancer and normal control samples were included in the eligible dataset, and samples were not treated by additional treatments such as drugs and radiation. As a result, two prostate cancer expression profiling datasets were chosen for analysis, GSE38241 (18) and GSE3933 (19). Data of 39 samples (18 prostate cancer samples and 21 normal samples) in GSE38241 were produced using platform GPL4133 Agilent-014850 Whole Human Genome Microarray 4×44K G4112F. Data of 112 samples in GSE3933 were produced using three platforms, consisting of GPL2695 SHBB (26 samples), GPL3044 SHCQ (45 samples) and GPL3289 SHBW (41 samples). In the dataset GSE3933, only the data of 45 samples (29 prostate cancer samples and 16 normal samples) obtained from platform GPL3044 were selected for additional analysis, since gene probes detected by platform GPL3044 overlapped more with those from platform GPL4133. Data and probe annotation files were downloaded for analysis.

Data preprocessing

Subsequent to obtaining the raw data, probe IDs of the matrix data were first translated into corresponding gene symbols. If one gene symbol was matched by a number of probe IDs, the mean expression value was selected as the expression level of this gene. In order to obtain reliable results, only the common genes in the two datasets were selected for the following analysis. During the process of merging the two different datasets, batch errors (21) were removed using the ComBat command of sva package in R language (http://www.bioconductor.org/packages/release/bioc/html/sva.html version 3.5) (22). Subsequently, quantile normalization of genes was performed by preprocessCore package in R (http://www.bioconductor.org/packages/release/bioc/html/preprocessCore.html; version 1.38.1) and an expression profile matrix was generated consisting of 12,621 genes.

Identification of DEGs

The linear models for microarray data package (http://www.bioconductor.org/packages/release/bioc/html/limma.html; version 3.22.7) (23), a widely-used tool for the identification of DEGs, was applied to identify DEGs between prostate cancer samples and normal samples. The raw P-value for each gene was calculated and subsequently adjusted into the false discovery rate (FDR) using the Benjamini-Hochberg method (24). Only the genes that met the threshold criteria of |log2 fold change| >1 and FDR<0.05 were identified as DEGs.

Functional enrichment analysis of DEGs

In order to investigate the signaling pathways and biological processes which may be involved in the progression of prostate cancer, GO (25) and KEGG (26) enrichment analysis were performed using the Database for Annotation, Visualization and Integrated Discovery (27,28). This provided a number of functional annotation tools to reveal the biological function of genes. Functional terms with P<0.05 were considered to indicate a statistically significant difference.

PPI networks and module analysis

PPIs of DEGs were searched in the Search Tool for the Retrieval of Interacting Genes/Proteins database (29), which integrates a number of known and predicted associations between proteins. The PPI network was visualized using Cytoscape (30), an open source software for integrating biomolecular networks. In the network, ‘node’ represents a gene or protein, and ‘line’ represents an interaction between two nodes. The degree of each node is equal to the number of nodes that the node interacted with. The node degree represents its topological importance; the higher the degree, the more important the node is (31). Hub genes were identified on the basis of the degree of genes in the PPI network. Molecular Complex Detection (MCODE) (32) is a tool used to determine the dense connections in large PPI networks, which may represent molecular complexes. In the present study, MCODE was utilized to screen the modules from the PPI network with a network aggregation score >10.

Constructing the transcriptional regulatory networks

TFs targeting DEGs were identified from DEGs on the basis of information in the TRANSFAC database (http://gene-regulation.com/pub/databases.html; version 7.0) (33), which provided data on eukaryotic transcription factors, consensus binding sequences (positional weight matrices), experimentally proven binding sites and regulated genes. Transcription regulatory networks were visualized using Cytoscape, as aforementioned, in order to observe the interactions between TFs and target DEGs.

Results

Identified DEGs

Prior to normalization, the medians of gene expression in each sample were markedly distinct (Fig. 1A). However, the medians became consistent and were at a similar level following normalization (Fig. 1B and C), suggesting that the normalization process was successful and the normalized data may be used for additional analysis.

On the basis of the threshold criteria, a total of 529 DEGs were obtained, including 129 upregulated and 400 downregulated genes in prostate cancer samples, compared with normal samples.

Enrichment analysis of DEGs

To reveal the biological functions of DEGs, GO and KEGG pathway enrichment analyses were performed for the up- and downregulated genes. Upregulated genes were primarily enriched in cell cycle-associated GO terms, including cell cycle phase, spindle and adenosine 5′-phosphate binding (Table I). Downregulated genes were identified to be significantly involved in a set of GO terms including muscle organ development, negative regulation of cell proliferation and cell adhesion (Table I).

Table I.

Top 5 most significant upregulated and downregulated DEGs from GO analysis across 3 categories including BP, CC and MM.

Table I.

Top 5 most significant upregulated and downregulated DEGs from GO analysis across 3 categories including BP, CC and MM.

A, Upregulated DEGs

IDTermCountP-value
GOTERM_BP_FATGO:0000279-M phase27 1.68×10−20
GOTERM_BP_FATGO:0022403-cell cycle phase29 2.93×10−20
GOTERM_BP_FATGO:0000280-nuclear division22 2.64×10−18
GOTERM_BP_FAT GO:0007067-mitosis22 2.64×10−18
GOTERM_BP_FATGO:0000087-M phase of mitotic cell cycle22 3.84×10−18
GOTERM_CC_FAT GO:0005819-spindle10 4.39×10−7
GOTERM_CC_FAT GO:0000775-chromosome, centromeric region  9 1.34×10−6
GOTERM_CC_FAT GO:0000793-condensed chromosome  9 1.81×10−6
GOTERM_CC_FAT GO:0000777-condensed chromosome kinetochore  7 1.93×10−6
GOTERM_CC_FAT GO:0000779-condensed chromosome, centromeric region  7 4.16×10−6
GOTERM_MF_FATGO:0005524-ATP binding27 1.39×10−5
GOTERM_MF_FATGO:0030554-adenyl nucleotide binding28 1.49×10−5
GOTERM_MF_FATGO:0032559-adenyl ribonucleotide binding27 1.76×10−5
GOTERM_MF_FATGO:0001883-purine nucleoside binding28 1.97×10−5
GOTERM_MF_FAT GO:0001882-nucleoside binding28 2.23×10−5

B, Downregulated DEGs

IDTermCountP-value

GOTERM_BP_FATGO:0007517-muscle organ development23 2.94×10−9
GOTERM_BP_FATGO:0008285-negative regulation of cell proliferation27 2.35×10−7
GOTERM_BP_FATGO:0007155-cell adhesion39 6.17×10−7
GOTERM_BP_FAT GO:0022610-biological adhesion39 6.27×10−7
GOTERM_BP_FATGO:0003012-muscle system process16 7.10×10−6
GOTERM_CC_FAT GO:0044421-extracellular region part59 2.25×10−12
GOTERM_CC_FAT GO:0005576-extracellular region90 4.38×10−11
GOTERM_CC_FAT GO:0031012-extracellular matrix29 3.94×10−9
GOTERM_CC_FAT GO:0005578-proteinaceous extracellular matrix26 5.96×10−8
GOTERM_CC_FAT GO:0043292-contractile fiber16 8.23×10−8
GOTERM_MF_FATGO:0046870-cadmium ion binding  7 3.11×10−8
GOTERM_MF_FATGO:0003779-actin binding27 5.32×10−8
GOTERM_MF_FAT GO:0008092-cytoskeletal protein binding34 9.74×10−8
GOTERM_MF_FATGO:0005507-copper ion binding12 5.45×10−7
GOTERM_MF_FAT GO:0005198-structural molecule activity33 3.76×10−5

[i] DEGs, differentially expressed genes; count, number of DEGs; BP, biological process; CC, cell component; MF, molecule function; ATP, adenosine 5′-phosphate; GO, Gene Ontology.

According to KEGG pathway enrichment analysis, upregulated genes were significantly associated with cell cycle and oocyte meiosis signaling pathways (Table II). Downregulated DEGs were principally implicated in vascular smooth muscle and focal adhesion signaling pathways (Table II).

Table II.

KEGG pathway analysis of the upregulated and downregulated differentially expressed genes.

Table II.

KEGG pathway analysis of the upregulated and downregulated differentially expressed genes.

A, Upregulated genes

Pathway termCountP-value
hsa04110: Cell cycle  7 3.79×10−4
hsa04114: Oocyte meiosis  5 1.04×10−2
hsa00983: Drug metabolism  3 4.45×10−2

B, Downregulated genes

Pathway termCountP-value

hsa04270: Vascular smooth muscle contraction15 2.66×10−7
hsa04510: Focal adhesion16 6.41×10−5
hsa00982: Drug metabolism  9 8.87×10−5
hsa05414: Dilated cardiomyopathy  9 1.35×10−3
hsa00980: Metabolism of xenobiotics by cytochrome P450  7 2.70×10−3

[i] Count, number of differentially expressed genes; KEGG, Kyoto Encyclopedia of Genes and Genomes; hsa, human.

PPI networks construction and MCODE analysis

To investigate interactions between the DEGs, PPI networks for the DEGs were constructed. There were 69 nodes and 180 edges in the PPI network of the upregulated genes (Fig. 2A). According to the degrees of nodes, four genes were selected as the hub nodes of the PPI network, cell division cycle associated 8 (CDCA8), cell division cycle associated 5 (CDCA5, ubiquitin-conjugating enzyme E2C (UBE2C) and thymidine kinase 1 (TK1). These four DEGs interacted with >45 nodes in the PPI network, suggesting the four DEGs served crucial roles in the PPI network. One sub-network was selected from the upregulated PPI network (network aggregation score, 19.366), containing 41 nodes and 794 edges (Fig. 2B). Enrichment analysis of genes in the sub-network revealed that genes in the sub-network were primarily associated with cell cycle and cell division (Tables III and IV). Furthermore, 257 nodes and 594 edges were included in the PPI network of the downregulated genes (Fig. 3). However, no significant module was screened with the threshold of network aggregation score >10.

Table III.

Top 5 most significant genes within the sub-network of upregulated genes from GO analysis of 3 categories including BP, CC and MM.

Table III.

Top 5 most significant genes within the sub-network of upregulated genes from GO analysis of 3 categories including BP, CC and MM.

CategoryTermCountP-value
GOTERM_BP_FATGO:0000279-M phase23 4.23×10−27
GOTERM_BP_FATGO:0022403-cell cycle phase24 1.28×10−26
GOTERM_BP_FAT GO:0007067-mitosis20 3.12×10−25
GOTERM_BP_FATGO:0000280-nuclear division20 3.12×10−25
GOTERM_BP_FATGO:0000087-M phase of mitotic cell cycle20 4.44×10−25
GOTERM_CC_FAT GO:0005819-spindle11 4.94×10−13
GOTERM_CC_FAT GO:0000775-chromosome, centromeric region10 4.83×10−12
GOTERM_CC_FAT GO:0000777-condensed chromosome kinetochore  8 3.94×10−11
GOTERM_CC_FAT GO:0015630-microtubule cytoskeleton14 5.31×10−11
GOTERM_CC_FAT GO:0000779-condensed chromosome, centromeric region  8 1.01×10−10
GOTERM_MF_FATGO:0005524-ATP binding13 3.65×10−5
GOTERM_MF_FATGO:0032559-adenyl ribonucleotide binding13 4.17×10−5
GOTERM_MF_FATGO:0030554-adenyl nucleotide binding13 7.02×10−5
GOTERM_MF_FATGO:0001883-purine nucleoside binding13 8.15×10−5
GOTERM_MF_FAT GO:0001882-nucleoside binding13 8.72×10−5

[i] Count, number of differentially expressed genes; GO, Gene Ontology; BP, biological process; CC, the cell component; MF, molecule function; count, number of differentially expressed genes; ATP, adenosine 5′-phosphate.

Table IV.

KEGG pathway analysis of the sub-network of upregulated genes.

Table IV.

KEGG pathway analysis of the sub-network of upregulated genes.

CategoryTermCountP-value
KEGG_PATHWAYhsa04110: Cell cycle5 1.48×10−4
KEGG_PATHWAYhsa04114: Oocyte meiosis4 1.88×10−3

[i] Count, number of differentially expressed genes; KEGG, Kyoto Encyclopedia of Genes and Genomes; hsa, human.

Construction of the transcriptional regulatory networks

As an important regulatory element, TFs regulate the expression of certain genes (34). In the present study, 14 upregulated genes were regulated by three upregulated TFs, and 10 genes [e.g. cell division cycle 6 (CDC6) and RAD51 recombinase (RAD51)] were regulated by E2F transcription factor 2 (E2F2) (Fig. 4A). Furthermore, six TFs were predicted to target the downregulated DEGs. Notably, early growth response 1 (EGR1) regulated a number of downregulated genes and one TF, NK3 homeobox 1, in the downregulated transcriptional regulatory network (Fig. 4B).

Discussion

Prostate cancer is the most common type of reproductive system cancer in males (1,2), particularly in men over 65 years of age. In the present study, analysis of GSE38241 and GSE3933 gene expression profiles identified a total of 529 DEGs (129 up- and 400 downregulated DEGs) between the prostate cancer and normal samples. Integrative analysis of two microarray data enhanced the reliability of the present study. Enrichment analysis of the upregulated genes predicted the cell cycle to be the primary biological process in the GO function and the KEGG pathway analyses. In addition, focal adhesion pathway was identified as a significant pathway of downregulated genes. The results of the present study were consistent with those of previous studies, which demonstrated that the cell cycle and focal adhesion are required for the progression of cancer (35,36).

A total of four genes, consisting of CDCA8, CDCA5, UBE2C and TK1, exhibited a high degree of interaction in the upregulated PPI network. All four genes were involved in cell cycle-associated biological processes and signaling pathways. In the sub-network, CDCA8 interacted with pituitary tumor-transforming gene-1 (PTTG1), which was upregulated in prostate cancer. There is evidence that knockdown of PTTG1 suppresses the proliferation and invasive potential of prostate cancer cells (37). Therefore, it was hypothesized that CDCA8 may be used as a target for prostate cancer treatment, and that the interaction between CDCA8 and PTTG1 may have a role in the progression of prostate cancer. In addition, UBE2C belongs to the ubiquitin-conjugating enzyme family and participates in the process of cell mitosis (38). A previous study identified that UBE2C, as an androgen receptor target gene, was involved in the progression of prostate cancer (39). Furthermore, serological TK1 protein concentration was used as a reliable marker for the risk assessment of pre/early cancerous progression (40). However, to the best of our knowledge, there is no evidence that TK1 is a target for cancer treatment. It was hypothesized that CDCA8, CDCA5, UBE2C and TK1 may be associated with the progression of prostate cancer, and these genes were expected to be used as potential treatment targets for prostate cancer. Limited information is known about the roles and underlying molecular mechanisms of these four genes in prostate cancer; therefore, the present study may provide novel insights into the study of treatment targets for prostate cancer.

In addition to the hub genes in the PPI network, TFs targeting DEGs were identified on the basis of the transcriptional regulatory network analysis. TFs are well known to regulate the transcription of a number of genes involved in distinct signaling pathways and biological processes (34). In the present study, the upregulated TF, E2F2, and the downregulated TF EGR1 regulated a number of DEGs. E2F2 regulates genes by binding the target sequence 5′-TTTSSCGC-3′ (S=C/G) (41). In the transcriptional regulatory network, DEGs, including CDC6 and RAD51, which contain the aforementioned sequence, may be bound by E2F2 (42). CDC6 is a protein that is required for the initiation of DNA replication and has been previously identified to be deregulated in prostate cancer (43). RAD51, a protein that catalyzes DNA repair via homologous recombination, is highly expressed in cancer cells (44). Additionally, overexpression of E2F2 leads to uncontrolled proliferation of ovarian cancer cells (45) and EGR1 regulates gene expression by binding the target sequence 5′-GCGC(G/T)GGGCG-3′ (46). The downregulated gene Dickkopf WNT Signaling Pathway Inhibitor 3 (DKK3) contained this sequence and was predicted to be regulated by EGR1. DKK3 promotes the proliferation and differentiation of fibroblasts and has a function in the pathogenic stromal remodeling of prostate cancer (47). Therefore, the TFs, E2F2 and EGR1, may have marked roles in the progression of prostate cancer.

The cell cycle signaling pathway may be closely associated with prostate cancer. A total of four genes (CDCA8, CDCA5, UBE2C and TK1) and two TFs (E2F2 and EGR1) were selected, and may have important roles in the progression of prostate cancer. The selected DEGs and TFs may be used as target genes for the treatment of prostate cancer and, although they were identified using bioinformatics, the specific roles and underlying molecular mechanisms in prostate cancer require further confirmation.

References

1 

Bosetti C, Bertuccio P, Chatenoud L, Negri E, La Vecchia C and Levi F: Trends in mortality from urologic cancers in Europe, 1970–2008. Eur Urol. 60:1–15. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Chung BH: The role of radical prostatectomy in high-risk prostate cancer. Prostate Int. 1:95–101. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Dell'Oglio P, Karnes RJ, Joniau S, Spahn M, Gontero P, Tosco L, Fossati N, Kneitz B, Chlosta P and Graefen M: Very long-term survival patterns of young patients treated with radical prostatectomy for high-risk prostate cancer. Urol Oncol. 34:234. e13–9. 2016.

5 

Saad F and Miller K: Treatment options in castration-resistant prostate cancer: Current therapies and emerging docetaxel-based regimens. Urol Oncol. 32:70–79. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Thomsen FB, Brasso K, Klotz LH, Røder MA, Berg KD and Iversen P: Active surveillance for clinically localized prostate cancer-a systematic review. J Surg Oncol. 109:830–835. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Cole G, McCaffrey J, Ali AA and McCarthy HO: DNA vaccination for prostate cancer: Key concepts and considerations. Cancer Nanotechnol. 6:22015. View Article : Google Scholar : PubMed/NCBI

8 

Cross D and Burmester JK: Gene therapy for cancer treatment: Past, present and future. Clin Med Res. 4:218–27. 2006. View Article : Google Scholar : PubMed/NCBI

9 

van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, Hoogerbrugge N, Verhoef S, Vasen HF, Ausems MG, Menko FH, Garcia Gomez EB and Klijn JG: Cancer risks in BRCA2 families: Estimates for sites other than breast and ovary. J Med Genet. 42:711–719. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Leongamornlert D, Mahmud N, Tymrakiewicz M, Saunders E, Dadaev T, Castro E, Goh C, Govindasami K, Guy M and O'Brien L: Germline BRCA1 mutations increase prostate cancer risk. Br J Cancer. 106:1697–1701. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Khandrika L, Kumar B, Koul S, Maroni P and Koul HK: Oxidative stress in prostate cancer. Cancer Lett. 282:125–136. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Campaner S and Amati B: Two sides of the Myc-induced DNA damage response: From tumor suppression to tumor maintenance. Cell Div. 7:62012. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Ngalame NN, Tokar EJ, Person RJ, Xu Y and Waalkes MP: Aberrant microRNA expression likely controls RAS oncogene activation during malignant transformation of human prostate epithelial and stem cells by arsenic. Toxicol Sci. 138:268–277. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, Ghosh D, Pienta KJ, Sewalt RG and Otte AP: The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 419:624–629. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Conesa-Zamora P, García-Solano J, García-García F, Mdel Turpin C, Trujillo-Santos J, Torres-Moreno D, Oviedo-Ramírez I, Carbonell-Muñoz R, Muñoz-Delgado E and Rodriguez-Braun E: Expression profiling shows differential molecular pathways and provides potential new diagnostic biomarkers for colorectal serrated adenocarcinoma. Int J Cancer. 132:297–307. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Wu C, Zhu J and Zhang X: Network-based differential gene expression analysis suggests cell cycle related genes regulated by E2F1 underlie the molecular difference between smoker and non-smoker lung adenocarcinoma. BMC Bioinformatics. 14:3652013. View Article : Google Scholar : PubMed/NCBI

18 

Li Y, Vongsangnak W, Chen L and Shen B: Integrative analysis reveals disease-associated genes and biomarkers for prostate cancer progression. BMC Med Genomics. 7 (Suppl 1):S32014. View Article : Google Scholar : PubMed/NCBI

19 

Aryee MJ, Liu W, Engelmann JC, Nuhn P, Gurel M, Haffner MC, Esopi D, Irizarry RA, Getzenberg RH and Nelson WG: DNA methylation alterations exhibit intraindividual stability and interindividual heterogeneity in prostate cancer metastases. Sci Transl Med. 5:169ra102013. View Article : Google Scholar : PubMed/NCBI

20 

Lapointe J, Li C, Higgins JP, van de Rijn M, Bair E, Montgomery K, Ferrari M, Egevad L, Rayford W and Bergerheim U: Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci USA. 101:811–816. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Johnson WE, Li C and Rabinovic A: Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 8:118–127. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Leek JT and Storey JD: A general framework for multiple testing dependence. Proc Natl Acad Sci USA. 105:18718–18723. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

24 

Pounds S and Morris SW: Estimating the occurrence of false positives and false negatives in microarray studies by approximating and partitioning the empirical distribution of P-values. Bioinformatics. 19:1236–1242. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Shoop E, Casaes P, Onsongo G, Lesnett L, Petursdottir EO, Donkor EK, Tkach D and Cosimini M: Data exploration tools for the gene ontology database. Bioinformatics. 20:3442–3454. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Kanehisa M, Goto S, Kawashima S, Okuno Y and Hattori M: The KEGG resource for deciphering the genome. Nucleic Acids Res. 32:D277–280. 2004. View Article : Google Scholar : PubMed/NCBI

27 

da Huang W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protoc. 4:44–57. 2009. View Article : Google Scholar

28 

da Huang W, 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 

Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, von Mering C and Jensen LJ: STRING v9.1: Protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41:D808–D815. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Albert R, Albert I and Nakarado GL: Structural vulnerability of the North American power grid. Physical Review E. 69:0251032004. View Article : Google Scholar

32 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

33 

Matys V, Kel-Margoulis OV, Fricke E, Liebich I, Land S, Barre-Dirrie A, Reuter I, Chekmenev D, Krull M and Hornischer K: TRANSFAC and its module TRANSCompel: Transcriptional gene regulation in eukaryotes. Nucleic Acids Res. 34:D108–110. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Chen K and Rajewsky N: The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. 8:93–103. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Seong J, Wang N and Wang Y: Mechanotransduction at focal adhesions: From physiology to cancer development. J Cell Mol Med. 17:597–604. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Fu LJ and Wang B: Investigation of the hub genes and related mechanism in ovarian cancer via bioinformatics analysis. J Ovarian Res. 6:922013. View Article : Google Scholar : PubMed/NCBI

37 

Huang SQ, Liao QJ, Wang XW, Xin DQ, Chen SX, Wu QJ and Ye G: RNAi-mediated knockdown of pituitary tumor-transforming gene-1 (PTTG1) suppresses the proliferation and invasive potential of PC3 human prostate cancer cells. Braz J Med Biol Res. 45:995–1001. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Summers MK, Pan B, Mukhyala K and Jackson PK: The unique N terminus of the UbcH10 E2 enzyme controls the threshold for APC activation and enhances checkpoint regulation of the APC. Mol Cell. 31:544–556. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Chen Z, Zhang C, Wu D, Chen H, Rorick A, Zhang X and Wang Q: Phospho-MED1-enhanced UBE2C locus looping drives castration-resistant prostate cancer growth. EMBO J. 30:2405–2419. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Huang S, Lin J, Guo N, Zhang M, Yun X, Liu S, Zhou J, He E and Skog S: Elevated serum thymidine kinase 1 predicts risk of pre/early cancerous progression. Asian Pac J Cancer Prev. 12:497–505. 2011.PubMed/NCBI

41 

Slansky JE and Farnham PJ: Introduction to the E2F family: Protein structure and gene regulation. Curr Top Microbiol Immunol. 208:1–30. 1996.PubMed/NCBI

42 

Bracken AP, Ciro M, Cocito A and Helin K: E2F target genes: Unraveling the biology. Trends Biochem Sci. 29:409–417. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Liu Y, Gong Z, Sun L and Li X: FOXM1 and androgen receptor co-regulate CDC6 gene transcription and DNA replication in prostate cancer cells. Biochim Biophys Acta. 1839:297–305. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Mason JM, Logan HL, Budke B, Wu M, Pawlowski M, Weichselbaum RR, Kozikowski AP, Bishop DK and Connell PP: The RAD51-stimulatory compound RS-1 can exploit the RAD51 overexpression that exists in cancer cells and tumors. Cancer Res. 74:3546–3555. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Reimer D, Sadr S, Wiedemair A, Concin N, Hofstetter G, Marth C and Zeimet AG: Heterogeneous cross-talk of E2F family members is crucially involved in growth modulatory effects of interferon-gamma and EGF. Cancer Biol Ther. 5:771–776. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Liu C, Rangnekar VM, Adamson E and Mercola D: Suppression of growth and transformation and induction of apoptosis by EGR-1. Cancer Gene Ther. 5:3–28. 1998.PubMed/NCBI

47 

Zenzmaier C, Sampson N, Plas E and Berger P: Dickkopf-related protein 3 promotes pathogenic stromal remodeling in benign prostatic hyperplasia and prostate cancer. Prostate. 73:1441–1452. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 14 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao R, Wang Y, Zhang M, Gu X, Wang W, Tan J, Wei X and Jin N: Screening of potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles. Oncol Lett 14: 5361-5369, 2017
APA
Zhao, R., Wang, Y., Zhang, M., Gu, X., Wang, W., Tan, J. ... Jin, N. (2017). Screening of potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles. Oncology Letters, 14, 5361-5369. https://doi.org/10.3892/ol.2017.6879
MLA
Zhao, R., Wang, Y., Zhang, M., Gu, X., Wang, W., Tan, J., Wei, X., Jin, N."Screening of potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles". Oncology Letters 14.5 (2017): 5361-5369.
Chicago
Zhao, R., Wang, Y., Zhang, M., Gu, X., Wang, W., Tan, J., Wei, X., Jin, N."Screening of potential therapy targets for prostate cancer using integrated analysis of two gene expression profiles". Oncology Letters 14, no. 5 (2017): 5361-5369. https://doi.org/10.3892/ol.2017.6879