Identification of hub genes and pathways associated with bladder cancer based on co-expression network analysis

  • Authors:
    • Dong‑Qing Zhang
    • Chang‑Kuo Zhou
    • Shou‑Zhen Chen
    • Yue Yang
    • Ben‑Kang Shi
  • View Affiliations

  • Published online on: May 26, 2017     https://doi.org/10.3892/ol.2017.6267
  • Pages: 1115-1122
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 identify hub genes and signaling pathways associated with bladder cancer (BC) utilizing centrality analysis and pathway enrichment analysis. The differentially expressed genes (DEGs) were screened from the ArrayExpress database between normal subjects and BC patients. Co‑expression networks of BC were constructed using differentially co‑expressed genes and links, and hub genes were investigated by degree centrality analysis of co‑expression networks in BC. The enriched signaling pathways were investigated by Kyoto Encyclopedia of Genes and Genomes database analysis based on the DEGs. The hub gene expression in BC tissues was validated using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blotting. A total of 329 DEGs were screened, including 147 upregulated and 182 downregulated genes. The co‑expression network constructed between BC and normal controls consisted of 182 nodes and 434 edges, and the two genes in each gene pair were differentially co‑expressed genes. Centrality analysis of co‑expression networks suggested that the top 5 hub genes with high degree included lectin, galactoside‑binding, soluble, 4 (LGALS4), protein tyrosine phosphatase, receptor type N2 (PTPRN2), transmembrane protease, serine 11E (TMPRSS11E), tripartite motif containing 31 (TRIM31) and potassium voltage‑gated channel subfamily D member 3 (KCND3). Pathway analysis revealed that the 329 DEGs were significantly enriched in 5 terms (cell cycle, DNA replication, oocyte meiosis, p53 signaling pathway and peroxisome proliferator‑activated receptor signaling pathway). According to RT‑qPCR and western blot analysis, 4/5 hub genes were significantly expressed, including LGALS4, PTPRN2, TMPRSS11E, TRIM31; however, KCND3 was not significantly expressed. In the present study, 5 hub genes were successfully identified (LGALS4, PTPRN2, TMPRSS11E, TRIM31 and KCND3) and 5 biological pathways that may be underlying biomarkers for early diagnosis and treatment associated with bladder cancer were revealed.

Introduction

Bladder cancer (BC) is the fourth most common cancer and the eighth leading cause of cancer-associated mortality worldwide. BC is an important public health issue as it is biologically very aggressive and highly prevalent in Western countries (1). In 2017, an estimated 79,030 new cases of BC and 16,870 mortalities will occur in the USA (2). In China, the incidence and mortality rates of BC rank the highest among tumors of the male urogenital system (3). Painless hematuria is the main symptom of BC, and its diagnosis is established based on urinary cytology and transurethral tumor resection (4). There is an increasing trend in incidence and mortality rates of BC. Numerous studies have identified various risk factors that may induce BC, including geography, race, gender, schistosomiasis infection, environmental or occupational exposure, smoking and genetic susceptibility (57).

Tumor progression is a complicated procedure of cancer cell development from normal epithelial cells, which involves changes in various genes, including oncogenes, cell cycle-associated genes, tumor suppressor genes and DNA damage repair genes. These are potential tumor markers in clinical practice; however, additional clinical studies are required to confirm their clinical utility (810). Development of molecular biology has increased the understanding of the mechanism underlying BC. Dyrskjøt et al (11) detected carcinoma in situ gene expression that is reflected in carcinoma in situ biopsies and superficial transitional cell carcinoma. Biton et al (12) demonstrated that a molecular urothelial differentiation program was maintained by applying independent component analysis to bladder cancer transcriptome data and exploiting additional molecular and clinic pathological data.

Network-based approaches, particularly co-expression networks, offer a more effective means to identify potential malignancy diagnostic molecules based on connecting them together. Co-expression networks are generally used to study disease mechanisms (13) and provide a systems level view of dysregulated signaling pathways (14). The basic premise of co-expression analysis is that strongly correlated genes are likely to be functionally associated. Furthermore, it is possible to gain a clear insight into the important tumorigenic genes and signaling pathways of a variety of diseases, many of which are applicable to early detection and treatment (15).

In order to investigate the molecular and genetic mechanisms of BC, the present study identified the differentially expressed genes (DEGs) between BC and normal controls and constructed co-expression networks of BC via differentially co-expressed genes and links (DCGL); subsequently, the hub genes and pathways were further investigated by centrality analysis and using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, respectively. In addition, the hub genes that were potential tumor markers for BC progression were validated in BC tissues using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting.

Materials and methods

Data collection and preprocessing

A total of two gene expression datasets from healthy people and BC patients, including E-MTAB-1940 and E-GEOD-3167, were obtained from the ArrayExpress database (www.ebi.ac.uk/arrayexpress/). A total of 146 samples (128 cases and 18 controls) were collected in the present study. The dataset E-MTAB-1940 (12) included 4 controls (samples from normal bladders) and 82 cases (samples from BC tissue); the dataset E-GEOD-3167 (11) included 14 controls and 46 cases.

Data preprocessing for all original expression information was performed prior to the analysis. In order to reduce the influence of nonspecific factors about, the background correction and normalization were performed using the robust multichip average method (16) and the quantile based algorithm (17), respectively. Perfect match and mismatch values were revised and selected using the Micro Array Suite 5.0 (MAS 5.0) algorithm (18) and the median method, respectively. Subsequently, the data were screened by the feature filter method of the genefilter package (http://bioconductor.org/packages/release/bioc/html/genefilter.html). Each probe was mapped to one gene using getSYMBOL, whoch is is a function in package annotate of the genefilter package (http://www.bioconductor.org/packages/release/bioc/html/annotate.html), and the probe was discarded if it did not match any genes. The two expression datasets were merged and synthetically analyzed using Batch Mean-centering, a merged data method (19), following adaptation according to Support Vector Machines, through the inSilicoMerging package (20).

Detection of DEGs

In the present study, the DEGs between BC patients and normal controls were screened by the linear models for microarray data (LIMMA) package (21). t-tests and F-tests were performed on the matrix, and the P-values were transformed to -log10. Empirical Bayes (22) statistics and false discovery rate (23) calibration of P-values for the data were conducted using the lmFit function (lmFit:http://lmfit.github.io//lmfit-py/). The DEGs were selected from following inspection for the following cut-off criteria: values of |log fold change (FC)|≥2 and P<0.05.

Construction of differential co-expression networks by DCGL

It is critical to construct a co-expression network for identifying modules and the intra-modular connectivity. DCGL 2.0 (24) is an R package for identifying differentially co-expressed genes (DCGs) and differentially co-expressed links (DCLs) from gene expression microarray data. It examines the expression correlation based on the exact co-expression changes of gene pairs between two conditions, and thus is able to differentiate significant co-expression changes from relatively trivial ones (25). It has four functional modules: Gene filtration, link filtration, differential co-expression analysis (DCEA) and differential regulation analysis. Differential co-expression profile (DCp) and differential co-expression enrichment (DCe) are involved in the DCEA module for extracting DCGs and DCLs. DCp worked on the filtered set of gene co-expression value pairs, where each pair was composed of two co-expression values worked out under two different conditions separately (24,26). The present study used a length-normalized Euclidean distance to measure differential co-expression (dC) of the co-expression value pairs associated with a particular gene. A permutation test was performed to assess the significance of dC. The sample permutation was repeated N times, and a large number of permutation dC statistics formed an empirical null distribution. The P-value for each gene could then be estimated.

DCe was also used to identify DCGs and DCLs, which were based on the ‘Limit Fold Change’ (LFC) model. Initially, correlation pairs were divided into 3 parts according to the pairing of signs of co-expression values and the multitude of co-expression values: Pairs with the same signs, pairs with different signs and pairs with differently-signed high co-expression values. The first two groups were processed with the ‘LFC’ model separately to produce two subsets of DCLs, while the third group was added to the set of DCLs directly. Therefore, a total of DCLs was determined from all gene links.

Centrality analysis

To additionally assess the key genes of BC, a centricity analysis was performed based on the nodes degree in the networks (27). Centrality measures mainly contain degree centrality, closeness centrality and shortest path between centrality, in which degree, the equivalent of the number of nodes directly adjacent to a given node (indicating the degree the vertex), is the simplest topological index (27). In the present study, centrality analysis, which was particularly useful to identify key players in biological processes, was implemented to study the differential co-expression networks. Calculation of the degree allows determining the ‘degree distribution’, which gives the probability that a selected node has exact links. Nodes with high degree (highly connected) are called ‘hubs’, which interact with several other genes, suggesting a central role in the interaction network (28). An obvious order of the vertices of a graph may be established by sorting them according to their degree (29). Genes with degree ≥9 were defined as hub genes in the present study.

Pathway enrichment analysis

To investigate the enriched signaling pathways of the DEGs, enrichment analysis was performed based on the KEGG database (www.genome.jp/kegg/). The DEGs were applied to this database for investigating the association between the biochemical pathways and the occurrence of bladder cancer. The analysis was conducted by DAVID (30) (david.abcc.ncifcrf.gov/tools.jsp). The enrichment pathway analysis of the DEGs contributed to additional observation of these genes at the macroscopic level. Categories were obtained according to the Expression Analysis Systematic Explorer (31) score for 0.01.

Patients

Tumor tissues from 10 BC patients were obtained during biopsy in Qilu Hospital of Shandong University (Jinan, China) between January 2015 and March 2015. Normal samples were collected from 2 cm away from the tumor. The present study was approved by the Ethical Committee of Qilu Hospital of Shandong University. Written informed consent was obtained from patients who agreed to participate in the study.

Validation of hub genes by RT-qPCR and western blotting

The tumor tissues of 10 BC patients and normal tissue samples obtained from 2 cm away from the tumor were analyzed with polymorphic DNA markers, and total RNA was prepared using TRIzol (Beyotime Institute of Biotechnology- Haimen, China). Total RNA was reverse transcribed with an oligo (dT18) primer and was treated with 2 µl RNasin (40 U/µl), 8.0 µl 5X reverse transcriptase buffer, 8.0 µl dNTPs and 2 µl AMV reverse transcriptase (5 U/µl) according to the manufacturer's protocol. The RT reagents were purchased from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA). For RT-qPCR, primer sequences of the 5 hub genes are listed in Table I. The qPCR mix composition was as follows: 10 µl of 10X qPCR buffer, 1 µl of Taq DNA polymerase, 3 µl of each forward and reverse primer and 8 µl of dNTPs. The qPCR reagents were purchased from Invitrogen (Thermo Fisher Scientific, Inc.). The qPCR conditions are shown in Table II. Complementary DNA was used as a template, and β-actin was used as an internal reference. RT-PCR products were separated using 1.5% agarose gel electrophoresis and gels were visualized using Quantity One Software v4.62 (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Every sample was run 3 times along with the internal control.

Table I.

Primer sequences and product length of the 5 hub genes.

Table I.

Primer sequences and product length of the 5 hub genes.

GenePrimer sequences (5′-3)Length (bp)
LGALS4F: GCCTTCCACTTCAATCCGC355
R: GGCTGTTCAGCTGTTGATGG
PTPRN2F: GTGGACCATGGAGTAGCTCG541
R: GTCCGAGAACCTCTCTGTCT
TMPRSS11EF: GTCTCAGGATCGTTGGTGGG720
R: ACAAGAAAGTTGGCAAGATACCAGT
TRIM31F: GTCTTGTGCAGAAGTGAAGAGTT178
R: TCACAAAACCAAGCCCGGAT
KCND3F: TTTACACTGGAGGTGGGGGA506
R: TGCAGTGCGATTTCAGGTCT
β-actinF: AAGTACTCCGTGTGGATCGG615
R: TCAAGTTGGGGGACAAAAAG

[i] LGALS4, lectin, galactoside-binding, soluble, 4; PTPRN2, protein tyrosine phosphatase, receptor type N2; TMPRSS11E, transmembrane protease, serine 11E; TRIM31, tripartite motif containing 31; KCND3, potassium voltage-gated channel subfamily D member 3; F, forward; R, reverse; bp, base pairs.

Table II.

Quantitative polymerase chain reaction amplification conditions for the 5 hub genes.

Table II.

Quantitative polymerase chain reaction amplification conditions for the 5 hub genes.

GeneReaction conditions
LGALS494°C 1 min; 35 cycles of 98°C 10 sec, 54°C 15 sec, 68°C 1 min; 72°C 7 min
PTPRN294°C 2 min; 35 cycles of 98°C 10 sec, 50°C 15 sec, 68°C 1 min; 72°C 7 min
TMPRSS11E94°C 2 min; 33 cycles of 98°C 10 sec, 55°C 15 sec, 68°C 1 min; 72°C 7 min
TRIM3194°C 2 min; 35 cycles of 98°C 10 sec, 54°C 15 sec, 68°C 1 min; 72°C 7 min
KCND394°C 2 min; 33 cycles of 98°C 10 sec, 54°C 15 sec, 68°C 1 min; 72°C 7 min
β-actin94°C 2 min; 30 cycles of 98°C 10 sec, 51°C 15 sec, 68°C 1 min; 72°C 7 min

[i] LGALS4, lectin, galactoside-binding, soluble, 4; PTPRN2, protein tyrosine phosphatase, receptor type N2; TMPRSS11E, transmembrane protease, serine 11E; TRIM31, tripartite motif containing 31; KCND3, potassium voltage-gated channel subfamily D member 3.

Proteins were extracted from tumor tissues and adjacent normal tissues according to the method reported by Yoon et al (32). Protein concentration was measured with a BCA Protein Assay kit (CW Biotech, Beijing, China). SDS-PAGE (12%) was performed for 10 µg protein, and the protein was electrotransferred (4°C, 300 mA, 2 h) to a nitrocellulose membrane. The membrane was blocked in TBST containing 5% skimmed milk powder at 37°C for 2 h, and incubated with rabbit anti-human antibodies (1:10,000) against lectin, galactoside-binding, soluble, 4 (LGALS4) [AP12391a; Abgent Biotech (SuZhou) Co., Ltd., Suzhou, China], protein tyrosine phosphatase, receptor type N2 (PTPRN2) (bs-19591R; BIOSS, Beijing, China), transmembrane protease, serine 11E (TMPRSS11E) [AP16520C; Abgent Biotech (SuZhou) Co., Ltd.], tripartite motif containing 31 (TRIM31) (bs-6220R; BIOSS), potassium voltage-gated channel subfamily D member 3 (KCND3) (bs-20219R; BIOSS) and GAPDH [AP50811; Abgent Biotech (SuZhou) Co., Ltd.] at 37°C for 2 h. Unbound antibody was washed away by TBST (3 times), and the membrane was incubated with horseradish peroxdiase-labeled sheep anti-rabbit immunoglobulin G secondary antibody (1:5,000; SC-2048; Beijing Zhongshan Jinqiao Biotechnology Co., Ltd., Beijing, China) at 37°C for 2 h. Following washing with TBST, the substrate was developed on the membrane for 3 min and exposed in the dark. Protein bands were visualized with Amersham ECL Western Blotting Detection kit (GE Healthcare Life Sciences, Chalfont, UK). GAPDH was used as an internal control, and western blots were analyzed using ImageJ 1.36b software (National Institutes of Health, Bethesda, MD, USA; http://rsb.info.nih.gov/ij/).

Statistical analysis

Statistical analysis was conducted using SPSS 20.0 software (IBM SPSS, Armonk, NY, USA). All data were presented as mean ± standard deviation. Statistical differences among groups were assessed using one way analysis of variance. P<0.05 was considered to indicate a tatistically significanct difference. Graphs were designed using GraphPad Prism 5.0 (GraphPad Software, Inc., La Jolla, CA, USA).

Results

Detection of DEGs

In the present study, a total of 146 samples of two datasets associated with BC were preprocessed to identify DEGs using the LIMMA package. A total of 329 DEGs, including 147 upregulated and 182 downregulated genes between BC patients and normal subjects were detected under the criteria of |logFC|≥2 and P<0.05.

Construction of differential co-expression networks by DCGL

The present study applied the DCGL 2.0 package in R to identify DCGs and DCLs, in which DCp and DCe methods involved in DCEA module were employed. A total of 434 DCLs were included in the co-expression network, and the two genes in each link were DCGs. A gene co-expression network containing 182 nodes and 434 edges was constructed in the analysis (Fig. 1).

Centrality analysis

By accessing degree centrality analysis of the co-expression network, 18 hub genes were obtained with degree ≥9 (Table III), of which 16 genes were upregulated and 2 were downregulated (Fig. 2). The genes LGALS4, PTPRN2, TMPRSS11E, TRIM31 and KCND3 were the top 5 hub genes with high degree, and all of them were upregulated.

Table III.

Eighteen hub genes where the degree was ≥9 in the co-expression network.

Table III.

Eighteen hub genes where the degree was ≥9 in the co-expression network.

GenesDegree
LGALS466
PTPRN256
TMPRSS11E45
TRIM3139
KCND335
NFIB34
LEPREL131
SH3GL229
CFI27
ADTRP21
PIGR16
MME15
ATF313
MMP712
PRKAR2B12
SCD510
IGF29
SF3B49
Pathway enrichment analysis

Pathway analysis based on the KEGG database revealed that these DEGs were significantly enriched in 5 terms, which were cell cycle (P=4.37×10−7), DNA replication (P=1.95×10−3), oocyte meiosis (P=6.47×10−3), p53 signaling pathway (P=7.17×10−3) and the peroxisome proliferator-activated receptor (PPAR) signaling pathway (P=7.70E-03). Among the 5 terms, the cell cycle pathway was the most significant term, which also enriched more genes compared with the other terms.

Validation by RT-qPCR and western blotting

In the present study, to confirm the messenger RNA (mRNA) and protein expression levels of 5 hub genes (LGALS4, PTPRN2, TMPRSS11E, TRIM31 and KCND3) from the co-expression network, RT-qPCR and western blotting was performed on BC patient samples. The results of the relative expression of all the hub genes are shown in Figs. 3 and 4. It was observed that 4/5 hub genes and the corresponding proteins including LGALS4, PTPRN2, TMPRSS11E and TRIM31 were significantly differentially expressed between BC patients and healthy subjects (P<0.05), while KCND3 was not significantly differentially expressed (P>0.05). Furthermore, it was noted that the relative expression of LGALS4, TMPRSS11E and TRIM31 was increasing, which was consistent with the result of the bioinformatics analysis, while PTPRN2 demonstrated the opposite trend. KCND3, which was downregulated in the bioinformatics investigation, was not significantly differentially expressed.

Discussion

BC is a heterogeneous disease, with 30% of cases presenting as muscle-invasive disease associated with a high risk of death from distant metastases, which may be managed with transurethral resection (33). However, BC has a notable tendency to recur (30–85%), therefore the present study investigated an adequate method for investigating biomarkers in BC, that may contribute to our understanding of the pathogenesis and diagnoses of the disease, substantially reducing the mortality associated with this disease.

In the present study, hub genes and pathways of BC were identified based on degree centrality analysis of the co-expression network and pathway enrichment analysis. A total of 18 hub genes were obtained, the top 5 of which were LGALS4, PTPRN2, TMPRSS11E, TRIM31 and KCND3, by conducting centrality analysis on the co-expression network. Furthermore, cell cycle, DNA replication, oocyte meiosis, the p53 signaling pathway and the PPAR signaling pathway were observed to be significant pathways of BC. In addition, RT-qPCR and western blotting were performed to verify network-based results and investigate significant genes of BC.

RT-qPCR analysis revealed that 3 hub genes (LGALS4, TMPRSS11E and TRIM31) were significantly upregulated in BC patients when compared with normal subjects, which was consistent with the bioinformatics analysis; however, KCND3 was not significantly differentially expressed between the conditions, and in contrast to the bioinformatics result, the relative content of the upregulated PTPRN2 was significantly reduced. This result was not entirely consistent with the network analysis. The probable reason for this was variations of samples; the microarray data was downloaded from the ArrayExpress database and RT-qPCR and western blotting were performed on patient samples. Therefore, it can be speculated that the 3 consistent hub genes (LGALS4, TMPRSS11E and TRIM31) may be potential markers of BC. LGALS4 has been identified as one of the genes involved in numerous types of human tumor, including sinonasal adenocarcinoma tumors (34), colorectal cancer (35) and breast cancer (36). There has also been a previous report that compared expression changes at mRNA and protein levels in the rat model and identified the gene exhibiting concordant changes with LGALS4 levels in bladder tumors (37). TRIM family proteins are involved in various cellular processes, including tumor development and antiviral response (38). One of the family proteins, TRIM31, was originally identified as a gene induced by growth-suppressive retinoid (39). A previous study showed that TRIM31 had the ability to regulate cell proliferation negatively in gastric adenocarcinoma (40) and its expression was reduced in lung cancer cell lines (41). However, the expression level of TRIM31 was increased in BC patients in the present study. Additional research is required to verify the biological properties of TRIM31.

Pathway analysis revealed that several significantly enriched signaling pathways included cell cycle, DNA replication, oocyte meiosis, the p53 signaling pathway and the PPAR signaling pathway. The cell cycle is the universal process through which cells reproduce and grow in all living organisms, and is concerned with the copying of the hereditary material, including replication of the chromosomal DNA during mitosis (42). Previous studies have indicated that several tumor suppressor candidates exert growth inhibitory effects by inducing cell cycle arrest at G2/M phase in leukemia cells (43), and exerted different tumor suppressive effects (44). In addition, Li et al (45) indicated the effect of the cell cycle on the susceptibility of SAS cells to sonodynamic therapy. p53 is a sequence-specific DNA-binding protein that promotes cell-cycle arrest or apoptosis in response to a variety of cellular stresses (46). The p53 signaling pathway had been suggested as a cellular surveillance mechanism for cancer prevention (47). Furthermore, drug development programs are underway to target the p53 signaling pathway (48).

In conclusion, the present study identified 5 hub genes associated with BC, and 3 of these were verified via molecular experiments, RT-qPCR and western blotting. The signaling pathways associated with these genes were presented systematically. These genes and pathways may be potential biomarkers for early detection and therapy for BC.

Glossary

Abbreviations

Abbreviations:

BC

bladder cancer

DEGs

differentially expressed genes

DCGL

differentially co-expressed genes

KEGG

Kyoto encyclopedia of genes and genomes

RT-qPCR

reverse transcription-quantitative polymerase chain reaction

DCGs

differentially co-expressed genes

DCLs

differentially co-expressed links

DCEA

differential co-expression analysis

DCp

differential co-expression profile

DCe

differential co-expression enrichment

References

1 

Rubio G, García-Mora B, Santamaria C and Pontones JL: A flowgraph model for bladder carcinoma. Theor Biol Med Model. 11 Suppl 1:S32014. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Chen WQ, Zeng HM, Zheng RS, Zhang SW and He J: Cancer incidence and mortality in China, 2007. Chin J Cancer Res. 24:1–8. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Sharma S, Ksheersagar P and Sharma P: Diagnosis and treatment of bladder cancer. Am Fam Physician. 80:717–723. 2009.PubMed/NCBI

5 

Burger M, Catto JW, Dalbagni G, Grossman HB, Herr H, Karakiewicz P, Kassouf W, Kiemeney LA, La Vecchia C, Shariat S and Lotan Y: Epidemiology and risk factors of urothelial bladder cancer. Eur Urol. 63:234–241. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Wyszynski A, Tanyos SA, Rees JR, Marsit CJ, Kelsey KT, Schned AR, Pendleton EM, Celaya MO, Zens MS, Karagas MR and Andrew AS: Body mass and smoking are modifiable risk factors for recurrent bladder cancer. Cancer. 120:408–414. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Letašiová S, Medve'ová A, Šovčíková A, Dušinská M, Volkovová K, Mosoiu C and Bartonová A: Bladder cancer, a review of the environmental risk factors. Environ Health. 11 Suppl 1:S112012. View Article : Google Scholar : PubMed/NCBI

8 

Xue Y, Wu G, Wang X, Zou X, Zhang G, Xiao R, Yuan Y, Long D, Yang J, Wu Y, et al: CIP2A is a predictor of survival and a novel therapeutic target in bladder urothelial cell carcinoma. Med Oncol. 30:4062013. View Article : Google Scholar : PubMed/NCBI

9 

Tabata K, Matsumoto K, Minami S, Ishii D, Nishi M, Fujita T, Saegusa M, Sato Y and Iwamura M: Nestin is an independent predictor of cancer-specific survival after radical cystectomy in patients with urothelial carcinoma of the bladder. PLoS One. 9:e915482014. View Article : Google Scholar : PubMed/NCBI

10 

Yan Y, Yang FQ, Zhang HM, Li J, Li W, Wang GC, Che JP, Zheng JH and Liu M: Bromodomain 4 protein is a predictor of survival for urothelial carcinoma of bladder. Int J Clin Exp Pathol. 7:4231–4238. 2014.PubMed/NCBI

11 

Dyrskjøt L, Kruhøffer M, Thykjaer T, Marcussen N, Jensen JL, Møller K and Ørntoft TF: Gene expression in the urinary bladder: A common carcinoma in situ gene expression signature exists disregarding histopathological classification. Cancer Res. 64:4040–4048. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Biton A, Bernard-Pierrot I, Lou Y, Krucker C, Chapeaublanc E, Rubio-Pérez C, López-Bigas N, Kamoun A, Neuzillet Y, Gestraud P, et al: Independent component analysis uncovers the landscape of the bladder tumor transcriptome and reveals insights into luminal and basal subtypes. Cell Rep. 9:1235–1245. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Nayak RR, Kearns M, Spielman RS and Cheung VG: Coexpression network based on natural variation in human gene expression reveals gene interactions and functions. Genome Res. 19:1953–1962. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, Mill J, Cantor RM, Blencowe BJ and Geschwind DH: Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature. 474:380–384. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Kristensen VN, Lingjærde OC, Russnes HG, Vollan HK, Frigessi A and Børresen-Dale AL: Principles and methods of integrative genomic analyses in cancer. Nat Rev Cancer. 14:299–313. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Ma L, Robinson LN and Towle HC: ChREBP*Mlx is the principal mediator of glucose-induced gene expression in the liver. J Biol Chem. 281:28721–28730. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Rifai N and Ridker PM: Proposed cardiovascular risk assessment algorithm using high-sensitivity C-reactive protein and lipid screening. Clin Chem. 47:28–30. 2001.PubMed/NCBI

18 

Pepper SD, Saunders EK, Edwards LE, Wilson CL and Miller CJ: The utility of MAS5 expression summary and detection call algorithms. BMC Bioinformatics. 8:2732007. View Article : Google Scholar : PubMed/NCBI

19 

Sims AH, Smethurst GJ, Hey Y, Okoniewski MJ, Pepper SD, Howell A, Miller CJ and Clarke RB: The removal of multiplicative, systematic bias allows integration of breast cancer gene expression datasets - improving meta-analysis and prediction of prognosis. BMC Med Genomics. 1:422008. View Article : Google Scholar : PubMed/NCBI

20 

Taminau J, Taminau MJ, Meganck S and BiocGenerics S: Package ‘inSilicoMerging’. 2013.https://bioc.ism.ac.jp/packages/3.1/bioc/html/inSilicoMerging.htmlFeb 06–2015

21 

Smyth G, Thorne N and Wettenhall J: LIMMA: Linear Models for Microarray Data User's Guide. 2003.http://www. bioconductor.org2005Feb 08–2015

22 

Datta S, Satten GA, Benos DJ, Xia J, Heslin MJ and Datta S: An empirical bayes adjustment to increase the sensitivity of detecting differentially expressed genes in microarray experiments. Bioinformatics. 20:235–242. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Reiner A, Yekutieli D and Benjamini Y: Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics. 19:368–375. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Yang J, Yu H, Liu BH, Zhao Z, Liu L, Ma LX, Li YX and Li YY: DCGL v2. 0: An R package for unveiling differential regulation from differential co-expression. PLoS One. 8:e797292013. View Article : Google Scholar : PubMed/NCBI

25 

Yu H, Liu BH, Ye ZQ, Li C, Li YX and Li YY: Link-based quantitative methods to identify differentially coexpressed genes and gene pairs. BMC Bioinformatics. 12:3152011. View Article : Google Scholar : PubMed/NCBI

26 

Liu BH, Yu H, Tu K, Li C, Li YX and Li YY: DCGL: An R package for identifying differentially coexpressed genes and links from gene expression microarray data. Bioinformatics. 26:2637–2638. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Scardoni G and Laudanna C: Centralities based analysis of complex networksNew Frontiers in Graph Theory. ISBN, 978-953-51-0115-4. INTECH Open Access Publisher; 2012, http://www.intechopen.com/books/new-frontiers-in-graph-theory/centralities-based-analysis-of-networks View Article : Google Scholar

28 

Zotenko E, Mestre J, O'Leary DP and Przytycka TM: Why do hubs in the yeast protein interaction network tend to be essential: Reexamining the connection between the network topology and essentiality. PLoS Comput Biol. 4:e10001402008. View Article : Google Scholar : PubMed/NCBI

29 

Koschützki D and Schreiber F: Centrality analysis methods for biological networks and their application to gene regulatory networks. Gene Regul Syst Bio. 2:193–201. 2008.PubMed/NCBI

30 

Huang DW, Sherman BT, Tan Q, Collins JR, Alvord WG, 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

31 

Hosack DA, Dennis G Jr, Sherman BT, Lane HC and Lempicki RA: Identifying biological themes within lists of genes with EASE. Genome Biol. 4:R702003. View Article : Google Scholar : PubMed/NCBI

32 

Yoon H and Liu RH: Effect of 2 alpha-hydroxyursolic acid on NF-kappaB activation induced by TNF-alpha in human breast cancer MCF-7 cells. J Agric Food Chem. 56:8412–8417. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Witjes JA, Compérat E, Cowan NC, De Santis M, Gakis G, Lebret T, Ribal MJ, Van der Heijden AG and Sherif A: European Association of Urology: EAU guidelines on muscle-invasive and metastatic bladder cancer: Summary of the 2013 guidelines. Eur Urol. 65:778–792. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Tripodi D, Quéméner S, Renaudin K, Ferron C, Malard O, Guisle-Marsollier I, Sébille-Rivain V, Verger C, Géraut C and Gratas-Rabbia-Ré C: Gene expression profiling in sinonasal adenocarcinoma. BMC Med Genomics. 2:652009. View Article : Google Scholar : PubMed/NCBI

35 

Selamat SA, Chung BS, Girard L, Zhang W, Zhang Y, Campan M, Siegmund KD, Koss MN, Hagen JA, Lam WL, et al: Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res. 22:1197–1211. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Miwa HE, Koba WR, Fine EJ, Giricz O, Kenny PA and Stanley P: Bisected, complex N-glycans and galectins in mouse mammary tumor progression and human breast cancer. Glycobiology. 23:1477–1490. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Lu Y, Liu P, Wen W, Grubbs CJ, Townsend RR, Malone JP, Lubet RA and You M: Cross-species comparison of orthologous gene expression in human bladder cancer and carcinogen-induced rodent models. Am J Transl Res. 3:8–27. 2010.PubMed/NCBI

38 

Hatakeyama S: TRIM proteins and cancer. Nat Rev Cancer. 11:792–804. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Dokmanovic M, Chang BD, Fang J and Roninson IB: Retinoid-induced growth arrest of breast carcinoma cells involves co-activation of multiple growth-inhibitory genes. Cancer Biol Ther. 1:24–27. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Sugiura T and Miyamoto K: Characterization of TRIM31, upregulated in gastric adenocarcinoma, as a novel RBCC protein. J Cell Biochem. 105:1081–1091. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Li H, Zhang Y, Zhang Y, Bai X, Peng Y and He P: TRIM31 is downregulated in non-small cell lung cancer and serves as a potential tumor suppressor. Tumour Biol. 35:5747–5752. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Nurse P: A long twentieth century of the cell cycle and beyond. Cell. 100:71–78. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Peng ZG, Yao YB, Yang J, Tang YL and Huang X: Mangiferin induces cell cycle arrest at G2/M phase through ATR-Chk1 pathway in HL-60 leukemia cells. Genet Mol Res. 14:4989–5002. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Andries V, Vandepoele K, Staes K, Berx G, Bogaert P, van Isterdael G, Ginneberge D, Parthoens E, Vandenbussche J, Gevaert K and van Roy F: NBPF1, a tumor suppressor candidate in neuroblastoma, exerts growth inhibitory effects by inducing a G1 cell cycle arrest. BMC Cancer. 15:3912015. View Article : Google Scholar : PubMed/NCBI

45 

Li N, Sun M, Wang Y, Lv Y, Hu Z, Cao W, Zheng J and Jiao X: Effect of cell cycle phase on the sensitivity of SAS cells to sonodynamic therapy using low-intensity ultrasound combined with 5-aminolevulinic acid in vitro. Mol Med Rep. 12:3177–3183. 2015.PubMed/NCBI

46 

de Stanchina E, McCurrach ME, Zindy F, Shieh SY, Ferbeyre G, Samuelson AV, Prives C, Roussel MF, Sherr CJ and Lowe SW: E1A signaling to p53 involves the p19ARFtumor suppressor. Genes Dev. 12:2434–2442. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Chakraborty A, Uechi T and Kenmochi N: Guarding the ‘translation apparatus’: Defective ribosome biogenesis and the p53 signaling pathway. Wiley Interdiscip Rev RNA. 2:507–522. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Stegh AH: Targeting the p53 signaling pathway in cancer therapy - the promises, challenges and perils. Expert Opin Ther Targets. 16:67–83. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 14 Issue 1

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
Zhang DQ, Zhou CK, Chen SZ, Yang Y and Shi BK: Identification of hub genes and pathways associated with bladder cancer based on co-expression network analysis. Oncol Lett 14: 1115-1122, 2017
APA
Zhang, D., Zhou, C., Chen, S., Yang, Y., & Shi, B. (2017). Identification of hub genes and pathways associated with bladder cancer based on co-expression network analysis. Oncology Letters, 14, 1115-1122. https://doi.org/10.3892/ol.2017.6267
MLA
Zhang, D., Zhou, C., Chen, S., Yang, Y., Shi, B."Identification of hub genes and pathways associated with bladder cancer based on co-expression network analysis". Oncology Letters 14.1 (2017): 1115-1122.
Chicago
Zhang, D., Zhou, C., Chen, S., Yang, Y., Shi, B."Identification of hub genes and pathways associated with bladder cancer based on co-expression network analysis". Oncology Letters 14, no. 1 (2017): 1115-1122. https://doi.org/10.3892/ol.2017.6267