Open Access

Identification of gene markers associated with metastasis in clear cell renal cell carcinoma

  • Authors:
    • Hailing Yang
    • Pengfei Huo
    • Guozhang Hu
    • Bo Wei
    • Dalian Kong
    • Hongjun Li
  • View Affiliations

  • Published online on: April 24, 2017     https://doi.org/10.3892/ol.2017.6084
  • Pages: 4755-4761
  • Copyright: © Yang 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 target genes for the early diagnosis and treatment of early metastatic clear cell renal cell carcinoma (ccRCC) using the microarray data of early metastatic and non‑metastatic ccRCC samples. The DNA microarray dataset GSE47352 was downloaded from Gene Expression Omnibus and included 4 early metastatic and 5 non‑metastatic ccRCC samples. Differentially expressed genes (DEGs) were screened using the limma package. Then, pheatmap package was used to conduct two‑way clustering for the DEGs. Subsequently, MAPPFinder and GenMAPP were employed separately to perform functional and pathway enrichment analysis for the DEGs. Additionally, a protein‑protein interaction (PPI) network was constructed using Cytoscape, and small drug molecules were searched using Connectivity map (cmap). In total, 196 upregulated and 163 downregulated genes were identified. DEGs, including JUN, tumor necrosis factor (TNF), Ras homolog family member B (RHOB) and transforming growth factor β2 (TGFβ2) were significantly enriched in the signaling pathway of renal cell carcinoma. Furthermore, nuclear receptor subfamily 4 group A member 1 (NR4A1) was significantly enriched in the mitogen‑activated protein kinase signaling pathway; in addition, laminin subunit α (LAMA) 1, LAMA2 and LAMA4 were significantly enriched in extracellular matrix‑receptor interaction. JUN (degree=6) had the highest degree in the PPI network. Thapsigargin (score=‑0.913) possessed the highest performance in terms of the treatment of early metastatic ccRCC. In the present study, it was discovered that certain DEGs, including JUN, TNF, RHOB, NR4A1, TGFβ2, LAMA1, LAMA2 and LAMA4 were potential target genes associated with early metastatic ccRCC. In addition, thapsigargin could be used as an efficient small drug molecule for the treatment of early metastatic ccRCC.

Introduction

Renal cell carcinoma (RCC), which is one of the most common types of cancer, accounts for almost 3% of all human malignancies (1). As the most common type of RCC, clear cell RCC (ccRCC) accounts for 70–80% of RCC cases (2). The metastasis and recurrence of ccRCC, as well as its poor prognosis, results in poor survival for patients (3).

At present, with the development of microarray technology, a large number of differentially expressed genes (DEGs) associated with ccRCC have been identified and the genes expression profiles have been uploaded to databases, including Gene Expression Omnibus (GEO) and Array Express Archive for researchers to study (4,5). Many genes and signaling pathways involved in the metastasis of ccRCC have been discovered. Downregulation of FOXO3a may promote tumor metastasis in ccRCC (6). C-X-C motif chemokine receptor 2 (CXCR2)/CXCR2 ligand biology is important in the promotion of angiogenesis and facilitation of tumor growth and metastasis in RCC cells (7). A previous study demonstrated that overexpression of brain-type fatty-acid-binding protein (FABP) may lead to the reduction of liver-type FABP in RCC, which serves a role in cell signaling, regulation of gene expression, cell growth and differentiation (8). Although the above researches have identified specific genes associated with metastasis of ccRCC, the mechanisms of ccRCC metastasis remain unclear. Furthermore, few drugs have been developed to be effective for treatment of metastatic ccRCC.

In the present study, in order to achieve an improved understanding of ccRCC, early metastatic and non-metastatic ccRCC samples were used to screen DEGs associated with metastatic ccRCC. Ni et al (6) used P<0.05 as the criterion to screen DEGs between metastatic and non-metastatic ccRCC samples; using identical data, the present study screened the DEGs by stricter cut-off criteria [false discovery rate (FDR) <0.05 and |log fold change (FC)|>1]. Subsequently, functional and pathway enrichment analysis was performed to predict the potential functions of DEGs. Furthermore, a protein-protein interaction (PPI) network was constructed to analyze the interactions between DEGs. In addition, small drug molecules associated with ccRCC were detected. It is anticipated that the results of the present study may lead to a potential breakthrough in the treatment of metastatic ccRCC.

Materials and methods

Microarray data

The microarray data GSE47352 deposited by Ni et al (6) was downloaded from the GEO (http://www.ncbi.nlm.nih.gov/geo/) of the National Center of Biotechnology Information. In addition, probes annotation information was also downloaded for mapping the probes to genes (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE47352). This dataset was generated based on the platform of GPL570 (HG-U133_Plus_2) Affymetrix Human Genome U133 Plus 2.0 Array. A total of 9 samples are enrolled in the GSE47352 dataset, including 4 early metastatic ccRCC samples (metastatic group) and 5 non-metastatic ccRCC samples (non-metastatic group). The ccRCC tissue samples were removed from ccRCC patients who underwent nephrectomy at the Chinese People's Liberation Army General Hospital between January 2009 and May 2012, and were snap-frozen in liquid nitrogen. Patients with negative abdomen and chest computed tomography or magnetic resonance imaging and without metastatic lesions were classed as non-metastatic ccRCC; patients with metastatic lesions were classed as early metastatic ccRCC (6).

Data preprocessing and DEGs screening

Based on the k-Nearest Neighbors method (9), Affymetrix (Affy) package (version 1.28.0; Affymetrix, Inc., Santa Clara, CA, USA) (10) in R language was employed to account for the missing values in the raw data from the DNA microarray. Subsequently, the data was normalized by the median normalization method (11). Compared with the non-metastatic group, the DEGs in the metastatic group were screened using the linear model for microarray data (Limma) package (12). The Benjamini-Hochberg method (13) was applied to conduct multiple testing adjustment to identify the FDR and the logFC was also calculated. Genes with FDR<0.05 and |logFC|>1 were taken as the DEGs between the early metastatic and non-metastatic groups.

Comparison of gene expression between the metastatic and non-metastatic groups

Generally, significant differences in gene expression are observed in tissues under different disease states (14). The gene expression values of DEGs were extracted, and the pheatmap package (15) in R was used to perform two-way clustering (16) based on Euclidean distance (17).

Functional and pathway enrichment analysis

Gene map annotator and pathway profiler (GenMAPP; version 2.1; http://www.GenMAPP.org) was used for visualizing, analyzing and demonstrating the microarray data in pathways (18). The MAPPFinder was used for coupling the annotations of the Gene Ontology (GO) database with GenMAPP and calculated the GO-values (19). In the present study, MAPPFinder and GenMAPP were employed separately to conduct functional and pathway enrichment analysis for the DEGs. P<0.05 was taken as the threshold.

PPI network construction

The Search Tool for the Retrieval of Interacting Genes (STRING) database (http://string-db.org/) provided comprehensive predicted PPI information (20). The PPI pairs (combined score >0.6) were screened from the STRING database, and the PPI network was subsequently visualized using Cytoscape software (The Cytoscape Consortium, San Diego, CA, USA; version 2.8; http://www.cytoscape.org) (21).

Screening of small drug molecules

The Connectivity Map (cmap; http://www.broadinstitute.org/CMAP/) database may be used to investigate connections among small drug molecules, genes and diseases (22,23). A higher negative score indicates a higher correlation between the small drug molecules and the DEGs. The DEGs were imported into cmap to screen the small drug molecules associated with DEGs. The small drug molecules with |score|>0.8 were recorded.

Results

DEGs screening

According to the microarray data analysis between early metastatic ccRCC and non-metastatic ccRCC samples by Limma, a total of 359 DEGs were obtained in metastatic group, including 196 upregulated genes and 163 downregulated genes. The top ten significantly upregulated (including vomeronasal 1 receptor 2 and homeobox A1) and downregulated [including epiregulin and RAR related orphan receptor A (RORA)] genes are listed in Table I.

Table I.

Top ten up- and downregulated genes.

Table I.

Top ten up- and downregulated genes.

Downregulated genesUpregulated genes


Gene symbolFDRLogFCGene symbolFDRLogFC
EREG0.0106464−4.63294VN1R20.00603994.758334
CCDC1580.0009937−4.13289TSPAN30.00782844.667987
HMGCLL10.0024176−4.12305KCTD40.01005704.158775
TRAF3IP2-AS10.0000508−4.10811CECR90.00898143.969089
RORA0.0056259−4.09241PCDH200.00341703.889183
TMEM51-AS10.0086847−3.87088FAM95A0.00821713.883404
LOC6454850.0063110−3.86383SEZ6L0.00781823.846624
UNC93A0.0024362−3.85761CYLC10.00101103.833110
RGPD10.0034170−3.78002SLC22A250.00698443.821152
FAHD2CP0.0009937−3.75996HOXA10.00545533.815357

[i] FDR, false discovery rate; FC, fold change.

Comparison of gene expression between metastatic and non-metastatic samples

Hierarchical cluster analysis of the expression values of DEGs revealed that the early metastatic ccRCC samples and the non-metastatic ccRCC samples were in significantly separated clusters (Fig. 1).

Functional and pathway enrichment analysis

A total of five Kyoto Encyclopedia Genes and Genomes pathways were obtained for the identified DEGs (Table II). The most significantly enriched pathway was the renal cell carcinoma pathway (P=0.003503), which involved 6 DEGs [endothelial PAS domain-containing protein 1 (EPAS1), ETS1, JUN, SOS2, TGFβ2, and protein tyrosine phosphatase, non-receptor type 11 (PTPN11)]. Furthermore, these 6 DEGs were all downregulated. In addition, 11 DEGs [including TGFβ2, nuclear receptor subfamily 4 group A member 1 (NR4A1) and dual specificity protein phosphatase 1 (DUSP1)] significantly participated in the mitogen-activate protein kinase (MAPK) signaling pathway (P=0.005407) and 5 DEGs [including laminin subunit α (LAMA) 2, LAMA1 and LAMA4] were enriched in the extracellular matrix (ECM)-receptor interaction pathway (P=0.034718).

Table II.

Enriched pathways for differentially expressed genes between early metastasis ccRCC and the non-metastasis ccRCC samples.

Table II.

Enriched pathways for differentially expressed genes between early metastasis ccRCC and the non-metastasis ccRCC samples.

Genes

IDPathwaysP-valueCountUpregulatedDownregulated
hsa05211Renal cell carcinoma0.003503  6EPAS1, ETS1, JUN, TGFβ2, SOS2, PTPN11
hsa04010MAPK signaling pathway0.00540711CACNA2D1, TNF, PTPN5, MAPK8IP3, CACNG2FGF8, DUSP1, JUN, SOS2, NR4A1, TGFβ2
hsa05410Hypertrophic cardiomyopathy0.008004  6CACNA2D1, TNF, SGCD, CACNG2TGFβ2 LAMA2
hsa05414Dilated cardiomyopathy0.011083  6CACNA2D1, TNF, SGCD, CACNG2TGFβ2, LAMA2,
hsa04512ECM-receptor interaction0.034718  5SV2BLAMA2, LAMA1, LAMA4, CD36

[i] ccRCC, clear cell renal cell carcinoma; MAPK, mitogen activated protein kinase; ECM, extracellular matrix.

The top 10 GO terms are listed in Table III, including regulation of transcription from RNA polymerase II promoter (P=7.26×10−6), positive regulation of the nucleic acid metabolic process (P=3.53×10−5) and positive regulation of the nitrogen compound metabolic process (P=5.82×10−5). In particular, JUN, EST1, RORA and TGFβ2 were significantly enriched in the majority of the GO terms.

Table III.

Top ten enriched functions for the differentially expressed genes.

Table III.

Top ten enriched functions for the differentially expressed genes.

IDGene ontology termCountP-valueGenes
GO:0006357Regulation of transcription from RNA polymerase II promoter29 7.26×10−6TNF, FOXK1, ONECUT2, NR6A1, FOXK2, TP63, RORA, MED20, WT1, HOXA1, NPAS1, SQSTM1, MED26, HSF4, RARB, DGKQ, KLF9, EPAS1, NR4A1, TEAD2, NR0B1, IL22, SP2, ETS1, JUN, MNX1, TFAP2E, KLF4, NFIB
GO:0045935Positive regulation of nucleic acid metabolic process25 3.53×10−5TNF, FOXK1, ONECUT2, TP63, ABCA1, RORA, WT1, HOXA1, SQSTM1, H2AFX, RARB, HSF4, EPAS1, TAF8, ESRRG, NR4A1, TEAD2, IL22, EREG, IRF6, ETS1, JUN, TFAP2E, KLF4, NFIB
GO:0045893Positive regulation of transcription21 5.04×10−5TNF, EPAS1, FOXK1, TAF8, ONECUT2, ESRRG, TP63, NR4A1, TEAD2, RORA, IL22, WT1, HOXA1, SQSTM1, ETS1, JUN, RARB, HSF4, TFAP2E, KLF4, NFIB
GO:0051254Positive regulation of RNA metabolic process21 5.72×10−5TNF, EPAS1, FOXK1, TAF8, ONECUT2, ESRRG, TP63, NR4A1, TEAD2, RORA, IL22, WT1, HOXA1, SQSTM1, ETS1, JUN, RARB, HSF4, TFAP2E, KLF4, NFIB
GO:0051173Positive regulation of nitrogen compound metabolic process25 5.82×10−5TNF, FOXK1, ONECUT2, TP63, ABCA1, RORA, WT1, HOXA1, SQSTM1, H2AFX, RARB, HSF4, EPAS1, TAF8, ESRRG, NR4A1, TEAD2, IL22, EREG, IRF6, ETS1, JUN, TFAP2E, KLF4, NFIB
GO:0045944Positive regulation of transcription from RNA polymerase II promoter18 6.37×10−5TNF, EPAS1, ONECUT2, TP63, NR4A1, TEAD2, RORA, IL22, WT1, HOXA1, SQSTM1, ETS1, JUN, RARB, HSF4, TFAP2E, KLF4, NFIB
GO:0009891Positive regulation of biosynthetic process26 7.16×10−5TNF, FOXK1, ONECUT2, TP63, APOC2, ABCA1, RORA, WT1, TGFβ2, HOXA1, SQSTM1, RARB, HSF4, EPAS1, TAF8, ESRRG, NR4A1, TEAD2, IL22, EREG, IRF6, ETS1, JUN, TFAP2E, KLF4, NFIB
GO:0031328Positive regulation of cellular biosynthetic process25 1.51s10−4TNF, FOXK1, ONECUT2, TP63, APOC2, ABCA1, RORA, WT1, HOXA1, SQSTM1, RARB, HSF4, EPAS1, TAF8, ESRRG, NR4A1, TEAD2, IL22, EREG, IRF6, ETS1, JUN, TFAP2E, KLF4, NFIB
GO:0010557Positive regulation of macromolecule biosynthetic process24 1.97×10−4TNF, EPAS1, FOXK1, TAF8, ONECUT2, ESRRG, TP63, NR4A1, TEAD2, RORA, IL22, WT1, TGFβ2, HOXA1, EREG, IRF6, SQSTM1, ETS1, JUN, RARB, HSF4, TFAP2E, KLF4, NFIB
GO:0010628Positive regulation of gene expression22 2.62×10−4TNF, EPAS1, FOXK1, TAF8, ONECUT2, ESRRG, TP63, NR4A1, TEAD2, RORA, IL22, WT1, HOXA1, IRF6, SQSTM1, ETS1, JUN, RARB, HSF4, TFAP2E, KLF4, NFIB
PPI network construction

In total, 87 PPI pairs were obtained from the STRING database. Subsequently, the six DEGs (EPAS1, ETS1, JUN, SOS2, TGFβ2 and PTPN11) that were enriched in the renal cell carcinoma pathway were mapped to the network. The network was visualized using Cytoscape (Fig. 2). In the PPI network, JUN possessed the highest degree of 6; additionally, ferritin light chain 1, NR4A1 and Ras homolog family member B (RHOB) demonstrated degrees of 5, 5 and 4, respectively. Furthermore, JUN could interact with ever shorter telomeres protein 1 (EST1), RHOB, DUSP1, tumor necrosis factor (TNF), MYC associated zinc finger protein (MAZ) and cyclin A1 (CCNA1). In addition, NR4A1 demonstrated an interaction with DUSP1.

Small drug molecule screening

For the screening of small molecular drugs, 7 small drug molecules with the |score|>0.8, including 4 negatively-correlated drugs (thapsigargin, score=−0.913; W-13, score=−0.885; trihexyphenidyl, score=−0.839; and lovastatin, score=−0.824) and 3 positively-correlated drugs (dioxybenzone, score=0.825; oxybuprocaine, score=0.853; and (−)-MK-801, score=0.887) were identified to be correlated with the DEGs (Table IV).

Table IV.

Small molecule drugs (|score|>0.8) associated with the differentially expressed genes between the primary metastatic ccRCC and the non-metastatic ccRCC samples.

Table IV.

Small molecule drugs (|score|>0.8) associated with the differentially expressed genes between the primary metastatic ccRCC and the non-metastatic ccRCC samples.

Connectivity map nameScoreP-value
Thapsigargin−0.9130.00112
W-13−0.8850.02630
Trihexyphenidyl−0.8390.00839
Lovastatin−0.8240.00181
Dioxybenzone0.8250.00149
Oxybuprocaine0.8530.00070
(−)-MK-8010.8870.00016

[i] ccRCC, clear cell renal cell carcinoma.

Discussion

In the present study, with the investigation of the gene expression profile between the early metastatic and non-metastatic ccRCC using bioinformatics methods, a total of 359 DEGs were obtained, including 196 upregulated DEGs and 163 downregulated DEGs. Hierarchical cluster analysis indicated that the metastatic ccRCC samples could be well distinguished from the non-metastatic ccRCC samples according to the identified DEGs. Furthermore, pathway enrichment analysis revealed that JUN was significantly enriched in renal cell carcinoma and the MAPK signaling pathway. Previous research has proven that MAPK serves a key role in tumor metastasis via regulating cell migration and apoptosis (24). Furthermore, in the PPI network, JUN was a hub node with the highest degree of 6 and could interact with RHOB, MAZ, DUSP1, CCNA1, TNF and EST1. JUN is identified as oncogene, which accelerates tumor cell metastasis (25). Zhang et al (26) demonstrated that JUN has a close association with metastasis of cancer cells, and overexpression of JUN may result in metastasis of breast cancer. A previous study demonstrated that epithelial-mesenchymal transition (EMT) is a key regulator of metastasis in cancer by conferring an invasive phenotype via the loss of cell-cell adhesions, cell-substrates and transition to a cell type that is capable of invading the ECM (27). Furthermore, EMT has been identified as a model by which the ccRCC occurs (28). In the present study, TNF and RHOB were significantly enriched in the pathway of early metastatic ccRCC. Previous studies have illustrated that TNF (or TNF-α) is able to elevate the migration and invasion of ccRCC cells together with downregulation of E-cadherin expression and promotion of EMT, suggesting that TNF has a close association with early metastatic ccRCC (29). RHOB is known as a tumor suppressor and is able to affect cell adhesion and migration by regulating surface integrin levels (30). Furthermore, it has also been observed that RHOB serves a distinct function in EMT by regulating cell-cell and cell- substrate contact in renal proximal tubular cells, suggesting that RHOB has a key role in early metastatic ccRCC (31). This appears to indicate that JUN, along with the interaction with TNF and RHOB, may participate in mediation of early metastatic ccRCC via regulation of cell migration and apoptosis.

In addition, NR4A1 and TGFβ2 were significantly enriched in the MAPK signaling pathway. Previous research has demonstrated that the MAPK signaling pathway is involved in inhibition of tumorigenesis, metastasis and angiogenesis in RCC via the disruption of tumor vasculature (32). NR4A1, which belongs to the Nur nuclear receptor family, has been implicated in cell cycle regulation, inflammation and apoptosis (33). It has also been reported that NR4A1 is able to promote the invasion and metastasis of breast cancer by activating TGFβ signaling (34). Furthermore, the loss of NR4A1 may enhance macrophage-mediated kidney injury and diseases due to a large increase in immune cell infiltration (predominantly macrophages, and to a lesser extent T cells and B cells) (35). Therefore, the authors hypothesized that NR4A1 had a close association with early metastatic ccRCC. TGFβ2, which belongs to the TGFβ family, is known to promote the invasion of tumor cells and allow metastasis to distant organs via induction of EMT, suppression of immune surveillance, promotion of angiogenesis and recruitment of inflammatory cells in human cancer cell lines and mouse tumor models (36,37). Consequently, the present study speculated that NR4A1 and TGFβ2 may serve a key role in the regulation of early metastatic ccRCC through the MAPK signaling pathway.

LAMA1, LAMA2 and LAMA4, which belong to the laminins family, were significantly enriched in the pathway of ECM-receptor interaction. Laminins, a family of ECM glycoproteins, are the major non-collagenous constituent of basement membranes (38). Laminins act as ECM fibers in lymph nodes, within which tumor cell metastasis occurs (39). A previous study has also reported that LAMA4 has a de-adhesive function and may serve a key role in detachment, migration and invasion of renal carcinoma cells in vivo (40). Therefore, LAMA1, LAMA2 and LAMA4 may be potential target genes in the treatment of early metastatic ccRCC.

Thapsigargin was discovered to have high efficiency for the treatment of early metastasis in ccRCC. Thapsigargin is a non-competitive inhibitor of sarco/endoplasmic reticulum Ca2+ ATPase (41). Thapsigargin is able to couple to a peptide carrier, producing a soluble non-toxic pro-drug, which induces apoptosis of prostate cancer cells (42). Research into the use of thapsigargin as a small drug molecule for cancer treatment has increased, including for the treatment of lung adenocarcinoma and prostate cancer (42,43). Due to its positive effects on prostate cancer and lung adenocarcinoma, the present authors speculated that it may be also effective for the treatment of early metastatic ccRCC.

In conclusion, a total of 359 DEGs were identified in the metastatic group compared with the non-metastatic group. Furthermore, the DEGs, including TGFβ2, JUN, NR4A1, RHOB, LAMA1, LAMA2 and LAMA4, were involved in early metastatic ccRCC. In addition, the present study screened a small drug molecule named thapsigargin, which may have high efficiency for the treatment of the early metastatic ccRCC. However, further studies to investigate the viability of the above assumptions are required and additional experimental research is needed to validate the results of the present study, as well as confirm thapsigargin's safety and efficacy for the treatment of early metastatic ccRCC.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Rosner I, Bratslavsky G, Pinto PA and Linehan WM: The clinical implications of the genetics of renal cell carcinoma. Urol Oncol. 27:131–136. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Novara G, Ficarra V, Antonelli A, Artibani W, Bertini R, Carini M, Cunico S Cosciani, Imbimbo C, Longo N, et al: Validation of the 2009 TNM version in a large multi-institutional cohort of patients treated for renal cell carcinoma: Are further improvements needed? Eur Urol. 58:588–595. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Barrett T, Troup DB, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, et al: NCBI GEO: Archive for functional genomics data sets-10 years on. Nucleic Acids Res. 39(Database Issue): D1005–D1010. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Parkinson H, Kapushesky M, Shojatalab M, Abeygunawardena N, Coulson R, Farne A, Holloway E, Kolesnykov N, Lilja P, Lukk M, et al: ArrayExpress-a public database of microarray experiments and gene expression profiles. Nucleic Acids Res. 35(Database issue): D747–D750. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Ni D, Ma X, Li HZ, Gao Y, Li XT, Zhang Y, Ai Q, Zhang P, Song EL, Huang QB, et al: Downregulation of FOXO3a promotes tumor metastasis and is associated with metastasis-free survival of patients with clear cell renal cell carcinoma. Clin Cancer Res. 20:1779–1790. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Mestas J, Burdick MD, Reckamp K, Pantuck A, Figlin RA and Strieter RM: The role of CXCR2/CXCR2 ligand biological axis in renal cell carcinoma. J Immunol. 175:5351–5357. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Tölle A, Jung M, Lein M, Johannsen M, Miller K, Moch H, Jung K and Kristiansen G: Brain-type and liver-type fatty acid-binding proteins: New tumor markers for renal cancer? BMC Cancer. 9:2482009. View Article : Google Scholar : PubMed/NCBI

9 

Zhang ML and Zhou ZH: A k-nearest neighbor based algorithm for multi-label classification. Journal. 2:718–721. 2005.

10 

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

11 

Rao Y, Lee Y, Jarjoura D, Ruppert AS, Liu CG, Hsu JC and Hagan JP: A comparison of normalization techniques for microRNA microarray data. Stat Appl Genet Mol Biol. 7:: 2008.PubMed/NCBI

12 

Gentleman R, Carey VJ, Huber W, Irizarry RA and Dudoit S: Bioinformatics and computational biology solutions using R and Bioconductor. Springer; 2005, View Article : Google Scholar

13 

Genovese C and Wasserman L: Operating characteristics and extensions of the false discovery rate procedure. J Royal Statistical Soci Series B (Statistical Methodology). 64:499–517. 2002. View Article : Google Scholar

14 

Ester M, Kriegel HP, Sander J and Xu X: A density-based algorithm for discovering clusters in large spatial databases with noise. Journal. 96:226–231. 1996.

15 

Kolde R: Pheatmap: Pretty Heatmaps. R Package Version 0.6.1. Journal. 2012.

16 

Szekely GJ and Rizzo ML: Hierarchical clustering via joint between-within distances: Extending Ward's minimum variance method. J Classification. 22:151–183. 2005. View Article : Google Scholar

17 

Deza MM and Deza E: Encyclopedia of Distances. Springer; 2009, View Article : Google Scholar

18 

Dahlquist KD, Salomonis N, Vranizan K, Lawlor SC and Conklin BR: GenMAPP, a new tool for viewing and analyzing microarray data on biological pathways. Nat Genet. 31:19–20. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Doniger SW, Salomonis N, Dahlquist KD, Vranizan K, Lawlor SC and Conklin BR: MAPPFinder: Using gene ontology and GenMAPP to create a global gene-expression profile from microarray data. Genome Biol. 4:R72003. View Article : Google Scholar : PubMed/NCBI

20 

Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, et al: The STRING database in 2011: Functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 39(Database issue): D561–D568. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: New features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Lamb J: The Connectivity Map: A new tool for biomedical research. Nat Rev Cancer. 7:54–60. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet JP, Subramanian A, Ross KN, et al: The connectivity map: Using gene-expression signatures to connect small molecules, genes and disease. Science. 313:1929–1935. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Reddy KB, Nabha SM and Atanaskova N: Role of MAP kinase in tumor progression and invasion. Cancer Metastasis Rev. 22:395–403. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Chaturvedi MM, Sung B, Yadav VR, Kannappan R and Aggarwal BB: NF-κB addiction and its role in cancer: ‘One size does not fit all’. Oncogene. 30:1615–1630. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Y, Pu X, Shi M, Chen L, Song Y, Qian L, Yuan G, Zhang H, Yu M, Hu M, et al: Critical role of c-Jun overexpression in liver metastasis of human breast cancer xenograft model. BMC Cancer. 7:1452007. View Article : Google Scholar : PubMed/NCBI

27 

Davis FM, Stewart TA, Thompson EW and Monteith GR: Targeting EMT in cancer: Opportunities for pharmacological intervention. Trends Pharmacol Sci. 35:479–488. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Tun HW, Marlow LA, Von Roemeling CA, Cooper SJ, Kreinest P, Wu K, Luxon BA, Sinha M, Anastasiadis PZ and Copland JA: Pathway signature and cellular differentiation in clear cell renal cell carcinoma. PLoS One. 5:e106962010. View Article : Google Scholar : PubMed/NCBI

29 

Mikami S, Mizuno R, Kosaka T, Saya H, Oya M and Okada Y: Expression of TNF-α and CD44 is implicated in poor prognosis, cancer cell invasion, metastasis and resistance to the sunitinib treatment in clear cell renal cell carcinomas. Int J Cancer. 136:1504–1514. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Wheeler AP and Ridley AJ: RhoB affects macrophage adhesion, integrin expression and migration. Exp Cell Res. 313:3505–3516. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Hutchison N, Hendry BM and Sharpe CC: Rho isoforms have distinct and specific functions in the process of epithelial to mesenchymal transition in renal proximal tubular cells. Cell Signal. 21:1522–1531. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Huang D, Ding Y, Luo WM, Bender S, Qian CN, Kort E, Zhang ZF, VandenBeldt K, Duesbery NS, Resau JH and Teh BT: Inhibition of MAPK kinase signaling pathways suppressed renal cell carcinoma growth and angiogenesis in vivo. Cancer Res. 68:81–88. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Pei L, Castrillo A and Tontonoz P: Regulation of macrophage inflammatory gene expression by the orphan nuclear receptor Nur77. Mol Endocrinol. 20:786–794. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Zhou F, Drabsch Y, Dekker TJ, de Vinuesa AG, Li Y, Hawinkels LJ, Sheppard KA, Goumans MJ, Luwor RB, de Vries CJ, et al: Nuclear receptor NR4A1 promotes breast cancer invasion and metastasis by activating TGF-β signalling. Nat Commun. 5:33882014. View Article : Google Scholar : PubMed/NCBI

35 

Westbrook L, Johnson AC, Regner KR, Williams JM, Mattson DL, Kyle PB, Henegar JR and Garrett MR: Genetic susceptibility and loss of Nr4a1 enhances macrophage-mediated renal injury in CKD. J Am Soc Nephrol. 25:2499–2510. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Janda E, Lehmann K, Killisch I, Jechlinger M, Herzig M, Downward J, Beug H and Grünert S: Ras and TGF[beta] cooperatively regulate epithelial cell plasticity and metastasis Dissection of Ras signaling pathways. J Cell Biol. 156:299–313. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Heldin C-H, Vanlandewijck M and Moustakas A: Regulation of EMT by TGFβ in cancer. FEBS Lett. 586:1959–1970. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Hohenester E and Yurchenco PD: Laminins in basement membrane assembly. Cell Adh Migr. 7:56–63. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Patarroyo M, Tryggvason K and Virtanen I: Laminin isoforms in tumor invasion, angiogenesis and metastasis. Semin Cancer Biol. 12:197–207. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Vainionpää N, Lehto VP, Tryggvason K and Virtanen I: Alpha4 chain laminins are widely expressed in renal cell carcinomas and have a de-adhesive function. Lab Invest. 87:780–791. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Rogers TB, Inesi G, Wade R and Lederer W: Use of thapsigargin to study Ca2+ homeostasis in cardiac cells. Biosci Rep. 15:341–349. 1995. View Article : Google Scholar : PubMed/NCBI

42 

Denmeade SR, Jakobsen CM, Janssen S, Khan SR, Garrett ES, Lilja H, Christensen SB and Isaacs JT: Prostate-specific antigen-activated thapsigargin prodrug as targeted therapy for prostate cancer. J Natl Cancer Inst. 95:990–1000. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Doan NT, Paulsen ES, Sehgal P, Møller JV, Nissen P, Denmeade SR, Isaacs JT, Dionne CA and Christensen SB: Targeting thapsigargin towards tumors. Steroids. 2014.PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 13 Issue 6

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
Yang H, Huo P, Hu G, Wei B, Kong D and Li H: Identification of gene markers associated with metastasis in clear cell renal cell carcinoma. Oncol Lett 13: 4755-4761, 2017
APA
Yang, H., Huo, P., Hu, G., Wei, B., Kong, D., & Li, H. (2017). Identification of gene markers associated with metastasis in clear cell renal cell carcinoma. Oncology Letters, 13, 4755-4761. https://doi.org/10.3892/ol.2017.6084
MLA
Yang, H., Huo, P., Hu, G., Wei, B., Kong, D., Li, H."Identification of gene markers associated with metastasis in clear cell renal cell carcinoma". Oncology Letters 13.6 (2017): 4755-4761.
Chicago
Yang, H., Huo, P., Hu, G., Wei, B., Kong, D., Li, H."Identification of gene markers associated with metastasis in clear cell renal cell carcinoma". Oncology Letters 13, no. 6 (2017): 4755-4761. https://doi.org/10.3892/ol.2017.6084