Open Access

Identification of glioblastoma‑specific prognostic biomarkers via an integrative analysis of DNA methylation and gene expression

  • Authors:
    • Yu Kun Mao
    • Zhi Bo Liu
    • Lin Cai
  • View Affiliations

  • Published online on: June 11, 2020     https://doi.org/10.3892/ol.2020.11729
  • Pages: 1619-1628
  • Copyright: © Mao 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 aggressive and lethal tumor of the central nervous system. The present study set out to identify reliable prognostic and predictive biomarkers for patients with GBM. RNA‑sequencing data were obtained from The Cancer Genome Atlas database and DNA methylation data were downloaded using the University of California Santa Cruz‑Xena database. The expression and methylation differences between patients with GBM, and survival times <1 and ≥1 year were investigated. A protein‑protein interaction network was constructed and functional enrichment analyses of differentially expressed and methylated genes were performed. Hub genes were identified using the Cytoscape plug‑in cytoHubba software. Survival analysis was performed using the survminer package, in order to determine the prognostic values of the hub genes. The present study identified 71 genes that were hypomethylated and expressed at high levels, and four genes that were hypermethylated and expressed at low levels in GBM. These genes were predominantly enriched in the ‘JAK‑STAT signaling pathway’, ‘transcriptional misregulation in cancer’ and the ‘ECM‑receptor interaction’, which are associated with GBM development. Among the 24 hub genes identified, 15 possessed potential prognostic value. An integrative analysis approach was implemented in order to analyze the association of DNA methylation with changes in gene expression and to assess the association of gene expression changes with GBM survival time. The results of the present study suggest that these 15 CpG‑based genes may be useful and practical tools in predicting the prognosis of patients with GBM. However, future research on gene methylation and/or expression is required in order to develop personalized treatments for patients with GBM.

Introduction

GBM is the most aggressive form of brain tumor, with high recurrence (~37%) and mortality rates (median survival time <1 year) in 2018 (1,2). GBM originates from astrocytes and is comprised of different types of cell (3), which multiply rapidly and support GBM development via large vascular networks (4). The current standard treatment for GBM includes surgery, followed by radiation therapy and temozolomide chemotherapy (5). However, the median survival time for this procedure is <1 year and very few patients survive >3 years. Despite great efforts, little progress has been made in the treatment of GBM over the past decade (6,7). Furthermore, a series of phase III clinical trials for targeted drugs has failed to improve the overall survival of patients with GBM (8). Thus, it is critical to investigate potential biomarkers of GBM and provide novel insights into the diagnosis and treatment of the disease.

Changes in the expression of biomarkers are commonly described in oncological research. Biomarkers provide a framework and assurance for the strategy of diagnostic and therapeutic methods, with the aim of eradicating complications. Previous studies have identified prognostic biomarkers for GBM, such as long coding RNAs, circular RNAs, microRNAs and somatic mutations (912). In the past decade, epigenetic modification, especially DNA methylation in carcinogenesis, has become a focus for cancer research. Several methylated markers have been reported. For example, O6-methylguanine DNA methyltransferase promoter methylation is the major emblematic biomarker in GBM, which may predict the response to temozolomide treatment (1315). Epigenetic changes are heritable and reversible, affecting the spatial conformation of DNA and its transcriptional activities (16). DNA methylation is one of the main epigenetic mechanisms of gene expression regulation in eukaryotes (17). Changes in DNA methylation may affect gene expression and feedback mechanisms, with various positive and negative interactions (18). Thus, abnormally methylated CpG sites are considered potential prognostic factors for GBM.

As the majority of previous studies were performed on relatively small sample sizes and limited to a single epigenetic level, the present study integrated 152 GBM RNA-sequencing (RNA-seq) samples and 151 GBM DNA methylation samples from public databases. Bioinformatics-based methods were implemented to identify novel markers of phenotypically important associations among DNA methylation, gene expression and survival time in patients with GBM, using The Cancer Genome Atlas (TCGA) datasets.

Materials and methods

Data preparation

RNA-seq data of 174 GBM samples were downloaded from the TCGA (https://portal.gdc.cancer.gov/) database in October, 2018 according to ‘glioblastoma’ search term. Repeat samples, non-cancerous samples, paracancerous samples and samples without follow-up information were removed using gdcRNA tools (version 4.6.3) (19). Thus, a total of 152 GBM samples were included in the present study. RNA-seq data were normalized using the trimmed mean of M-values method (20).

DNA methylation data of 155 GBM samples were downloaded from the University of California Santa Cruz (UCSC)-Xena database (https://xenabrowser.net/datapages/) in October, 2018 according to ‘glioblastoma’ search term. UCSC-Xena is a bioinformatics tool used to visualize functional genomics data from several sources, including TCGA database. DNA methylation was represented as β values, which are bounded variables of the form [M/(M + U + 100)] that are generated by the Illumina 450 k BeadChip array (Illumina, Inc.) (21). Samples without follow-up information were removed, thus a total of 151 samples were used in the present study.

The DNA methylation dataset (151 patients) and the RNA-seq dataset (152 patients) were compared, and 49 patients were found to be shared between the two datasets. The research flow chart is presented in Fig. S1.

Identification of differentially expressed genes (DEGs) according to survival time

Gene expression data were used for the differential expression analysis. The median survival time of patients with GBM is <1 year (1). Thus, the patients (n=152) were divided into two groups, survival time <1 year and survival time ≥1 year, according to the median survival time. Differential expression analysis was performed between the two groups using the edgeR package (version 3.8) (22). P<0.05 and |logFC|>0.585 were considered to indicate DEGs.

Identification of differentially methylated genes

Methylation data were used for the differential methylation analysis. The patients (n=151) were divided into two groups, survival time <1 year and survival time ≥1 year, according to the median survival time. Differentially methylated CpG sites were identified by comparing the two groups. CpG sites with ≥50% missing values were removed and CpG sites with <50% missing values were filled using K-means (23). The remaining missing values were imputed using the ChAMP package (version 3.8) (24). Low-quality probes were removed if they met the following criteria: Probes with non-CpG sites, probes associated with single-nucleotide polymorphisms, cross-reactive probes and probes on the X and Y chromosomes. The β-Mixture Quantile Normalization method was used for further type I and II probe correction (25,26). P<0.01 was considered to indicate differentially methylated CpG sites.

Identification of differentially expressed and methylated genes

DNA methylation analysis focused on CpG sites that were located simultaneously in differentially methylated sites and differentially expressed genes. Thus, the differentially expressed genes and the differentially methylated genes were integrated.

Protein-protein (PPI) network construction

The Search Tool for the Retrieval of interacting Genes/Proteins (STRING) database (http://string-db.org) is used to identify associations between known proteins and predicted proteins (27), and was used in the pwresent study to construct a PPI network. The PPI network was visualized using Cytoscape software (version 3.8.0) (28). Each node represents a gene, protein or molecule. Furthermore, the connections between nodes represent the interactions of these biological molecules, which can be used to identify interactions between the proteins encoded by DEGs and methylated genes in GBM. The corresponding protein in the central node can be a core protein or a key candidate gene with critical physiological regulatory functions.

Gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses

Functional enrichment analyses were performed to determine potential biological processes and pathways enriched by the DEGs and methylated genes in GBM. KEGG pathway enrichment analysis of the differentially expressed and methylated genes was performed using the clusterProfiler package (version 3.8) (29,30), and GO enrichment analysis was performed using Cytoscape plug-in ClueGO (version 2.5.3), which visualizes the non-redundant biological terms for large clusters of genes in a functionally grouped network (31). The cut-off value for significant terms was set as P<0.05.

Identification of hub genes

Cytoscape plug-in cytoHubba (version 1.6) is commonly used to identify hub genes and subnetworks from complex interactomes (32). The four local-based methods; Density of maximum neighborhood component (DMNC), MNC, maximal clique centrality (MCC) and degree, were used to predict and evaluate essential proteins according to ranking nodes in the network. The top 30 genes within the four methods were screened and overlapped, and the overlapping genes were considered to be hub genes.

Survival analysis

The expression values of the hub genes were visualized in box plots and heat maps. Survival analysis was performed using the survminer package (version 0.4.6; http://cran.r-project.org/) and the cut-off values were identified according to the surv_cutpoint function. Survival times were evaluated using Kaplan-Meier survival curves and differences were analyzed using the log-rank test. P<0.05 was considered to indicate a statistically significant difference. Subsequently, Cox proportional hazards regression analysis was performed to further assess the results of Kaplan-Meier survival analysis.

Results

Identification of DEGs according to survival time

A total of 152 GBM gene expression profiles were obtained from the UCSC-Xena database. The patients were divided into 2 groups; survival time <1 year (short group) and survival time ≥1 year (long group) according to the median survival time. A total of 429 downregulated genes and 672 upregulated genes were identified, according to survival time, with P<0.05 and |logFC|>0.585. The results were visualized as hierarchical clusters and volcano plots, as presented in Fig. 1.

Identification of differentially methylated genes

A total of 151 GBM DNA methylation profiles were obtained from TCGA database. The clinical characteristics of patients with GBM in the TCGA database are presented in Table I. A total of 4,079 differentially methylated CpG sites were identified, including 509 hypermethylated sites and 3,570 hypomethylated sites (P<0.01; Fig. S2A). The present study screened the top 50 methylation sites in descending order of the absolute value of logFC, as presented in Fig. S2B. Following annotation of the differentially methylated CpG sites, 370 hypermethylated genes and 1,987 hypomethylated genes were identified.

Table I.

Clinical characteristics of patients with glioblastoma.

Table I.

Clinical characteristics of patients with glioblastoma.

CharacteristicTCGA (n=152)UCSC-Xena (n=151)
Age, years
  Median  61  60
  Range21-8921-85
Sex
  Male  98  88
  Female  54  63
Ethnicity, n
  Asian  5  0
  Black or African American  10  24
  White136118
  N/A  1  9
Vital status, n
  Living  30  45
  Dead122106

[i] TCGA, The Cancer Genome Atlas; UCSC-Xena, University of California Santa Cruz-Xena.

Identification of differentially expressed and methylated genes

DNA methylation may downregulate gene expression. Thus, the present study examined the intersection of the differentially expressed and methylated genes. A total of 49 genes from 152 patients overlapped the DNA methylation array data from 151 patients. The clinical characteristics of patients with GBM from TCGA dataset are presented in Table I. A total of 71 genes that were hypomethylated and expressed at high levels, and four genes that were hypermethylated and expressed at low levels in GBM were identified.

PPI network construction and functional enrichment analyses

Within the STRING database, the expression products of the differentially expressed and methylated genes in GBM were used to construct the PPI network (Fig. 2A), with a total of 63 genes (confidence score, >0.15). KEGG pathway enrichment analysis demonstrated that these genes were predominantly enriched in the ‘JAK-STAT signaling pathway’, ‘protein digestion and absorption’, ‘steroid hormone biosynthesis’, ‘cytokine-cytokine receptor interaction’, ‘prolactin signaling pathway’, ‘transcriptional misregulation in cancer’ and ‘ECM-receptor interaction’ (Fig. 2B). GO enrichment analysis demonstrated that these genes were predominantly enriched in ‘cellular response to corticosteroid stimulus’, ‘cell fate determination’, ‘collagen fibril organization’ and ‘collagen biosynthetic process’ (Fig. 2C), which may contribute to GBM development.

Hub genes and survival analysis

The top 30 genes were screened by combining the four local-based methods (DMNC, MNC, MCC and degree) in the Cytoscape plug-in cytoHubba based on differentially expressed and methylated genes. The 24 overlapping genes were considered hub genes (Fig. 3). Subsequently, the expression values of the 24 hub genes were visualized as box plots, as presented in Figs. 4 and S3. These hub genes had significantly differential expression in patients who had a survival time ≥1 year (long group) compared with patients who had a survival time <1 year (short group) (P<0.05). Survival analysis was performed in order to further assess the prognostic values of the hub genes. Among the 24 hub genes, 15 possessed potential prognostic value by comparing patients who experienced longer survival times with patients who experienced shorter survival times. Patients with hypomethylated genes expressed at high levels had shorter survival times compared with patients expressing hypermethylated genes at low levels, including COL1A1 (P=0.0280), CYP1B1 (P=0.0370), CYR61 (P=0.0087), ELANE (P=0.0051), FAM20C (P=0.0019), HAL (P=0.0160), HIC1 (P=0.0210), KRT19 (P=0.0150), LIF (P=0.0097), LOX (P=0.0180), PAX3 (P=0.0110), SFRP2 (P=0.0110) and SOCS3 (P=0.0200) (Fig. 5). Furthermore, patients with high expression levels of the hypomethylated COMP gene had longer survival times compared with patients with low levels of the hypermethylated COMP gene (P=0.0400). In addition, patients with low levels of the hypermethylated DLL1 gene had shorter survival times compared with patients with high expression levels of the hypomethylated DLL1 gene (P=0.0047). The details of the 15 hub genes are presented in Table II. Cox proportional hazards regression analysis was performed to evaluate Kaplan-Meier survival analysis results, which are presented in Table III.

Table II.

Hub genes (n=15) in glioblastoma.

Table II.

Hub genes (n=15) in glioblastoma.

SymbolGroupLogFCLogCPMP-valueFDRChangeMethylation
COL1A1protein_coding0.9483499.177457 2.369×10−30.116097UP Hypomethylation
CYP1B1protein_coding0.9050224.648310 7.350×10−40.053914UP Hypomethylation
CYR61protein_coding0.8577877.004545 4.660×10−50.007693UP Hypomethylation
ELANEprotein_coding0.820009−0.896520 5.280×10−40.043721UP Hypomethylation
FAM20Cprotein_coding0.6458447.793972 1.160×10−40.015224UP Hypomethylation
HALprotein_coding0.860970−1.280750 5.460×10−50.008582UP Hypomethylation
HIC1protein_coding0.6203052.999093 5.230×10−40.043431UP Hypomethylation
KRT19protein_coding0.967123−1.356580 2.650×10−40.027310UP Hypomethylation
LIFprotein_coding0.8786224.793528 1.020×10−30.066857UP Hypomethylation
LOXprotein_coding0.6355355.807618 5.588×10−30.190609UP Hypomethylation
PAX3protein_coding0.7045071.890802 3.279×10−20.430150UP Hypomethylation
SFRP2protein_coding1.3506523.849805 6.910×10−50.010316UP Hypomethylation
SOCS3protein_coding0.7593826.741612 1.930×10−40.021922UP Hypomethylation
COMPprotein_coding3.3520020.442354 9.550×10−145.78E-10UP Hypomethylation
DLL1protein_coding−1.8064205.384099 2.780×10−126.22E-09DOWN Hypermethylation

[i] LogFC, log-fold change; LogCPM, log-counts per million; FDR, false discovery rate.

Table III.

Cox proportional-hazards regression model analysis of the 15 hub genes.

Table III.

Cox proportional-hazards regression model analysis of the 15 hub genes.

Hub geneHRCIP-value
COMP0.9720.848–1.1150.688
DLL10.8600.703–1.0530.144
CYR610.9780.776–1.2340.853
HAL1.0320.873–1.2190.715
SFRP20.9950.866–1.1430.947
FAM20C0.9300.676–1.2790.656
SOCS31.3080.975–1.7540.073
KRT191.0030.868–1.1590.967
HIC11.0850.784–1.5030.622
ELANE1.0280.884–1.1960.722
CYP1B10.9710.752–1.2530.819
LIF0.8780.717–1.0740.206
COL1A10.9280.712–1.2100.580
LOX1.0000.789–1.2680.999
PAX30.9010.821–0.9890.029

[i] HR, hazard ratio; CI, confidence interval.

Functional enrichment analysis of the 15 hub genes

Functional enrichment analyses were performed to determine the potential biological processes and signaling pathways enriched by the differentially expressed and methylated genes in GBM. KEGG pathway analysis demonstrated that the hub genes were predominantly enriched in the ‘JAK-STAT signaling pathway’, ‘transcriptional misregulation in cancer’, ‘focal adhesion’ and ‘PI3K-Akt signaling pathway’ (Fig. 6A). These pathways have been confirmed to be involved in GBM development and progression. GO enrichment analysis demonstrated that these hub genes were predominantly enriched in ‘skeletal system development’, ‘ossification’ and ‘striated muscle cell differentiation’ (Fig. 6B).

Discussion

GBM is a brain tumor associated with a poor prognosis. Despite advancements made in the treatment of GBM, the median survival time remains <1 year, and few patients survive >3 years (33,34). In the present study, patients from TCGA database were divided into two groups, survival time <1 year and survival time ≥1 year, according to the median survival time. Gene expression and DNA methylation were compared between the genes in both groups. DNA methylation has been demonstrated to be associated with survival outcome via changes in gene expression (35). A previous study of GBM used a joint analysis method of DNA methylation and changes in gene expression to identify prognostic biomarkers for patients with GBM (36). The hypermethylation or hypomethylation of DEGs has a major impact on the development and progression of different types of tumor, either acting as oncogenes or anticancer genes. The current study successfully presented a number of genes that were significantly modulated by DNA methylation.

In the present study, KEGG pathway enrichment analysis demonstrated that the differentially expressed and methylated genes were predominantly enriched in the ‘JAK-STAT signaling pathway’. Along with the ‘JAK-STAT signaling pathway’, ‘transcriptional misregulation in cancer’ and ‘ECM-receptor interaction’ have been confirmed to serve a critical role in GBM development (3741). GO enrichment analysis demonstrated that these genes were predominantly enriched in ‘cell fate determination’, ‘collagen fibril organization’ and ‘collagen biosynthetic process’, which may contribute to GBM development (4245).

Cancer involves a complex regulatory network and the integration of multiple biomarkers into an aggregation model can improve the prognostic value compared with a single biomarker (46). For example, the expression of transmembrane protein 41A is associated with metastasis by modulating E-cadherin in radically resected gastric cancer (47). In the present study, 24 hub genes were identified by overlapping the top 30 genes within four local-based methods (DMNC, MNC, MCC and degree), in the Cytoscape plug-in cytoHubba. Among the 24 hub genes, 15 possessed potential prognostic value between patients who experienced longer survival times and patients who experienced shorter survival times. The hypomethylated genes expressed at high levels, including COL1A1, CYP1B1, CYR61, ELANE, FAM20C, HAL, HIC1, KRT19, LIF, LOX, PAX3, SFRP2 and SOCS3, were associated with a poor prognosis of GBM. Furthermore, the hypermethylated DLL1 gene and hypomethylated COMP gene, expressed at low levels, were demonstrated to have worse clinical outcomes. Among the 15 hub genes corresponding with candidate CpG sites, 7 have been reported as GBM-associated genes. For example, COL1A1 has the potential to stratify patients with GBM into subgroups, to determine the risk of recurrence (48). Overexpression of CYR61 has been reported to enhance the tumorigenicity of glioma cells and stimulate the β-catenin-TCF/Lef and Akt signaling pathways via the activation of integrin-linked kinase (49,50). Furthermore, LIF possesses a self-renewal capacity that prevents the differentiation of glioma-initiating cells (51). As an evolutionarily conserved metabolic gene, LOX is involved in gliomagenesis (52). PAX3 is overexpressed in GBM and strictly regulates the tumorigenicity of glioma cells (53). The transcriptional inhibition of p53 by PAX3 may contribute to glioma formation and the differentiation of glioma stem cells (54). The hypermethylation of SFRP2 occurs in >40% of primary GBMs (55,56). A previous study demonstrated that patients with hypermethylated SOCS3 were associated with a significantly poor prognosis compared with healthy controls (57). However, the results of the present study demonstrated that patients with hypomethylated SOCS3 at low levels had a shorter survival time compared with patients with hypermethylated SOCS3 at high levels Thus, further research is required in order to clarify these inconsistencies.

Overall, the present study identified 15 prognostic biomarkers for GBM based on DNA methylation and mRNA expression levels, including COL1A1, CYP1B1, CYR61, ELANE, FAM20C, HAL, HIC1, KRT19, LIF, LOX, PAX3, SFRP2, SOCS3, COMP and DLL1. The results of the current study demonstrated that these genes are regulated by aberrant methylation, which could serve a crucial role in GBM. These genes have the potential to serve as candidate prognostic biomarkers and therapeutic targets in GBM; however, the results require validation within larger cohorts to evaluate their potential as biomarkers in GBM. Furthermore, future studies are required to determine the molecular mechanisms underlying these genes.

The present study implemented an integrative analysis approach to analyze DNA methylation with changes in gene expression and assess the association of gene expression changes with GBM survival time. A total of 15 CpG-based genes were identified to possess potential value to predict the prognosis of patients with GBM. However, future studies are required on these gene methylation and/or gene expression biomarkers to develop personalized treatments for patients with GBM.

There are a number of limitations associated with the present study. GBM isocitrate dehydrogenase (IDH)-wild-type is the most common astrocytic glioma (58). Thus, existing prognostic factors for this type of tumor should commonly be used for comparative analysis of novel prognostic factors for GBM. However, there are currently no available data within TCGA cohorts. Thus, the present study lacks comparative analyses between the status of IDH-1/2 genes and the 15 CpG-based genes. Furthermore, although GBM-specific prognostic biomarkers were identified via integrative analysis of DNA methylation and gene expression, the present study lacks validation by functional studies both in vitro and in vivo.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was funded by The Health Care of Yellow Crane Talent Plan (grant no. 17).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the TCGA repository (https://portal.gdc.cancer.gov/).

Authors' contributions

LC, YM and ZL designed the present study and reviewed the manuscript. ZL acquired the data, YM interpreted the data, and YM and ZL analyzed the data. YM drafted the initial 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 interest.

Glossary

Abbreviations

Abbreviations:

GBM

glioblastoma

TCGA

The Cancer Genome Atlas

PPI

protein-protein interaction

KEGG

Kyoto Encyclopedia of Genes and Genomes

GO

Gene Ontology

MNC

maximum neighborhood component

DMNC

density of MNC

MCC

maximal clique centrality

References

1 

Jackson CM, Choi J and Lim M: Mechanisms of immunotherapy resistance: Lessons from glioblastoma. Nat Immunol. 20:1100–1109. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Lu VM, Jue TR, McDonald KL and Rovin RA: The survival effect of repeat surgery at glioblastoma recurrence and its trend: A systematic review and meta-analysis. World Neurosurg. 115:453–459. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Brandao M, Simon T, Critchley G and Giamas G: Astrocytes, the rising stars of the glioblastoma microenvironment. Glia. 67:779–790. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Saadatpour L, Fadaee E, Fadaei S, Mansour RN, Mohammadi M, Mousavi SM, Goodarzi M, Verdi J and Mirzaei H: Glioblastoma: Exosome and microRNA as novel diagnosis biomarkers. Cancer Gene Ther. 23:415–418. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Brandes AA, Franceschi E, Paccapelo A, Tallini G, Biase DD, Ghimenton C, Danieli D, Zunarelli E, Lanza G, Silini EM, et al: Role of MGMT methylation status at time of diagnosis and recurrence for patients with glioblastoma: Clinical implications. Oncologist. 22:432–437. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Jhanwar-Uniyal M, Amin AG, Cooper JB, Das K, Schmidt MH and Murali R: Discrete signaling mechanisms of mTORC1 and mTORC2: Connected yet apart in cellular and molecular aspects. Adv Biol Regul. 64:39–48. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Jhanwar-Uniyal M, Wainwright JV, Mohan AL, Tobias ME, Murali R, Gandhi CD and Schmidt MH: Diverse signaling mechanisms of mTOR complexes: MTORC1 and mTORC2 in forming a formidable relationship. Adv Biol Regul. 72:51–62. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Klughammer J, Kiesel B, Roetzer T, Fortelny N, Nemc A, Nenning KH, Furtner J, Sheffield NC, Datlinger P, Peter N, et al: The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat Med. 24:1611–1624. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Tan SK, Pastori C, Penas C, Komotar RJ, Ivan ME, Wahlestedt C and Ayad NG: Serum long noncoding RNA HOTAIR as a novel diagnostic and prognostic biomarker in glioblastoma multiforme. Mol Cancer. 17:742018. View Article : Google Scholar : PubMed/NCBI

10 

Li X and Diao H: Circular RNA circ_0001946 acts as a competing endogenous RNA to inhibit glioblastoma progression by modulating miR-671-5p and CDR1. J Cell Physiol. 234:13807–13819. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Zhao H, Shen J, Hodges TR, Song R, Fuller GN and Heimberger AB: Serum microRNA profiling in patients with glioblastoma: A survival analysis. Mol Cancer. 16:592017. View Article : Google Scholar : PubMed/NCBI

12 

Shajani-Yi Z, de Abreu FB, Peterson JD and Tsongalis GJ: Frequency of somatic TP53 mutations in combination with known pathogenic mutations in colon adenocarcinoma, non-small cell lung carcinoma, and gliomas as identified by next-generation sequencing. Neoplasia. 20:256–262. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Chen X, Zhang M, Gan H, Wang H, Lee JH, Fang D, Kitange GJ, He L, Hu Z, Parney IF, et al: A novel enhancer regulates MGMT expression and promotes temozolomide resistance in glioblastoma. Nat Commun. 9:29492018. View Article : Google Scholar : PubMed/NCBI

14 

Kessler T, Sahm F, Sadik A, Stichel D, Hertenstein A, Reifenberger G, Zacher A, Sabel M, Tabatabai G, Steinbach J, et al: Molecular differences in IDH wildtype glioblastoma according to MGMT promoter methylation. Neuro Oncol. 20:367–379. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Cloughesy T, Finocchiaro G, Belda-Iniesta C, Recht L, Brandes AA, Pineda E, Mikkelsen T, Chinot OL, Balana C, Macdonald DR, et al: Randomized, double-blind, placebo-controlled, multicenter phase II study of onartuzumab plus bevacizumab versus placebo plus bevacizumab in patients with recurrent glioblastoma: Efficacy, safety, and hepatocyte growth factor and O6-Methylguanine-DNA methyltransferase biomarker analyses. J Clin Oncol. 35:343–351. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Arantes LM, de Carvalho AC, Melendez ME, Carvalho AL and Goloni-Bertollo EM: Methylation as a biomarker for head and neck cancer. Oral Oncol. 50:587–592. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Anastasiadi D, Esteve-Codina A and Piferrer F: Consistent inverse correlation between DNA methylation of the first intron and gene expression across tissues and species. Epigenetics Chromatin. 11:372018. View Article : Google Scholar : PubMed/NCBI

18 

Portela A and Esteller M: Epigenetic modifications and human disease. Nat Biotechnol. 28:1057–1068. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Li R, Qu H, Wang S, Wei J, Zhang L, Ma R, Lu J, Zhu J, Zhong WD and Jia Z: GDCRNATools: An R/bioconductor package for integrative analysis of lncRNA, miRNA and mRNA data in GDC. Bioinformatics. 34:2515–2517. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Robinson MD and Oshlack A: A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11:R252010. View Article : Google Scholar : PubMed/NCBI

21 

Weinhold L, Wahl S, Pechlivanis S, Hoffmann P and Schmid M: A statistical model for the analysis of beta values in DNA methylation studies. BMC Bioinformatics. 17:4802016. View Article : Google Scholar : PubMed/NCBI

22 

Robinson MD, McCarthy DJ and Smyth GK: EdgeR: A bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Tseng GC: Penalized and weighted K-means for clustering with scattered objects and prior information in high-throughput biological data. Bioinformatics. 23:2247–2255. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Morris TJ, Butcher LM, Feber A, Teschendorff AE, Chakravarthy AR, Wojdacz TK and Beck S: ChAMP: 450 k chip analysis methylation pipeline. Bioinformatics. 30:428–430. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Chen YA, Lemire M, Choufani S, Butcher DT, Grafodatskaya D, Zanke BW, Gallinger S, Hudson TJ and Weksberg R: Discovery of cross-reactive probes and polymorphic CpGs in the illumina infinium humanmethylation450 microarray. Epigenetics. 8:203–209. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Teschendorff AE, Marabita F, Lechner M, Bartlett T, Tegner J, Cabrero DG and Beck S: A beta-mixture quantile normalization method for correcting probe design bias in illumina infinium 450 k DNA methylation data. Bioinformatics. 29:189–196. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, et al: The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45(D1): D362–D368. 2017. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pagès F, Trajanoski Z and Galon J: ClueGO: A cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 25:1091–1093. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Chin CH, Chen SH, Wu HH, Ho CW, Ko MT and Lin CY: cytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 8 (Suppl 4):S112014. View Article : Google Scholar : PubMed/NCBI

33 

Woehrer A, Bauchet L and Barnholtz-Sloan JS: Glioblastoma survival: Has it improved? Evidence from population-based studies. Curr Opin Neurol. 27:666–674. 2014.PubMed/NCBI

34 

Ramos AR, Elong Edimo W and Erneux C: Phosphoinositide 5-phosphatase activities control cell motility in glioblastoma: Two phosphoinositides PI(4,5)P2 and PI(3,4)P2 are involved. Adv Biol Regul. 67:40–48. 2018. View Article : Google Scholar : PubMed/NCBI

35 

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

36 

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

37 

Senft C, Priester M, Polacin M, Schröder K, Seifert V, Kögel D and Weissenberger J: Inhibition of the JAK-2/STAT3 signaling pathway impedes the migratory and invasive potential of human glioblastoma cells. J Neurooncol. 101:393–403. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Mukthavaram R, Ouyang X, Saklecha R, Jiang P, Nomura N, Pingle SC, Guo F, Makale M and Kesari S: Effect of the JAK2/STAT3 inhibitor SAR317461 on human glioblastoma tumorspheres. J Transl Med. 13:2692015. View Article : Google Scholar : PubMed/NCBI

39 

Park AK, Kim P, Ballester LY, Esquenazi Y and Zhao Z: Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma. Neuro Oncol. 21:59–70. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Nawaz Z, Patil V, Paul Y, Hegde AS, Arivazhagan A, Santosh V and Somasundaram K: PI3 kinase pathway regulated miRNome in glioblastoma: Identification of miR-326 as a tumour suppressor miRNA. Mol Cancer. 15:742016. View Article : Google Scholar : PubMed/NCBI

41 

Agrawal R, Garg A, Benny Malgulwar P, Sharma V, Sarkar C and Kulshreshtha R: P53 and miR-210 regulated NeuroD2, a neuronal basic helix-loop-helix transcription factor, is downregulated in glioblastoma patients and functions as a tumor suppressor under hypoxic microenvironment. Int J Cancer. 142:1817–1828. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Park NI, Guilhamon P, Desai K, McAdam RF, Langille E, O'Connor M, Lan X, Whetstone H, Coutinho FJ, Vanner RJ, et al: ASCL1 reorganizes chromatin to direct neuronal fate and suppress tumorigenicity of glioblastoma stem cells. Cell Stem Cell. 21:209–224. 2017. View Article : Google Scholar : PubMed/NCBI

43 

John S, Sivakumar KC and Mishra R: Extracellular proton concentrations impacts ln229 glioblastoma tumor cell fate via differential modulation of surface lipids. Front Oncol. 7:202017. View Article : Google Scholar : PubMed/NCBI

44 

Mammoto T, Jiang A, Jiang E, Panigrahy D, Kieran MW and Mammoto A: Role of collagen matrix in tumor angiogenesis and glioblastoma multiforme progression. Am J Pathol. 183:1293–1305. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Liu J, Li W, Liu S, Zheng X, Shi L, Zhang W and Yang H: Knockdown of collagen triple helix repeat containing 1 (CTHRC1) inhibits epithelial-mesenchymal transition and cellular migration in glioblastoma cells. Oncol Res. 25:225–232. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Ng SW, Mitchell A, Kennedy JA, Chen WC, McLeod J, Ibrahimova N, Arruda A, Popescu A, Gupta V, Schimme AD, et al: A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature. 540:433–437. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Lin B, Xue Y, Qi C, Chen X and Mao W: Expression of transmembrane protein 41A is associated with metastasis via the modulation of E-cadherin in radically resected gastric cancer. Mol Med Rep. 18:2963–2972. 2018.PubMed/NCBI

48 

Balbous A, Cortes U, Guilloteau K, Villalva C, Flamant S, Gaillard A, Milin S, Wager M, Sorel N, Guilhot J, et al: A mesenchymal glioma stem cell profile is related to clinical outcome. Oncogenesis. 3:e912014. View Article : Google Scholar : PubMed/NCBI

49 

Xie D, Yin D, Tong X, O'Kelly J, Mori A, Miller C, Black K, Gui D, Said JW and Koeffler HP: Cyr61 is overexpressed in gliomas and involved in integrin-linked kinase-mediated akt and beta-catenin-TCF/Lef signaling pathways. Cancer Res. 64:1987–1996. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Jeansonne D, Pacifici M, Lassak A, Reiss K, Russo G, Zabaleta J and Peruzzi F: Differential effects of microRNAs on glioblastoma growth and migration. Genes (Basel). 4:46–64. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Peñuelas S, Anido J, Prieto-Sánchez RM, Folch G, Barba I, Cuartas I, Dorado DG, Poca MA, Sahuquillo J, Baselga J and Seoane J: TGF-beta increases glioma-initiating cell self-renewal through the induction of LIF in human glioblastoma. Cancer Cell. 15:315–327. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Chi KC, Tsai WC, Wu CL, Lin TY and Hueng DY: An adult drosophila glioma model for studying pathometabolic pathways of gliomagenesis. Mol Neurobiol. 56:4589–4599. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Xia L, Huang Q, Nie D, Shi J, Gong M, Wu B, Gong P, Zha L, Zuo H, Ju S, et al: PAX3 is overexpressed in human glioblastomas and critically regulates the tumorigenicity of glioma cells. Brain Res. 1521:68–78. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Zhu H, Wang H, Huang Q, Liu Q, Guo Y, Lu J, Li X, Xue C and Han Q: Transcriptional repression of p53 by PAX3 contributes to gliomagenesis and differentiation of glioma stem cells. Front Mol Neurosci. 11:1872018. View Article : Google Scholar : PubMed/NCBI

55 

Götze S, Wolter M, Reifenberger G, Müller O and Sievers S: Frequent promoter hypermethylation of Wnt pathway inhibitor genes in malignant astrocytic gliomas. Int J Cancer. 126:2584–2593. 2010.PubMed/NCBI

56 

Stricker SH, Feber A, Engström PG, Carén H, Kurian KM, Takashima Y, Watts C, Way M, Dirks P, Bertone P, et al: Widespread resetting of DNA methylation in glioblastoma-initiating cells suppresses malignant cellular behavior in a lineage-dependent manner. Genes Dev. 27:654–669. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Martini M, Pallini R, Luongo G, Cenci T, Lucantoni C and Larocca LM: Prognostic relevance of SOCS3 hypermethylation in patients with glioblastoma multiforme. Int J Cancer. 123:2955–2960. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Wirsching HG, Galanis E and Weller M: Glioblastoma. Handb Clin Neurol. 134:381–397. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 20 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
Mao YK, Liu ZB and Cai L: Identification of glioblastoma‑specific prognostic biomarkers via an integrative analysis of DNA methylation and gene expression. Oncol Lett 20: 1619-1628, 2020
APA
Mao, Y.K., Liu, Z.B., & Cai, L. (2020). Identification of glioblastoma‑specific prognostic biomarkers via an integrative analysis of DNA methylation and gene expression. Oncology Letters, 20, 1619-1628. https://doi.org/10.3892/ol.2020.11729
MLA
Mao, Y. K., Liu, Z. B., Cai, L."Identification of glioblastoma‑specific prognostic biomarkers via an integrative analysis of DNA methylation and gene expression". Oncology Letters 20.2 (2020): 1619-1628.
Chicago
Mao, Y. K., Liu, Z. B., Cai, L."Identification of glioblastoma‑specific prognostic biomarkers via an integrative analysis of DNA methylation and gene expression". Oncology Letters 20, no. 2 (2020): 1619-1628. https://doi.org/10.3892/ol.2020.11729