Open Access

DNA methylation profiling identifies potentially significant epigenetically‑regulated genes in glioblastoma multiforme

  • Authors:
    • Shifeng Kan
    • Song Chai
    • Wenhua Chen
    • Bo Yu
  • View Affiliations

  • Published online on: June 21, 2019     https://doi.org/10.3892/ol.2019.10512
  • Pages: 1679-1688
  • Copyright: © Kan 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 multiforme (GBM) is one of the most lethal and damaging types of human cancer. The current study was conducted to identify differentially methylated genes (DMGs) between GBM and normal controls, and to improve our understanding of GBM at the epigenetic level. The DNA methylation profile of GBM was downloaded from the Gene Expression Omnibus (GEO) database using the accession number GSE50923. The MethyAnalysis package was applied to identify DMGs between GBM and controls, which were then analyzed by functional enrichment analysis. Protein‑protein interaction (PPI) networks were constructed using the hypermethylated and hypomethylated genes. Finally, transcription factors (TFs) that can regulate the hypermethylated and hypomethylated genes were predicted, followed by construction of transcriptional regulatory networks. Furthermore, another relevant dataset, GSE22867, was downloaded from the GEO database for data validation. A total of 476 hypermethylated and 850 hypomethylated genes were identified, which were mainly associated with the functions of ‘G‑protein‑coupled receptors ligand binding’, ‘cytokine activity’, ‘cytokine‑cytokine receptor interaction’, and ‘D‑glutamine and D‑glutamate metabolism’. The hypermethylated gene neuropeptide Y (NPY) and the hypomethylated gene tumor necrosis factor (TNF) demonstrated high degrees in the PPI network. Forkhead box protein A1 (FOXA1), potassium voltage‑gated channel subfamily C member 3 (KCNC3) and caspase‑8 (CASP8) exhibited high degrees in the transcriptional regulatory networks. In addition, the methylation profiles of NPY, TNF, FOXA1, KCNC3 and CASP8 were confirmed by another dataset. In summary, the present study systematically analyzed the DNA methylation profile of GBM using bioinformatics approaches and identified several abnormally methylated genes, providing insight into the molecular mechanism underlying GBM.

Introduction

Glioblastoma multiforme (GBM), which develops from astrocytes, is the most common primary brain tumor in adults and one of the most damaging types of human cancer. Despite aggressive multimodal treatment with surgery, radiotherapy and chemotherapy, the prognosis of GBM is extremely poor (1,2). Additionally, GBM demonstrates a high potential to infiltrate the brain parenchyma, which poses a challenge to the available treatment strategies. Typically, GBM results in mortality at 12–15 months post-diagnosis (1). Therefore, novel therapies for GBM are urgently required.

Identifying the mechanisms underlying the development of GBM is important for the development of new treatments. The genetic alterations that affect genes controlling cell growth, apoptosis and invasion have been widely examined in GBM (3). Epigenetic alterations have also been identified to be involved in GBM by affecting the expression of cancer-associated genes alone or in combination with genetic mechanisms (4). Aberrant methylation of gene promoters is the most widely studied epigenetic change that occurs during oncogenesis. It is understood that increased methylation, termed hypermethylation, in the CpG island promotes carcinogenesis by silencing tumor suppressor genes, while loss of methylation, termed hypomethylation, enhances the transcriptional activation of oncogenes and induces chromosomal instability (5,6). Decreased expression of tumor suppressor genes, including retinoblastoma gene, phosphatase and tensin homolog, and TP53, is associated with CpG island promoter hypermethylation and has been reported in GBM (79). Epigenetic silencing of O6-methylguanine-DNA methyltransferase, a DNA repair gene that can protect cancer cells from chemotherapeutic alkylating agents, has been revealed to be significantly associated with longer survival times in patients with GBM who are treated with alkylating agents (10,11). Such epigenetic changes may be promising targets for epigenetic anticancer treatments. Indeed, the DNA-demethylating agents 5-azacitidine and 5-aza-2′-deoxycitidine have been approved by the Food and Drug Administration for the treatment of myelogenous leukemia and myelodysplastic syndromes (12).

Bioinformatics tools and algorithms assist the processing and analysis of high-throughput DNA methylation data (13,14). For example, a previous study used a joint analysis of DNA methylation and gene expression data of GBM to demonstrate that changes in DNA methylation can be associated with survival outcome (15). In addition, a recent study used a computational approach to integrate gene expression and genomic or methylation data to investigate biological networks in GBM (16). The current study used bioinformatics approaches to reanalyze the DNA methylation data deposited by Lai et al (17). The present results may improve understanding of the epigenetic regulation mechanism of GBM and provide potential gene methylation biomarkers for GBM, which may contribute to the development of treatments.

Materials and methods

Microarray data

DNA methylation data from the GBM study by Lai et al (17) were retrieved from the National Center for Biotechnology Information Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/projects/geo/) with the accession number GSE50923. The DNA methylation profiles of 54 GBM samples and 24 control brain samples were previously investigated using Illumina HumanMethylation27 BeadChip. The patient details are presented in the original study by Lai et al (17). Only data from 31 GBM and 24 control brain samples were present in the GEO database, therefore, only these data were analyzed in the current study.

Identification of differentially methylated regions (DMRs)

The original methylation data were processed using the Bioconductor lumi package version 2.18.0, which is designed to process the Illumina microarray data (18,19). Following quality control and background correction, these data were scaled by quantile normalization as implemented in the lumi package. The methyAnalysis package version 1.8.0 (20) was applied to identify DMRs between GBM and controls. P<0.001 and a difference in methylation levels, calculated as the M-value (19), of >1 or <-1 were considered to indicate a statistically significant DMR. The identified DMRs were then annotated using the methyAnalysis package and DMRs located at gene promoter regions, within 2 kb upstream of the transcription start site, were selected for further analysis.

Functional enrichment analysis

Functional enrichment analysis was conducted to explore the most significant differentially methylated genes (DMGs) with relevant biological functions. Gene Ontology (GO) is a bioinformatics tool for annotating genes, gene products and sequences using defined GO terms (21). The Kyoto Encyclopedia of Genes and Genomes (KEGG) is a comprehensive database linking genomic data, stored in the GENES database, with higher order functional information, stored in the PATHWAY database (22). Reactome is a free online database of biological pathways (23). The KEGG pathway, Reactome pathway and GO functional terms for the DMGs were identified using the following cut-off criteria: P<0.05 and number of over-represented genes >2.

Construction of a protein-protein interaction (PPI) network

The PPI data were retrieved from the STRING database (http://string.embl.de/). Subsequently, the DMGs were mapped into these interactions, and DMG pairs with an interaction score >0.9 were selected to construct the PPI networks, which were visualized using Cytoscape software version 3.1.0 (24).

Classification of DMGs

TSGene (25) is a database for tumor suppressor genes (TSGs) and the TAG database (26) provides information on well-characterized oncogenes and TSGs. The known TSGs and oncogenes were identified from the list of DMGs based on information retrieved from the TSGene and TAG databases.

Identification of transcription factors (TFs) regulating DMGs and construction of a transcriptional regulatory network

The TF regulation data were downloaded from the Encyclopedia of DNA Elements data portal (27,28), and TFs that regulate the hypermethylated genes and hypomethylated genes were identified. The hypergeometric distribution was used to assign a P-value for the prediction of these TFs and an adjusted P-value of <0.05 was considered significant. Furthermore, transcriptional regulatory networks of hypermethylated genes and hypomethylated genes were constructed using Cytoscape (version 3.6.0) (24).

Data validation

To validate the identified DMGs from GSE50923, another relevant DNA Methylation Profiling dataset, GSE22867 (29), was downloaded from the GEO database and used for data validation. GSE22867 included 55 GBM samples and 3 control brain samples. The platform was GPL8490 Illumina HumanMethylation27 BeadChip (HumanMethylation27_270596_v.1.2). The β-value was calculated based on the methylated and unmethylated signal of the sample data, and a t-test was implemented using the genefilter package (version 1.56.0) (30) to identify significant differentially methylated CpGs (P<0.05). In addition, selected CpG sites exhibited a mean methylation (β-value) difference ≥0.05 between the disease group and the control group. Subsequently, the genes covering the differentially methylated CpGs were identified and compared with the identified DMGs from GSE50923.

Results

Identification of DMGs

Based on the methyAnalysis package, a total of 2,407 DMRs were identified between the GBM samples and normal controls. Among these DMRs, 476 hypermethylated and 850 hypomethylated regions were located in gene promoters, which clearly distinguished GBM from normal samples (Fig. 1).

Functional enrichment analysis

To gain insight into the dysregulated biological processes induced by DMGs, separate functional enrichment analyses were performed for the hypermethylated and hypomethylated genes. The data indicated that certain KEGG pathways, including ‘neuroactive ligand-receptor interaction’, and ‘D-glutamine and D-glutamate metabolism’, were enriched by hypermethylated genes, while ‘cytokine-cytokine receptor interaction’ and ‘hematopoietic cell lineage’ were enriched by hypomethylated genes (Table I). GO terms were grouped into the following three categories: Biological process, molecular function and cellular component. The most significantly enriched GO biological process and molecular function terms were ‘anatomical structure development’ and ‘sequence-specific DNA-binding transcription factor activity’, respectively, while no statistically significant enriched cellular component terms were identified (Table II). For hypomethylated genes, the most significant GO terms were ‘defense response’, ‘cytokine activity’ and ‘extracellular region’ (Table II). Reactome pathway analysis demonstrated that both hypermethylated and hypomethylated genes were closely associated with the ‘G-protein-coupled receptor (GPCR) ligand binding pathway’ (Table III).

Table I.

Top five KEGG pathways enriched by hypermethylated genes and hypomethylated genes.

Table I.

Top five KEGG pathways enriched by hypermethylated genes and hypomethylated genes.

A, Hypermethylated genes

KEGG IDKEGG pathwayP-value
4080Neuroactive ligand-receptor interaction 1.18×10−3
471D-Glutamine and D-glutamate metabolism 3.62×10−3
4114Oocyte meiosis 6.76×10−3
250Alanine, aspartate and glutamate metabolism 7.87×10−3
4971Gastric acid secretion 1.02×10−2

B, Hypomethylated genes

KEGG IDKEGG pathwayP-value

4060Cytokine-cytokine receptor interaction 1.22×10−10
4640Hematopoietic cell lineage 1.49×10−5
5150Staphylococcus aureus infection 1.35×10−4
4610Complement and coagulation cascades 2.72×10−4

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes.

Table II.

Top five GO terms enriched by hypermethylated genes and hypomethylated genes.

Table II.

Top five GO terms enriched by hypermethylated genes and hypomethylated genes.

A, Hypermethylated genes

GO IDGO termP-value
Biological process
  GO:0048856Anatomical structure development 3.19×10−8
  GO:0032502Developmental process 4.06×10−8
  GO:0044707 Single-multicellular organism process 4.85×10−8
  GO:0032501Multicellular organismal process 8.26×10−8
  GO:0007275Multicellular organismal development 3.47×10−7
Molecular function
  GO:0003700Sequence-specific DNA binding transcription factor activity 5.04×10−9
  GO:0001071Nucleic acid binding transcription factor activity 5.51×10−9
  GO:0043565Sequence-specific DNA binding 1.76×10−8
  GO:0044212Transcription regulatory region DNA binding 6.04×10−7
  GO:0000975Regulatory region DNA binding 9.99×10−7
Cellular component
  GO:0044459Plasma membrane part 2.37×10−1
  GO:0019897Extrinsic to plasma membrane 2.61×10−1
  GO:0008076Voltage-gated potassium channel complex 4.53×10−1
  GO:0034705Potassium channel complex 4.53×10−1
  GO:0019898Extrinsic to membrane 5.85×10−1

B, Hypomethylated genes

GO IDGO termP-value

Biological process
  GO:0006952Defense response 8.19×10−12
  GO:0006954Inflammatory response 9.55×10−12
  GO:0050896Response to stimulus 6.68×10−11
  GO:0002376Immune system process 8.73×10−11
  GO:0009611Response to wounding 6.82×10−10
Molecular function
  GO:0005125Cytokine activity 3.13×10−7
  GO:0005126Cytokine receptor binding 1.30×10−5
  GO:0004872Receptor activity 6.25×10−5
  GO:0005102Receptor binding 1.39×10−3
  GO:0005506Iron ion binding 8.86×10−3
Cellular component
  GO:0005576Extracellular region <1.00×10−3
  GO:0005615Extracellular space <1.00×10−3
  GO:0044421Extracellular region part <1.00×10−3
  GO:0071944Cell periphery 3.84×10−9
  GO:0005886Plasma membrane 5.56×10−9

Table III.

Top five Reactome pathways enriched by hypermethylated genes and hypomethylated genes.

Table III.

Top five Reactome pathways enriched by hypermethylated genes and hypomethylated genes.

A, Hypermethylated genes

Reactome IDNameP-value
500792GPCR ligand binding 8.88×10−2
112316Neuronal system 1.61×10−1
373076Class A/1 (Rhodopsin-like receptors) 3.55×10−1
418597G α (z) signaling events 4.98×10−1
1296072Voltage-gated potassium channels 9.92×10−1

B, Hypomethylated genes

Reactome IDNameP-value

500792GPCR ligand binding 5.94×10−4
211897Cytochrome P450-arranged by substrate type 1.05×10−3
373076Class A/1 (Rhodopsin-like receptors) 5.68×10−3
211945Phase 1-Functionalization of compounds 6.96×10−3

[i] GPCR, G-protein-coupled receptor.

Construction of PPI networks

A total of 476 hypermethylated and 850 hypomethylated genes were mapped to the STRING database and significant interactions with scores >0.9 were selected. A total of 83 hypermethylated genes and 204 hypomethylated genes were screened to separately construct PPI networks involving hypermethylated genes (Fig. 2) and hypomethylated genes (Fig. 3). The DMGs adenylate cyclase type 2 (ADCY2; degree, 18), neuropeptide Y (NPY; degree, 11) and somatostatin (SST; degree, 11) demonstrated the highest degrees, determined by the number of interactions in the hypermethylated gene network. Kininogen 1 (KNG1; degree, 27), proto-oncogene tyrosine protein kinase (SRC; degree, 18) and tumor necrosis factor (TNF; degree, 15) exhibited the highest degrees in the hypomethylated gene network.

Classification of the DMGs

A total of 43 cancer-associated genes were identified among the hypermethylated genes, including 8 oncogenes and 25 TSGs, while 41 hypomethylated genes were revealed to be cancer-associated genes, including 10 oncogenes and 24 TSGs.

Prediction of TFs that target the DMGs and construction of transcriptional regulatory networks

A total of 55 TFs that could regulate the hypermethylated genes were identified, including polycomb repressive complex 2 subunit, neuron-restrictive silencer factor and cohesin complex component. Additionally, 55 TFs that could regulate the hypomethylated genes were identified, including signal transducer and activator of transcription 3, and forkhead box protein A1 (FOXA1). Notably, FOXA1 was also revealed to be hypermethylated. The transcriptional regulatory networks of hypermethylated genes and hypomethylated genes are presented in Figs. 4 and 5, respectively. The top 10 hypermethylated genes and hypomethylated genes with high node degrees in the regulatory networks are listed in Table IV, including the hypermethylated genes, FOXA1 (degree, 35), adenine phosphoribosyltransferase (degree, 17) and potassium voltage-gated channel subfamily C member 3 (KCNC3; degree, 15), and the hypomethylated genes, proliferating cell nuclear antigen-associated factor (degree, 14) and caspase-8 (CASP8; degree, 12).

Table IV.

Top 10 hypermethylated and hypomethylated genes with high node degree in the transcriptional regulatory networks.

Table IV.

Top 10 hypermethylated and hypomethylated genes with high node degree in the transcriptional regulatory networks.

GeneNode degree
Hypermethylated
  FOXA135
  APRT17
  KCNC315
  LSR15
  SNAI113
  NUP9813
  PDE4D12
  RPL3112
  RPL3112
  CREM12
Hypomethylated
  KIAA010114
  CASP812
  TMA1612
  FAM107B11
  LINC0026311
  RXRB11
  PHLDA110
  DEGS110
  KAZALD110
  C1RL-AS1  9
Data validation

From the GSE22867 validation dataset, 2,151 DMGs were identified. As demonstrated in Fig. 6, a total of 690 overlapping DMGs were revealed. In particular, the hypermethylated genes NPY, FOXA1 and KCNC3, as well as the hypomethylated genes TNF and CASP8, were among the overlapping DMGs.

Discussion

The current study systemically analyzed the DNA methylation profile of GBM samples to improve our understanding of GBM at the epigenetic level and identify potential biomarkers as therapeutic targets for GBM.

Differential analysis revealed that the methylation levels of 1,326 genes were altered in patients with GBM compared with those in the controls. The biological functions most associated with the DMGs were explored by functional enrichment analysis. The results revealed that DMGs were closely associated with the functions of ‘GPCR ligand binding’, ‘cytokine activity’, ‘cytokine-cytokine receptor interaction’, and ‘D-glutamine and D-glutamate metabolism’. Both ‘cytokine activity’ and ‘cytokine-cytokine receptor interaction’ are associated with inflammatory responses, and there is a close association between inflammation and cancer. Unresolved inflammation resulting from a failure in the regulation of the immune response creates a tumor microenvironment, an important aspect of tumorigenesis proliferation, survival and migration (31,32). GPCR ligands bind to GPCRs and activate the downstream signaling that regulates cellular physiology. Aberrant G-protein signaling is closely associated with cancer development and progression (33).

To investigate whether abnormally methylated genes are associated with GBM, PPI networks of the screened genes were constructed and the degrees of the nodes were analyzed in the present study. The results demonstrated that the top three genes with the highest node degrees were ADCY2, NPY and SST in the hypermethylated genes network, and KNG1, SRC and TNF in the hypomethylated genes network. Among these genes, NPY and TNF were validated in the alternative dataset. NPY encodes a 36-amino acid neuropeptide that acts as a neurotransmitter in the brain and autonomic nervous system of humans. Using bioinformatic analysis, abnormal methylation of NPY has been observed in numerous types of cancer (3436). Furthermore, a previous study reported that NPY is expressed in various types of intracranial tumor in humans and that NPY mRNA is detectable in the temporal lobe in higher quantities compared with that in tumors (37). In addition, the present study identified that TNF was hypomethylated, which may lead to upregulation of the gene. The expression of TNF-α has been identified to be increased in GBM (38), which corresponds with the current study. Therefore, NPY and TNF may be involved in GBM due to their abnormal methylation and may result in the disturbance of biological processes.

The hypermethylated genes FOXA1 and KCNC3, as well as the hypomethylated gene CASP8, exhibited high degrees in the transcriptional regulatory networks, which was also confirmed by the second dataset. FOXA1 acts as a TF and has been demonstrated to be a potential regulator of human glioma progression (39). KCNC3, encoding the Kv3. 3 voltage-gated potassium channel, is expressed in various neuronal cell types that are involved in motor function (40). Previous studies have demonstrated an association between KCNC3 expression and the poor prognosis of patients with GBM (41). Patients with GBM with higher KCNC3 expression exhibit improved survival times (42). Genomic loss of CASP8 by DNA methylation may result in tumor resistance to therapies targeting TNF-related apoptosis-inducing ligand-associated apoptosis pathways (43). In summary, the methylation of FOXA1, KCNC3 and CASP8 in GBM should be investigated in future studies, as this may promote the development of therapeutic approaches.

In conclusion, the current study comprehensively analyzed the DNA methylation profile of GBM using bioinformatics approaches and identified several abnormally methylated genes, including NPY, TNF, FOXA1, KCNC3 and CASP8. The findings of the present study improve the understanding of the molecular mechanism underlying GBM, and provide potential biomarkers for GBM and the development of novel treatment strategies. However, the number of samples included in this study was relatively small. Experimental verification and additional studies with larger sample sizes are required to confirm the present results.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets generated and/or analyzed during the current study are available as datasets GSE50923 and GSE22867 from the GEO database (http://www.ncbi.nlm.nih.gov/projects/geo/).

Authors contributions

SK and SC conceived and designed the study, and drafted the manuscript. WC acquired, analyzed and interpreted the data, and conducted the statistical analysis. BY acquired and interpreted data, and revised the manuscript for important intellectual content. 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 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Rock K, McArdle O, Forde P, Dunne M, Fitzpatrick D, O'Neill B and Faul C: A clinical review of treatment outcomes in glioblastoma multiforme - the validation in a non-trial population of the results of a randomised phase III: Has a more radical approach improved survival? Br J Radiol. 85:e729–e733. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Holland EC: Gliomagenesis: Genetic alterations and mouse models. Nat Rev Genet. 2:120–129. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Nagarajan RP and Costello JF: Epigenetic mechanisms in glioblastoma multiforme. Semin Cancer Biol. 19:188–197. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Herman JG and Baylin SB: Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 349:2042–2054. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Karpf AR and Matsui SI: Genetic disruption of cytosine DNA methyltransferase enzymes induces chromosomal instability in human cancer cells. Cancer Res. 65:8635–8639. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Nakamura M, Yonekawa Y, Kleihues P and Ohgaki H: Promoter hypermethylation of the RB1 gene in glioblastomas. Lab Invest. 81:77–82. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Amatya VJ, Naumann U, Weller M and Ohgaki H: TP53 promoter methylation in human gliomas. Acta Neuropathol. 110:178–184. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Baeza N, Weller M, Yonekawa Y, Kleihues P and Ohgaki H: PTEN methylation and expression in glioblastomas. Acta Neuropathol. 106:479–485. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Kreth S, Thon N, Eigenbrod S, Lutz J, Ledderose C, Egensperger R, Tonn JC, Kretzschmar HA, Hinske LC and Kreth FW: O-methylguanine-DNA methyltransferase (MGMT) mRNA expression predicts outcome in malignant glioma independent of MGMT promoter methylation. PLoS One. 6:e171562011. View Article : Google Scholar : PubMed/NCBI

11 

Lalezari S, Chou AP, Tran A, Solis OE, Khanlou N, Chen W, Li S, Carrillo JA, Chowdhury R, Selfridge J, et al: Combined analysis of O6-methylguanine-DNA methyltransferase protein expression and promoter methylation provides optimized prognostication of glioblastoma outcome. Neuro Oncol. 15:370–381. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Garcia-Manero G: Demethylating agents in myeloid malignancies. Curr Opin Oncol. 20:705–710. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Li D, Xie Z, Pape ML and Dye T: An evaluation of statistical methods for DNA methylation microarray data analysis. BMC Bioinformatics. 16:2172015. View Article : Google Scholar : PubMed/NCBI

14 

Xu C, Zhang JG, Lin D, Zhang L, Shen H and Deng HW: A systemic analysis of transcriptomic and epigenomic data to reveal regulation patterns for complex disease. G3 (Bethesda). 7:2271–2279. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Smith AA, Huang YT, Eliot M, Houseman EA, Marsit CJ, Wiencke JK and Kelsey KT: A novel approach to the discovery of survival biomarkers in glioblastoma using a joint analysis of DNA methylation and gene expression. Epigenetics. 9:873–883. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Gadaleta F, Bessonov K and Van Steen K: Integration of gene expression and methylation to unravel biological networks in glioblastoma patients. Genet Epidemiol. 41:136–144. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Lai RK, Chen Y, Guan X, Nousome D, Sharma C, Canoll P, Bruce J, Sloan AE, Cortes E, Vonsattel JP, et al: Genome-wide methylation analyses in glioblastoma multiforme. PLoS One. 9:e893762014. View Article : Google Scholar : PubMed/NCBI

18 

Du P, Kibbe WA and Lin SM: Lumi: A pipeline for processing Illumina microarray. Bioinformatics. 24:1547–1548. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Du P, Zhang X, Huang CC, Jafari N, Kibbe WA, Hou L and Lin SM: Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics. 11:5872010. View Article : Google Scholar : PubMed/NCBI

20 

Du P and Bourgon R: methyAnalysis: An R package for DNA methylation data analysis and visualization. 2013.

21 

Harris MA, Clark J, Ireland A, Lomax J, Ashburner M, Foulger R, Eilbeck K, Lewis S, Marshall B, Mungall C, et al: The gene ontology (GO) database and informatics resource. Nucleic Acids Res. 32:D258–D261. 2004. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Joshi-Tope G, Gillespie M, Vastrik I, D'Eustachio P, Schmidt E, de Bono B, Jassal B, Gopinath GR, Wu GR, Matthews L, et al: Reactome: A knowledgebase of biological pathways. Nucleic Acids Res. 33:D428–D432. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Zhao M, Sun J and Zhao Z: TSGene: A web resource for tumor suppressor genes. Nucleic Acids Res. 41:D970–D976. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Chen JS, Hung WS, Chan HH, Tsai SJ and Sun HS: In silico identification of oncogenic potential of fyn-related kinase in hepatocellular carcinoma. Bioinformatics. 29:420–427. 2013. View Article : Google Scholar : PubMed/NCBI

27 

ENCODE Project Consortium: The ENCODE (ENCyclopedia of DNA elements) project. Science. 306:636–640. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Landt SG, Marinov GK, Kundaje A, Kheradpour P, Pauli F, Batzoglou S, Bernstein BE, Bickel P, Brown JB, Cayting P, et al: ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22:1813–1831. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Etcheverry A, Aubry M, de Tayrac M, Vauleon E, Boniface R, Guenot F, Saikali S, Hamlat A, Riffaud L, Menei P, et al: DNA methylation in glioblastoma: Impact on gene expression and clinical outcome. BMC Genomics. 11:7012010. View Article : Google Scholar : PubMed/NCBI

30 

Gentleman R, Carey V, Huber W and Hahne F: Genefilter: Methods for filtering genes from microarray experiments. R Package version 1.66.0. 2011.

31 

Eiró N and Vizoso FJ: Inflammation and cancer. World J Gastrointest Surg. 4:62–72. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Spiegelberg BD and Hamm HE: Roles of G-protein-coupled receptor signaling in cancer biology and gene transcription. Curr Opin Genet Dev. 17:40–44. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Anglim PP, Alonzo TA and Laird-Offringa IA: DNA methylation-based biomarkers for early detection of non-small cell lung cancer: An update. Mol Cancer. 7:812008. View Article : Google Scholar : PubMed/NCBI

35 

Hill VK, Ricketts C, Bieche I, Vacher S, Gentle D, Lewis C, Maher ER and Latif F: Genome-wide DNA methylation profiling of CpG islands in breast cancer identifies novel genes associated with tumorigenicity. Cancer Res. 71:2988–2999. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Shin SH, Kim BH, Jang JJ, Suh KS and Kang GH: Identification of novel methylation markers in hepatocellular carcinoma using a methylation array. J Korean Med Sci. 25:1152–1159. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Knerr I, Schuster S, Nomikos P, Buchfelder M, Dötsch J, Schoof E, Fahlbusch R and Rascher W: Gene expression of adrenomedullin, leptin, their receptors and neuropeptide Y in hormone-secreting and non-functioning pituitary adenomas, meningiomas and malignant intracranial tumours in humans. Neuropathol Appl Neurobiol. 27:215–222. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Kore RA and Abraham EC: Inflammatory cytokines, interleukin-1 beta and tumor necrosis factor-alpha, upregulated in glioblastoma multiforme, raise the levels of CRYAB in exosomes secreted by U373 glioma cells. Biochem Biophys Res Commun. 453:326–331. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Wang L, Qin H, Li L, Feng F, Ji P, Zhang J, Li G, Zhao Z and Gao G: Forkhead-box A1 transcription factor is a novel adverse prognosis marker in human glioma. J Clin Neurosci. 20:654–658. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Hurlock EC, McMahon A and Joho RH: Purkinje-cell-restricted restoration of Kv3.3 function restores complex spikes and rescues motor coordination in Kcnc3 mutants. J Neurosci. 28:4640–4648. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Martinez R, Stuhmer W, Martin S, Schell J, Reichmann A, Rohde V and Pardo L: Analysis of the expression of Kv10.1 potassium channel in patients with brain metastases and glioblastoma multiforme: Impact on survival. BMC Cancer. 15:8392015. View Article : Google Scholar : PubMed/NCBI

42 

Arvind S, Arivazhagan A, Santosh V and Chandramouli BA: Differential expression of a novel voltage gated potassium channel-Kv 1.5 in astrocytomas and its impact on prognosis in glioblastoma. Br J Neurosurg. 26:16–20. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Qi L, Bellail AC, Rossi MR, Zhang Z, Pang H, Hunter S, Cohen C, Moreno CS, Olson JJ, Li S and Hao C: Heterogeneity of primary glioblastoma cells in the expression of caspase-8 and the response to TRAIL-induced apoptosis. Apoptosis. 16:1150–1164. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

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
Kan S, Chai S, Chen W and Yu B: DNA methylation profiling identifies potentially significant epigenetically‑regulated genes in glioblastoma multiforme. Oncol Lett 18: 1679-1688, 2019
APA
Kan, S., Chai, S., Chen, W., & Yu, B. (2019). DNA methylation profiling identifies potentially significant epigenetically‑regulated genes in glioblastoma multiforme. Oncology Letters, 18, 1679-1688. https://doi.org/10.3892/ol.2019.10512
MLA
Kan, S., Chai, S., Chen, W., Yu, B."DNA methylation profiling identifies potentially significant epigenetically‑regulated genes in glioblastoma multiforme". Oncology Letters 18.2 (2019): 1679-1688.
Chicago
Kan, S., Chai, S., Chen, W., Yu, B."DNA methylation profiling identifies potentially significant epigenetically‑regulated genes in glioblastoma multiforme". Oncology Letters 18, no. 2 (2019): 1679-1688. https://doi.org/10.3892/ol.2019.10512