Open Access

Bioinformatics analyses of significant genes, related pathways and candidate prognostic biomarkers in glioblastoma

  • Authors:
    • Lingqi Zhou
    • Hai Tang
    • Fang Wang
    • Lizhi Chen
    • Shanshan Ou
    • Tong Wu
    • Jie Xu
    • Kaihua Guo
  • View Affiliations

  • Published online on: August 21, 2018     https://doi.org/10.3892/mmr.2018.9411
  • Pages: 4185-4196
  • Copyright: © Zhou 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

Glioblastoma (GBM) is the most common type of malignant tumor of the central nervous system. The prognosis of patients with GBM is very poor, with a survival time of ~15 months. GBM is highly heterogeneous and highly aggressive. Surgical removal of intracranial tumors does provide a good advantage for patients as there is a high rate of recurrence. The understanding of this type of cancer needs to be strengthened, and the aim of the present study was to identify gene signatures present in GBM and uncover their potential mechanisms. The gene expression profiles of GSE15824 and GSE51062 were downloaded from the Gene Expression Omnibus database. Normalization of the data from primary GBM samples and normal samples in the two databases was conducted using R software. Then, joint analysis of the data was performed. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed, and the protein‑protein interaction (PPI) network of the differentially expressed genes (DEGs) was constructed using Cytoscape software. Identification of prognostic biomarkers was conducted using UALCAN. In total, 9,341 DEGs were identified in the GBM samples, including 9,175 upregulated genes and 166 downregulated genes. The top 1,000 upregulated DEGs and all of the downregulated DEGs were selected for GO, KEGG and prognostic biomarker analyses. The GO results showed that the upregulated DEGs were significantly enriched in biological processes (BP), including immune response, cell division and cell proliferation, and the downregulated DEGs were also significantly enriched in BP, including cell growth, intracellular signal transduction and signal transduction by protein phosphorylation. KEGG pathway analysis showed that the upregulated DEGs were enriched in circadian entrainment, cytokine‑cytokine receptor interaction and maturity onset diabetes of the young, while the downregulated DEGs were enriched in the TGF‑β signaling pathway, MAPK signaling pathway and pathways in cancer. All of the downregulated genes and the top 1,000 upregulated genes were selected to establish the PPI network, and the sub‑networks revealed that these genes were involved in significant pathways, including olfactory transduction, neuroactive ligand‑receptor interaction and viral carcinogenesis. In total, seven genes were identified as good prognostic biomarkers. In conclusion, the identified DEGs and hub genes contribute to the understanding of the molecular mechanisms underlying the development of GBM and they may be used as diagnostic and prognostic biomarkers and molecular targets for the treatment of patients with GBM in the future.

Introduction

Glioma is the most common type of tumor in the central nervous system. According to the literature (1,2), the incidence of gliomas is ~12 per 100,000 people and the proportion of intracranial tumors that are gliomas is ~35–60%, which is a serious harm to human health. As stated in the pathological classification of the WHO 2007) (3), the degree of differentiation of tumor cells can be divided into four levels, with the higher the grade of the tumor, the worse the prognosis of the patient with GBM. Level IV of the WHO classification has a very poor prognosis. According to statistics, each year ~77,000 new cases of patients with glioblastoma (GBM) are diagnosed in the United States and Europe, but the follow-up 5-year survival rate is <3% (4). Compared with other pathological types of glioma, the survival of patients with GBM is also calculated weekly and the majority of patients with GBM die due to tumor recurrence. It is therefore imperative to find a new solution in order to treat patients with GBM.

Due to the popularization and development of supercomputer technology, high-throughput platforms for the analysis of gene expression are growing. Microarray analysis and next-generation sequencing are increasingly valued as essential tools in the field of medical oncology, with great clinical applications such as molecular classification of cancers, molecular diagnosis, prognosis prediction, patient stratification, tumor response prediction and new drug targets discovery (57). A number of tumor-related databases have been established, such as Gene Expression Omnibus (GEO) (8) and TCGA (9). Further, the uses of bioinformatics analysis are changing constantly. The 2016 edition of the WHO pathological grading criteria was the first to include guidelines for the molecular markers of glioma, indicating that genetic molecular diagnosis and gene analysis of the disease play an increasingly important role (10).

In the present study, original data (GSE15824 and GSE51062) were downloaded from the GEO database (http://www.ncbi.nlm.nih.gov/geo/). A combined analysis was conducted using limma package (11) in R software (version 3.3.2). In total, 74 GBM samples and 5 normal samples were selected to identify differentially expressed genes (DEGs). Subsequently, the DEGs were screened using Morpheus online software (https://software.broadinstitute.org/morpheus/), followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. By analyzing the biological functions and pathways of the DEGs, the present study provides insight into the development of GBM at the molecular level and identifies candidate biomarkers for the future treatment, diagnosis and prognosis of patients with GBM.

Materials and methods

Microarray data

GSE15824 and GSE51062 were downloaded from the GEO database. Both datasets were based on the Affymetrix GPL570 platform (Affymetrix Human Genome U133 Plus 2.0 Array). In total, 5 normal samples and 22 primary GBM samples from GSE15824 and 52 primary GBM samples from GSE51062 were selected.

Data quality control

Affy package (12) R software was used to perform quality control of all of the microarray CEL files and RNA degradation plots were drawn at the same time. Good uniformity of the curve due to the quality of the RNA extraction with a small level of error laid the foundation for the merging operation of the two datasets.

Identification of DEGs

Analysis of gene expression was conducted using limma package (11) in R software. The Affy package (12) Robust Multichip Average (13) algorithm was used to identify DEGs with a fold-change ≥2 and a P-value cutoff of <0.05 was defined as statistically significant. A heatmap of the DEGs was produced using Morpheus online software and it showed the top 10 upregulated and downregulated genes, respectively. All significant DEGs were shown in a volcano plot generated using R software.

GO and pathway enrichment analyses of DEGs

GO analysis is a common and useful method for annotating genes and gene products and for identifying characteristic biological attributes of high-throughput genome or transcriptome data (14,15). KEGG (http://www.genome.jp/) is a well-known database for systematic analysis of gene functions in biological pathways, which links genomic information with higher-order functional information (16). The DEGs were mapped to the relevant biological annotation using the DAVID online database (https://david.ncifcrf.gov/) (17). DAVID is an essential online tool for high-throughput gene functional analysis, which provides the functionality to perform simultaneous GO and KEGG analysis. P<0.05 was considered to indicate a statistically significant difference.

Integration of protein-protein interaction (PPI) network and module analysis

Search Tool for the Retrieval of Interacting Genes (18) (STRING, version 10.0, http://string-db.org/) was used to conduct PPI network and module analysis. STRING (version 10.0) includes 9.6 million proteins from 2,031 organisms. These proteins have 184 million interactions. The top 1,000 upregulated DEGs and all of the downregulated DEGs were uploaded to the search tool. Only experimentally-validated interactions with a combined score >0.4 were selected as significant. The whole giant network was then inputted into Cytoscape (19) software (version 3.4) to evaluate primary modules of the PPI sub-network using the plug-in Molecular Complex Detection (MCODE). The criteria were set as default. GO analysis and KEGG pathway enrichment analyses were performed for DEGs in the modules using DAVID. P<0.05 was considered to indicate a statistically significant difference.

Identification of prognostic biomarker

UALCAN (20) (http://ualcan.path.uab.edu) was used to identify prognostic biomarkers in the top 100 regulated DEGs. UALCAN was used to construct an algorithm based on the TCGA level 3 RNA-seq database. All data were divided into 31 types according to the type of cancer. The top 100 upregulated and downregulated DEGs, respectively, were uploaded to the UALCAN GBM database. P<0.05 was considered to indicate a statistically significant difference. The influence of genes and sex as an influencing factor on the prognosis of disease were retrieved. Genes with insufficient data were automatically eliminated.

Results

Microarray chip quality control

Affy package in R software was used to conduct quality control of all of the microarray CEL files. The RNA degradation plot shows that the two datasets exhibit a good consistency in the slope of the line. All of the samples have a lower numerical value in the 5′ end, which is shown in Fig. 1.

Identification of DEGs

A total 74 primary GBM samples and five normal samples were analyzed. By using R software and the Morpheus online tool, 9,341 DEGs were identified in the GBM samples, which included 9,175 upregulated genes and 166 downregulated genes. A heatmap of the top 10 upregulated and downregulated DEGs, respectively, was produced using the Morpheus online tool. A volcano plot of all of the DEGs was generated using ggplot2 package and R software. The heatmap and volcano plot are shown in Figs. 2 and 3, respectively.

GO term enrichment analysis

The online software DAVID was used to identify overrepresented GO categories and KEGG pathways. The top 1,000 upregulated DEGs and all of the downregulated DEGs were uploaded to the gene list separately. GO analysis showed that the upregulated DEGs were significantly enriched in biological processes (BP), including defense response, nervous system development and cell development. The downregulated DEGs were also significantly enriched in BP, including regulation of signaling, positive regulation of transport and regulation of cell communication. As for molecular function, the upregulated DEGs were enriched in molecular transducer activity, receptor activity and transmembrane receptor activity and the downregulated DEGs were enriched in cytokine receptor binding, metal ion binding and growth factor activity. In addition, with regards to cell components, the upregulated DEGs were enriched in intrinsic component of plasma membrane, integral component of plasma membrane and extracellular space, and the downregulated DEGs were enriched in regulation of signaling, positive regulation of transport and regulation of cell communication. The results are shown in full in Fig. 4.

KEGG pathway analysis

The enrichment of the top 1,000 upregulated DEGs and all of the downregulated DEGs was identified using KEGG analysis separately. The top 1,000 upregulated DEGs were enriched in circadian entrainment, cytokine-cytokine receptor interaction and maturity onset diabetes of the young, while all of the downregulated DEGs were enriched in TGF-β signaling pathway, MAPK signaling pathway and pathways in cancer. The results are shown in full in Table I.

Table I.

KEGG pathway analysis of DEGs associated with GBM.

Table I.

KEGG pathway analysis of DEGs associated with GBM.

A, Upregulated
Pathway IDNameGene count%P-valueGenes
hsa04713Circadian entrainment181.204.60E-04ADCY1, ADCYAP1R1, GRIA3, KCNJ3, KCNJ5, RPS6KA5, GNGT1, FOS, GRIA2, RYR3, RYR1, RYR2, GUCY1A3, GUCY1B3, ADCY10, RASD1, GNG7, MTNR1A
hsa04060Cytokine-cytokine receptor interaction312.070.0013OSMR, BMPR2, TNFSF15, CXCL11, PF4V1, IFNA2, IFNA1, IL23A, IFNA6, CXCR4, IL10RB, TNFRSF19, IFNA8, IFNK, IL13RA1, THPO, IL3, IL18RAP, IL5, FLT4, TGFBR1, IL9, IL26, TNFSF8, CCR8, TNFSF11, PRLR, PPBP, PDGFRB, IL12B, IL22RA2
hsa04950Maturity onset diabetes of the young80.540.0021ONECUT1, FOXA3, IAPP, SLC2A2, PAX4, HNF4G, NR5A2, NKX2-2
hsa04080Neuroactive ligand-receptor interaction342.280.0035OPRM1, GLRA1, DRD3, TACR1, ADCYAP1R1, GABBR1, GNRHR, SCTR, AGTR2, S1PR1, GRID2, TAAR1, TAAR2, GABRD, GABRA4, RXFP3, RXFP2, GRIA3, NPY1R, NTSR2, FSHR, P2RX7, P2RY10, ADRB2, GABRR1, PRLR, CHRM3, GRIA2, P2RX3, CHRNB4, ADRA1B, ADRA1A, GHSR, MTNR1A
hsa04630Jak-STAT signaling pathway191.270.0185PIK3CG, IL3, IL5, OSMR, IL9, IFNA2, IFNA1, IL23A, PRLR, IFNA6, IL10RB, IFNA8, IL12B, IFNK, IL13RA1, MYC, THPO, IL22RA2, IL13RA2

B, Downregulated

Pathway IDNameGene count%P-valueGenes

hsa04350TGF-β signaling pathway42.700.0181BMP4, INHBA, NOG, GDF5
hsa04010MAPK signaling pathway64.050.0262CACNA2D1, BDNF, NTF3, RASGRF2, PPP3CB, CHUK
hsa05200Pathways in cancer74.730.0445BMP4, VEGFC, CCND1, GNAI1, HHIP, FZD5, CHUK
hsa05217Basal cell carcinoma32.030.0515BMP4, HHIP, FZD5
hsa04068FoxO signaling pathway42.700.0591CCND1, PLK2, IRS1, CHUK

[i] GBM, glioblastoma; KEGG, Kyoto Encyclopedia of Genes and Genomes; DEGs, differentially expressed genes.

Module screening from the PPI network

All of the downregulated DEGs and the top 1,000 upregulated DEGs were uploaded to STRING to draw the PPI network (data not shown). Cytoscape was used to evaluate primary modules of the PPI sub-network using the plug-in MCODE. A total of 1,222 nodes and 5,223 edges were analyzed in Cytoscape and the top two sub-networks are displayed in Fig. 5. There are 42 genes involved in sub-network 1, 13 genes involved in sub-network 2 and 47 genes involved in sub-network 3. GO and KEGG analyses were conducted separately for genes in the three top-score sub-networks. All of the results are shown in Tables II and III. In addition, the top 10 hub genes were identified using STRING software and were as follows: AURKB, RACGAP1, MCM2, MCM5, TGFBR1, SMAD3, RPN1, RPN2, VEGFC and FLT4.

Table II.

GO analysis of top 3 primary modules of PPI sub-network genes.

Table II.

GO analysis of top 3 primary modules of PPI sub-network genes.

Sub-networkCategoryTerm/gene functionGene count%P-value
Module 1GOTERM_BP_FAT GO:0007186~G-protein coupled receptor signaling pathway4197.623.95E-44
GOTERM_BP_FATGO:0007606~sensory perception of chemical stimulus21  501.25E-19
GOTERM_BP_FAT GO:0050907~detection of chemical stimulus involved in sensory perception2047.624.32E-19
GOTERM_MF_FAT GO:0004930~G-protein coupled receptor activity3173.817.39E-29
GOTERM_MF_FATGO:0004871~signal transducer activity3583.336.72E-26
GOTERM_MF_FAT GO:0004888~transmembrane signaling receptor activity3173.816.75E-24
GOTERM_CC_FATGO:0098552~side of membrane  819.054.08E-06
GOTERM_CC_FAT GO:0005834~heterotrimeric G-protein complex  49.521.84E-05
GOTERM_CC_FAT GO:0031226~intrinsic component of plasma membrane1228.572.00E-05
Module 2GOTERM_BP_FAT GO:0007200~phospholipase C-activating G-protein coupled receptor signaling pathway  758.334.05E-11
GOTERM_BP_FAT GO:0007186~G-protein coupled receptor signaling pathway1191.676.50E-11
GOTERM_BP_FAT GO:0006940~regulation of smooth muscle contraction  541.675.28E-08
GOTERM_MF_FAT GO:0004930~G-protein coupled receptor activity  9751.69E-08
GOTERM_MF_FAT GO:0004888~transmembrane signaling receptor activity  9753.48E-07
GOTERM_MF_FAT GO:0099600~transmembrane receptor activity  9754.76E-07
GOTERM_CC_FATGO:0005887~integral component of plasma membrane  866.675.03E-05
GOTERM_CC_FAT GO:0031226~intrinsic component of plasma membrane  866.676.55E-05
GOTERM_CC_FATGO:0043005~neuron projection  433.340.038
Module 3GOTERM_BP_FATGO:0006323~DNA packaging1328.261.31E-13
GOTERM_BP_FAT GO:0006334~nucleosome assembly1123.915.31E-12
GOTERM_BP_FATGO:0071103~DNA conformation change1328.266.24E-12
GOTERM_MF_FATGO:0046982~protein heterodimerization activity1226.095.17E-08
GOTERM_MF_FATGO:0046983~protein dimerization activity1532.612.25E-06
GOTERM_MF_FATGO:0042393~histone binding  613.041.75E-04
GOTERM_CC_FAT GO:0098687~chromosomal region1328.261.53E-10
GOTERM_CC_FAT GO:0005694~chromosome1839.132.91E-10
GOTERM_CC_FAT GO:0000786~nucleosome  919.576.45E-10

[i] GO, Gene Ontology; PPI, protein-protein interaction; BP, biological processes; CC, cellular component; MF, molecular function.

Table III.

KEGG pathway analysis of top 3 primary modules of PPI sub-network genes.

Table III.

KEGG pathway analysis of top 3 primary modules of PPI sub-network genes.

Sub-networkPathway IDNameGene count%P-valueGenes
Module 1hsa04740Olfactory transduction1740.482.86E-11OR5H1, OR2A4, OR5P2, OR1A2, OR5K1, OR1J4, OR7E24, OR2M4, OR2L2, OR1G1, OR6B1, OR2F1, OR3A1, OR8D1, OR51B4, OR2C3, OR7A10
hsa04062Chemokine signaling pathway716.673.49E-04GNGT1, CCR8, ADCY1, PPBP, GNAI1, CXCR4, CXCL11
hsa05032Morphine addiction49.520.01OPRM1, GNGT1, ADCY1, GNAI1
Module 2hsa04080Neuroactive ligand-receptor interaction866.671.55E-10P2RY10, CHRM3, TACR1, ADRA1B, ADRA1A, GNRHR, GHSR, NTSR2
hsa04020Calcium signaling pathway433.345.54E-04CHRM3, TACR1, ADRA1B, ADRA1A
hsa04970Salivary secretion3250.003CHRM3, ADRA1B, ADRA1A
Module 3hsa05203Viral carcinogenesis1226.092.68E-09HIST1H2BA, MAD1L1, HIST1H4L, HDAC3, KAT2B, HIST1H4A, HIST1H4B, HIST1H2BH, ACTN1, HIST1H4F, ACTN2, HIST1H4C
hsa05322Systemic lupus erythematosus1021.741.36E-08HIST1H2BA, HIST1H4L, HIST1H4A, HIST1H4B, HIST1H2BH, H2AFY2, ACTN1, HIST1H4F, ACTN2, HIST1H4C
hsa05034Alcoholism915.572.06E-06HIST1H2BA, HIST1H4L, HDAC3, HIST1H4A, HIST1H4B, HIST1H2BH, H2AFY2, HIST1H4F, HIST1H4C

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; PPI, protein-protein interaction.

Identification of prognostic biomarkers

After verification, the top four genes related to survival were identified as prognostic biomarkers of GBM and were as follows: RANBP17, ZNF734, NLRP2 and GPR1. In addition, the top four prognostic genes affected by sex were also identified and were as follows: CCDC81, NLRP2, SH3RF1 and TM7SF4. All of the results are shown in Figs. 6 and 7.

Discussion

Given the heterogeneity of GBM, which includes a variety of gene expression patterns and imaging-based phenotypes, understanding the molecular mechanism of GBM is of critical importance for the diagnosis and treatment of patients with GBM (21). As high-throughput sequencing and microarray analyses provide data on thousands of expressed genes in the human genome simultaneously, these techniques have been widely used to predict the potential therapeutic targets for GBM. In the present study, data from two datasets, GSE15824 and GSE51062, was extracted to identify the DEGs between GBM and normal brain control samples. The results showed that there are 9,175 upregulated and 166 downregulated DEGs between the GBM and normal brain control samples by using bioinformatics analysis. According to the function annotation results of the data, these DEGs were mainly involved in the defense response, cell development, regulation of signaling and of cell communication. By analyzing the PPI network, a number of key genes were identified that may provide new approaches for therapeutic studies of GBM.

In the present study, microarray data of 74 GBM samples and 5 normal brain control samples were downloaded from the GEO datasets under the accession numbers GSE15824 and GSE51062. Using a series of essential packages in R software, all raw CEL data from the two datasets was extracted and microarray chip quality control was conducted. It is worth noting that there were many more upregulated than downregulated genes. In order to understand the interactions of the DEGs, GO and KEGG pathway analyses were performed. GO term analysis of the top 1,000 upregulated genes and all of the downregulated genes showed that the upregulated DEGs were mainly involved defense response, nervous system development and cell development, and the downregulated DEGs were involved in regulation of signaling, positive regulation of transport and regulation of cell communication. Genes that control tumor progression and enhancement of tumor defense were upregulated and genes that control tumor signaling and cell communication were downregulated, which shows genome alteration consistent with the clinical manifestations. As observed in clinical practice, many patients with low-grade gliomas develop a high-grade glioma after recurrence. Genes controlling tumor progression may be good biomarkers for postoperative recurrence monitoring. A previous study has indicated that MGMT methylation status has great importance in tumor prognosis, especially in patients who have undergone the first surgery (22). Although MGMT methylation status was not reflected in the 2016 edition of the WHO pathological grading criteria, it is still an influential gene biomarker for the prognosis of patients with GBM.

KEGG analysis of the upregulated genes in the present study identified that they are mainly involved in cytokine relevant pathways and ligand-receptor relevant pathways, including the pathway of neuroactive ligand-receptor interaction, cytokine-cytokine receptor interaction and Jak-STAT signaling pathway. The downregulated genes were mainly involved in pathways of classical tumor signaling, including TGF-β signaling pathway, MAPK signaling pathway, pathways in cancer, basal cell carcinoma and FoxO signaling pathway (2328). It is noteworthy that a number of the downregulated genes (BMP4, FZD5 and HHIP) in the GBM samples are involved in the basal cell carcinoma pathway, which may be because GBM originates from the source of nerve epithelium. These three genes may exert some of the same effects in both basal cell carcinoma and GBM. On the other hand, it is also notable that the majority of the downregulated genes are not involved in KEGG pathways and therefore exist in a scattered form.

The PPI network of the DEGs was constructed using the plug-in MCODE in Cytoscape and the top degree hub genes were listed as follows: AURKB, RACGAP1, MCM2, MCM5, TGFBR1, SMAD3, RPN1, RPN2, VEGFC and FLT4. These genes were identified by the degree of connection of every gene according to the connectivity measured by the prior analysis with STRING. All of the hub genes are upregulated except VEGFC. AURKB was identified as one of the hub genes which exhibits the highest degree of connectivity. AURKB encodes Aurora B, which is an important kinase that regulates cell mitosis. A recent study claims that Aurora B activity is an important modulator of the taxane response in non-small cell lung cancer cells (29). A 2012 study of GBM showed that inhibition of Aurora kinases enhances chemosensitivity to temozolomide and causes radiosensitization in GBM cells (30). RACGAP1 encodes Rac GTPase activating protein 1, a component of the centralspindlin complex that serves as a microtubule-dependent and Rho-mediated signal required for myosin contractile ring formation during cell cycle cytokinesis. RACGAP1 plays key roles in the control of cell growth and differentiation of hematopoietic cells through mechanisms other than regulating Rac GTPase activity. A previous study showed a strong connection in the expression of AURKA and RACGAP1 (31). However, there is a lack of data regarding whether AURKB has a strong connection with the expression of RACGAP1. MCM2 and MCM5 regulate cell proliferation. A study in 2017 showed that MCM2 can be used as an alternative to Ki-67 for measuring breast cancer cell proliferation (32). MCM5 also has a regulatory effect in cell proliferation (33). There is little research regarding MCM2 and MCM5 in GBM. As for TGFBR1, a study in 2016 showed that mi-181c inhibits cell invasion, migration and mesenchymal transition by downregulating TGFBR1 (34). SAMD3 is also a miR-regulated target in GBM (35). As shown in the present study, these six genes are all involved in the cell cycle. TGFBR1, SMAD3, VEGFC and FLT4 are involved in the regulation of epithelial cell proliferation. VEGFC has been shown to have an important role in GBM angiogenesis (36). FLT4 has been found to be differentially expressed in angiosarcoma of the breast and lymphatic filariasis (37,38). However, to the best of our knowledge, there are no studies concerning FLT4 in GBM. Previous studies have shown that little is known about these two genes (VEGFC, FLT4). RPN1 has been demonstrated to function in proteasome-mediated proteolysis (39). RPN2 has been shown as a promising biomarker in colorectal cancer (40). Both of these genes have not been studied clearly in GBM; therefore, they are two potential biomarkers which merit further studies.

KEGG analysis of the whole PPI network in the present study shows that the development of GBM is associated with the TGF-β signaling pathway, MAPK signaling pathway, pathways in cancer, basal cell carcinoma, FoxO signaling pathway, neuroactive ligand-receptor interaction, Jak-STAT signaling pathway, circadian entrainment and cytokine-cytokine receptor interaction. In addition, certain genes were also involved in the Wnt pathway, cell cycle and cell proliferation.

As shown in the survival curve, RANBP17, ZNF734, NLRP2, GPR1, CCDC81, SH3RF1 and TM7SF4 are good prognostic biomarkers for GBM. It is noteworthy that the expression of a number of the genes not only affects survival time, but also shows sex differences in the GBM samples. According to a recent study (41), the female sex plays an important role in the survival of one subtype of GBM. This may mean that sex plays an important role in cancer. The prognosis of patients of different sexes varies greatly. To date, the international diagnostic and treatment criteria for glioma does not include sex differences. The present study reveals that different expression levels of key genes and sex may provide more information with regards to treating patients with GBM.

In conclusion, the large-scale data analysis of the present study provides a comprehensive bioinformatics analysis of DEGs that may be involved in the progress of GBM. Joint analysis of two databases helped to overcome the shortcomings of insufficient samples, and existing data was reused to conduct a novel analysis. This study provides a set of important targets for future investigation into the molecular mechanisms and prognostic biomarkers involved in GBM. However, follow-up molecular biological experiments and verification with many more clinical samples are required to confirm the functions of the identified genes in patients with different subtypes of GBM.

Acknowledgements

The present study was supported by International Program for Ph.D. Candidates, Sun Yat-sen University.

Funding

The present study was financially supported by the National Natural Science Foundation of China (grant no. 31360258), the Fundamental Research Funds for the Central Universities (grant no. 17ykpy84), Guangdong Provincial Natural Science Foundation (grant nos. 2015A030313047, 2015A030313077 and 2017A030310192), and Science and Technology Planning Project of Guangdong Province of China (grant no. 2016A020215036).

Availability of data and materials

The datasets analyzed in the present study are available from the GEO repository, https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE15824 and https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE51062.

Authors' contributions

LZ, HT, JX and KG conceived and designed the study; LZ analyzed and interpreted the microarray datasets, and produced the manuscript. LZ, HT, FW, LC, SO and TW analyzed the data. LZ wrote the paper, HT submitted the manuscript, and LZ and HT revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Steiner HH, Herold-Mende C, Bonsanto M, Geletneky K and Kunze S: Prognosis of brain tumors: Epidemiology, survival time and clinical course. Versicherungsmedizin. 50:173–179. 1998.(In French). PubMed/NCBI

2 

Fisher JL, Schwartzbaum JA, Wrensch M and Wiemels JL: Epidemiology of brain tumors. Neurol Clin. 25:867–890. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Asthagiri AR, Pouratian N, Sherman J, Ahmed G and Shaffrey ME: Advances in brain tumor surgery. Neurol Clin. 25:975–1003. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Grzmil M, Morin P Jr, Lino MM, Merlo A, Frank S, Wang Y, Moncayo G and Hemmings BA: MAP kinase-interacting kinase 1 regulates SMAD2-dependent TGF-β signaling pathway in human glioblastoma. Cancer Res. 71:2392–2402. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Walsh AM, Kapoor GS, Buonato JM, Mathew LK, Bi Y, Davuluri RV, Martinez-Lage M, Simon MC, O'Rourke DM and Lazzara MJ: Sprouty2 drives drug resistance and proliferation in glioblastoma. Mol Cancer Res. 13:1227–1237. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Gerber NK, Goenka A, Turcan S, Reyngold M, Makarov V, Kannan K, Beal K, Omuro A, Yamada Y, Gutin P, et al: Transcriptional diversity of long-term glioblastoma survivors. Neuro Oncol. 16:1186–1195. 2014. View Article : Google Scholar : PubMed/NCBI

8 

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:D1005–D1010. 2011. View Article : Google Scholar : PubMed/NCBI

9 

NIH: The Cancer Genome Atlas, . TCGA Data Portal. https://tcga-data.nci.nih.gov/docs/publications/tcga/?June 30–2016

10 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 world health organization classification of tumors of the central nervous system: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Diboun I, Wernisch L, Orengo CA and Koltzenburg M: Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics. 7:2522006. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

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

14 

Gene Ontology Consortium, . The gene ontology (GO) project in 2006. Nucleic Acids Res. 34:D322–D336. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nature Genetics. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization and integrated discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

18 

Franceschini A, Szklarezyk 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

19 

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

20 

Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Rodriguez I, Chakravarthi BVSK and Varambally S: UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 19:649–658. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Liu TT, Achrol AS, Mitchell LA, Rodriguez SA, Feroze A, Iv M, Kim C, Chaudhary N, Gevaert O, Stuart JM, et al: Magnetic resonance perfusion image features uncover an angiogenic subgroup of glioblastoma patients with poor survival and better response to antiangiogenic treatment. Neuro Oncol. 19:997–1007. 2017.PubMed/NCBI

22 

Brandes A, Franceschi E, Tosoni A, Fioravanti A, Agati R, Andreoli A, Mazzocchi V, Morandi L, Bartolini S and Ermani M: Change in MGMT methylation status between first and second surgery for recurrence: Clinical implications. Eur J Cancer Suppl. 7:4952009. View Article : Google Scholar

23 

Akbari A, Farahnejad Z, Akhtari J, Abastabar M, Mobini GR and Mehbod AS: Staphylococcus aureus enterotoxin B down-regulates the expression of transforming growth factor-beta (TGF-β). Signaling Transducers in Human Glioblastoma. Jundishapur J Microbiol. 9:e272972016. View Article : Google Scholar : PubMed/NCBI

24 

Jiang C, Shen F, Du J, Hu Z, Li X, Su J, Wang X and Huang X: MicroRNA-564 is downregulated in glioblastoma and inhibited proliferation and invasion of glioblastoma cells by targeting TGF-β1. Oncotarget. 7:56200–56208. 2016.PubMed/NCBI

25 

Li J, Ren S, Liu Y, Lian Z, Dong B, Yao Y and Xu Y: Knockdown of NUPR1 inhibits the proliferation of glioblastoma cells via ERK1/2, p38 MAPK and caspase-3. J Neuro Oncol. 132:15–26. 2017. View Article : Google Scholar

26 

Kim EH, Song HS, Yoo SH and Yoon M: Tumor treating fields inhibit glioblastoma cell migration, invasion and angiogenesis. Oncotarget. 7:65125–65136. 2016.PubMed/NCBI

27 

Firat E and Niedermann G: FoxO proteins or loss of functional p53 maintain stemness of glioblastoma stem cells and survival after ionizing radiation plus PI3K/mTOR inhibition. Oncotarget. 7:54883–54896. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Masui K, Tanaka K, Akhavan D, Babic I, Gini B, Matsutani T, Iwanami A, Liu F, Villa GR, Gu Y, et al: mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 18:726–739. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Al-Khafaji AS, Davies MP, Risk JM, Marcus MW, Koffa M, Gosney JR, Shaw RJ, Field JK and Liloglou T: Aurora B expression modulates paclitaxel response in non-small cell lung cancer. Br J Cancer. 116:592–599. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Borges KS, Castro-Gamero AM, Moreno DA, da Silva Silveira V, Brassesco MS, de Paula Queiroz RG, de Oliveira HF, Carlotti CG Jr, Scrideli CA and Tone LG: Inhibition of Aurora kinases enhances chemosensitivity to temozolomide and causes radiosensitization in glioblastoma cells. J Cancer Res Clin Oncol. 138:405–414. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Bornschein J, Nielitz J, Drozdov I, Selgrad M, Wex T, Jechorek D, Link A, Vieth M and Malfertheiner P: Expression of aurora kinase A correlates with the Wnt-modulator RACGAP1 in gastric cancer. Cancer Med. 5:516–526. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Yousef EM, Furrer D, Laperriere DL, Tahir MR, Mader S, Diorio C and Gaboury LA: MCM2: An alternative to Ki-67 for measuring breast cancer cell proliferation. Mod Pathol. 30:682–697. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Su Z, Zheng X, Zhang X, Wang Y, Zhu S, Lu F, Qu J and Hou L: Sox10 regulates skin melanocyte proliferation by activating the DNA replication licensing factor MCM5. J Dermatol Sci. 85:216–225. 2017. View Article : Google Scholar : PubMed/NCBI

34 

He X, Liu Z, Peng Y and Yu C: MicroRNA-181c inhibits glioblastoma cell invasion, migration and mesenchymal transition by targeting TGF-β pathway. Biochem Biophys Res Commun. 469:1041–1085. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Wu ZB, Cai L, Lin SJ, Lu JL, Yao Y and Zhou LF: The miR-92b functions as a potential oncogene by targeting on Smad3 in glioblastomas. Brain Res. 1529:16–25. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Carpenter RL, Paw I, Zhu H, Sirkisoon S, Xing F, Watabe K, Debinski W and Lo HW: The gain-of-function GLI1 transcription factor TGLI1 enhances expression of VEGF-C and TEM7 to promote glioblastoma angiogenesis. Oncotarget. 6:22653–22655. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Sheik Y, Qureshi SF, Mohhammed B and Nallari P: FOXC2 and FLT4 gene variants in lymphatic filariasis. Lymphat Res Biol. 13:112–119. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Cornejo KM, Deng A, Wu H, Cosar EF, Khan A, St Cyr M, Tomaszewicz K, Dresser K, O'Donnell P and Hutchinson L: The utility of MYC and FLT4 in the diagnosis and treatment of postradiation atypical vascular lesion and angiosarcoma of the breast. Hum Pathol. 46:868–875. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Rani N, Aichem A, Schmidtke G, Kreft SG and Groettrup M: FAT10 and NUB1L bind to the VWA domain of Rpn10 and Rpn1 to enable proteasome-mediated proteolysis. Nat Commun. 3:7492012. View Article : Google Scholar : PubMed/NCBI

40 

Zhang J, Yan B, Späth SS, Qun H, Cornelius S, Guan D, Shao J, Hagiwara K, Van Waes C, Chen Z, et al: Integrated transcriptional profiling and genomic analyses reveal RPN2 and HMGB1 as promising biomarkers in colorectal cancer. Cell Biosci. 5:532015. View Article : Google Scholar : PubMed/NCBI

41 

Liau CT, Chou WC, Wei KC, Chang CN, Toh CH and Jung SM: Female sex, good performance status and bevacizumab-induced hypertension associated with survival benefit in Asian patients with recurrent glioblastoma treated with bevacizumab. Asia Pac J Clin Oncol. 14:e8–e14. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 18 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou L, Tang H, Wang F, Chen L, Ou S, Wu T, Xu J and Guo K: Bioinformatics analyses of significant genes, related pathways and candidate prognostic biomarkers in glioblastoma. Mol Med Rep 18: 4185-4196, 2018
APA
Zhou, L., Tang, H., Wang, F., Chen, L., Ou, S., Wu, T. ... Guo, K. (2018). Bioinformatics analyses of significant genes, related pathways and candidate prognostic biomarkers in glioblastoma. Molecular Medicine Reports, 18, 4185-4196. https://doi.org/10.3892/mmr.2018.9411
MLA
Zhou, L., Tang, H., Wang, F., Chen, L., Ou, S., Wu, T., Xu, J., Guo, K."Bioinformatics analyses of significant genes, related pathways and candidate prognostic biomarkers in glioblastoma". Molecular Medicine Reports 18.5 (2018): 4185-4196.
Chicago
Zhou, L., Tang, H., Wang, F., Chen, L., Ou, S., Wu, T., Xu, J., Guo, K."Bioinformatics analyses of significant genes, related pathways and candidate prognostic biomarkers in glioblastoma". Molecular Medicine Reports 18, no. 5 (2018): 4185-4196. https://doi.org/10.3892/mmr.2018.9411