Open Access

DNA hypermethylation of aurora kinase A in hepatitis C virus‑positive hepatocellular carcinoma

  • Authors:
    • Zuohong Ma
    • Yefu Liu
    • Zhiqiang Hao
    • Xiangdong Hua
    • Wenxin Li
  • View Affiliations

  • Published online on: July 11, 2019     https://doi.org/10.3892/mmr.2019.10487
  • Pages: 2519-2532
  • Copyright: © Ma 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

Changes in the methylation levels of tumor suppressor genes or proto‑oncogenes are involved in the pathogenesis of hepatitis C virus (HCV) infection‑induced hepatocellular carcinoma (HCC). The aim of the present study was to identify novel aberrantly methylated differentially expressed genes by integrating mRNA expression profile (GSE19665 and GSE62232) and methylation profile (GSE60753) microarrays downloaded from the Gene Expression Omnibus database. Functional enrichment analysis of screened genes was performed using the DAVID software and BinGO database. Protein‑protein interaction (PPI) networks were constructed using the STRING database, followed by module analysis with MCODE software. The transcriptional and translational expression levels of crucial genes were confirmed using The Cancer Genome Atlas (TCGA) datasets and Human Protein Atlas database (HPA). A total of 122 downregulated/hypermethylated genes and 63 upregulated/hypomethylated genes were identified. These genes were enriched in the Gene Ontology biological processes terms of ‘inflammatory response’ [Fos proto‑oncogene, AP‑1 transcription factor subunit (FOS)] and ‘cell cycle process’ [aurora kinase A (AURKA), cyclin dependent kinase inhibitor 3 (CDKN3) and ubiquitin conjugating enzyme E2 C (UBE2C)]. PPI network and module analysis indicated that human oncogenes FOS, AURKA, CDKN3 and UBE2C may be hub genes. mRNA, protein expression and methylation levels of AURKA and FOS were validated by TCGA and HPA data. In conclusion, aberrantly methylated AURKA and FOS may be potential therapeutic targets for HCV‑positive HCC.

Introduction

Hepatocellular carcinoma (HCC) is a common malignancy and the leading cause of cancer-related mortality, with an estimated 42,220 new cases diagnosed and 30,200 mortalities in the United States in 2018 (1). Despite curative surgical resection and recent advances in adjuvant chemotherapy, radiotherapy and liver transplantation, recurrence and metastasis occur frequently, leading to the overall 5-year survival rate <20% (2). Although multiple factors have been linked to the development and progression of HCC, hepatitis virus infection is considered to be the predominant underlying cause. It has been reported that the burden of HCC parallels the prevalence of hepatitis C virus (HCV) (3). The 10-year survival rate was reported to be approximately 35% in HCC patients with HCV (4). Thus, it is necessary to explore the molecular mechanisms of HCV-associated hepatocarcinogenesis to screen novel prognostic biomarkers and to develop effective therapeutic strategies.

Although the mechanism of the pathogenesis by which HCV induces HCC is currently unclear, epigenetic changes (such as DNA methylation) have been demonstrated to serve fundamental roles. For example, aberrant hypermethylation of tumor suppressor genes or hypomethylation of proto-oncogenes may result in the decrease or increase in their expression levels and induce excessive proliferation, migration and invasion of hepatocytes (5). Methylation of several genes has been reported in HCV-associated HCC (6). For example, Ramadan et al demonstrated that the frequency of aberrant methylation in the promoter region of serine protease inhibitor kunitz-type 2 gene was significantly higher in HCV-positive HCC cases compared with HCV-positive cirrhosis and normal control patients (7). Takagi et al reported that CpG islands in zygote arrest 1 exon 1 had a higher methylation level in HCV-positive HCC compared with non-tumorous tissues (8). Tsunedomi et al not only demonstrated a correlation between DNA methylation and mRNA expression levels of ATP-binding cassette subfamily B member 6 (ABCB6), but also revealed that aberrant mRNA and DNA methylation levels of ABCB6 may serve as predictive biomarkers for early intrahepatic recurrence of HCV-positive HCC (9). In vitro studies by Mileo et al (10) and Quan et al (11) demonstrated that HCV may promote the progression of HCC cells by downregulating the protein and mRNA levels of proline rich protein BstNI subfamily 2/p130 and secreted frizzled-related protein, a Wnt antagonist, by inducing promoter hypermethylation. However, genes with aberrant DNA methylation for HCV-positive HCC remain largely under-investigated.

The aim of the present study was to identify novel genes to explain the development of HCV-positive HCC by combining mRNA expression profile and methylation profile microarrays, and to confirm their transcriptional and translational expression using The Cancer Genome Atlas (TCGA) datasets and Human Protein Atlas database (HPA). The results of the present study may provide novel therapeutic targets for HCV-positive HCC.

Materials and methods

Microarray data collection

Three microarray datasets: GSE19665 (12), GSE62232 (13) and GSE60753 (14) were downloaded from the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo) on July 25, 2018. The GSE19665 dataset [platform, GPL570 Affymetrix Human Genome U133 Plus 2.0 Array (HG-U133_Plus_2)] was used to analyze the gene expression profile in 20 HCC and 20 matched non-cancerous tissues, among which 5 pairs were HCV-positive; the GSE62232 dataset (platform, HG-U133_Plus_2) was used to detect the gene expression profile in 81 HCC (including 9 HCV-positive) and 10 normal liver tissues; and the GSE60753 dataset (platform, GPL13534, Illumina HumanMethylation450 BeadChip) was used to determine the DNA methylation profile in 156 HCC (including 12 HCV-positive), 34 normal liver tissues and 1 HCC cell line. Normal liver tissues were not matched with HCC in GSE62232 and GSE60753, but only collected from patients without HCC (such as benign cysts). Only the HCV-positive HCC and normal control samples were used for our following analyses.

Microarray data preprocessing

For the GSE19665 and GSE62232 datasets, the raw data were preprocessed using the oligo package (version 1.42.0; http://www.bioconductor.org/packages/release/bioc/html/oligo.html) in Bioconductor R package (version 3.4.1; http://www.R-project.org), including data transformation, missing value imputation with median, background correction with microarray analysis suite method and quantile normalization. For the GSE60753 dataset, the DNA methylation β values were downloaded and the genes were annotated according to the annotation information from the corresponding platform.

Differential gene expression and methylation analysis

Differentially expressed genes (DEGs) and differentially methylated genes (DMGs) between HCV-positive HCC and control samples were identified using the Linear Models for Microarray Data method (version 3.34.0; http://bioconductor.org/packages/release/bioc/html/limma.html) (15) in the Bioconductor R package. False discovery rate (FDR) <0.05 and |logFC| >1 were defined as the statistical threshold value; where FC is fold change. Hierarchical clustering (16) was performed for the DEGs and DMGs using pheatmap R package (version 1.0.8; http://cran.r-project.org/web/packages/pheatmap) based on Euclidean distance and the results were displayed as a heat map. The upregulated and downregulated shared DEGs in GSE19665 and GSE62232 datasets were then overlapped with the hypomethylated and hypermethylated DMGs, respectively, to identify the methylated-mediated genes. Additionally, the methylated-mediated genes were compared with the human oncogenes downloaded from the ONGene database (http://ongene.bioinfo-minzhao.org) (17) to screen for HCC-related oncogenes.

Protein-protein interaction (PPI) network construction and module analysis

The STRING database (version 10.0; http://string-db.org) (18) was used to predict the interactions between DEGs, and a PPI network was constructed using the obtained interaction pairs using the Cytoscape software (version 3.6.1; http://www.cytoscape.org) (19). The topological characteristics of the nodes (proteins) in the PPI network were computed using the CytoNCA plugin in the Cytoscape software (http://apps.cytoscape.org/apps/cytonca) (20) to determine the hub genes, including ‘degree’ [the number of edges (interactions) of a node (protein)], ‘betweenness’ (the number of shortest paths that run through a node), ‘closeness centrality’ (CC; the average length of the shortest paths to access all other proteins in the network) and ‘average path length’ (APL; the average distance between all pairs of nodes). Functionally related modules with well-interconnected genes were further identified in the PPI network using the Molecular Complex Detection (MCODE; version 1.4.2; http://apps.cytoscape.org/apps/mcode) algorithm (21) with the following scoring options: Degree cutoff=2; node score cutoff=0.2; K-core=2. Modules with MCODE score (Density*Nodes) >3 and node number >6 were considered to be significant.

Function enrichment analysis

Gene Ontology (GO) biological process terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed for the methylated-mediated DEGs using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool (version 6.8; http://david.abcc.ncifcrf.gov) (22) and BinGO (23) plugin in Cytoscape to predict their underlying functions. Statistical significance was defined as P<0.05 or FDR <0.05.

Validation of the selected methylation-mediated DEGs

The mRNA and methylation sequencing data of HCC tissues and normal liver tissues from patients without HCC were extracted from TCGA database (https://portal.gdc.cancer.gov) prior to July 25, 2018, which were measured on the Illumina HiSeq 2000 RNA Sequencing platform. Only the HCV-positive HCC samples were included in the present study to confirm the expression consistency of the methylated-mediated DEGs. The expression difference between HCC and controls was determined by Student's independent t-test using the TCGA data. P<0.05 was considered to indicate a statistically significant difference.

In addition, protein expression levels of the methylated-mediated DEGs were also validated by the HPA database (version 18; http://www.proteinatlas.org) (24), which was used to evaluate the translational levels of the DEGs by immunohistochemistry. Results are presented as the sum of scores of staining intensity (negative, weak, moderate or strong) and the percentage of stained cells (<25, 25–75 or >75%): Negative-not detected; weak + <25%-not detected; weak + 25–75 or 75%-low; moderate <25%-low; moderate + 25–75 or 75%-medium; strong <25%-medium, strong + 25–75 or 75%-high.

Results

Differential gene expression and methylation

A flowchart depicting the analytical process is presented as Fig. 1. Following preprocessing, a total of 1,306 (735 downregulated and 571 upregulated) and 1,249 (330 downregulated and 919 upregulated) DEGs were identified between HCV-positive HCC and control tissues in GSE19665 and GSE62232 datasets, respectively, using the cut-off criteria FDR <0.05 and |logFC| >1. The hierarchical-clustering heat map (Fig. 2A and B) indicated that DEGs may be used to distinguish HCV-positive HCC from control samples.

A total of 23,408 methylated probes were annotated to genes in the GSE60753 dataset. By comparing with normal samples, 1,448 DMGs (903 hypomethylated and 545 hypermethylated) were also obtained in HCV-positive HCC tissues. The hierarchical-clustering heat map (Fig. 2C) revealed that DMGs were different between HCV-positive HCC and control samples.

Following comparison of the DEGs identified in GSE19665 and GSE62232 datasets, 173 downregulated and 278 upregulated DEGs were revealed to be common and their expression trends were consistent in the two datasets (Fig. 3A). Further integration with the DMGs found 122 DEGs were downregulated by DNA hypermethylation and 63 DEGs were upregulated by DNA hypomethylation (Fig. 3B). Among the methylated DEGs, nine were suggested as human oncogenes according to the prediction by ONGene database; five were downregulated: Inhibitor of DNA binding 1, HLH protein (ID1), epithelial cell adhesion molecule (EPCAM), Fos proto-oncogene, AP-1 transcription factor subunit (FOS), ID2 and placenta specific 8 (PLAC8), whereas four were upregulated: Aurora kinase A (AURKA), ubiquitin conjugating enzyme E2 C (UBE2C), erb-b2 receptor tyrosine kinase 3 (ERBB3) and cyclin-dependent kinase inhibitor 3 (CDKN3).

Functional enrichment for the DEGs

The 122 downregulated/hypermethylated and 63 upregulated/hypomethylated DEGs were respectively uploaded to DAVID to predict their functions. Using the threshold value of FDR <0.05, 18 GO biological process terms were obtained for the downregulated/hypermethylated DEGs, including ‘response to wounding’ (FOS) and ‘inflammatory response’ (FOS), whereas 14 GO biological process terms were enriched for the upregulated/hypomethylated DEGs, including ‘cell cycle process’ (AURKA, CDKN3 and UBE2C) and ‘cell cycle’ (CDKN3 and MCM6) (Fig. 4A; Table I). Furthermore, KEGG pathway enrichment analysis was also performed, which resulted in 11 KEGG pathways identified as enriched for downregulated/hypermethylated and 4 enriched for upregulated/hypomethylated DEGs, using the threshold value of P<0.05 (FDR >0.05 for all pathways) (Fig. 4B; Table II). The KEGG pathway enrichment results were consistent with GO biological process term analysis, in which inflammatory-related ‘cytokine-cytokine receptor interaction’ pathway was enriched for downregulated/hypermethylated DEGs, and ‘DNA replication’ and ‘cell cycle’ were enriched for upregulated/hypomethylated DEGs.

Table I.

GO enrichment for methylation-related differentially expressed genes.

Table I.

GO enrichment for methylation-related differentially expressed genes.

A, Downregulated/hypermethylated genes
GO IDGO termFDRGenes
0051605Protein maturation by peptide bond cleavage 2.87×10−2CFP, C8B, C7, FCN3, KLKB1, C1R
0048545Response to steroid hormone stimulus 1.47×10−2PRSS8, FOS, GOT1, CCL2, ACADS, WFDC1, CA2, NPY1R, GHR
0046395Carboxylic acid catabolic process 2.18×10−2ASPA, GOT1, ACADS, IDO2, KMO, UROC1, PON3
0045087Innate immune response 2.26×10−3CFP, C8B, C7, FCN3, IL1RAP, VNN1, C1R, CD1D, GCH1
0031960Response to corticosteroid stimulus 2.93×10−2PRSS8, FOS, GOT1, CCL2, ACADS, GHR
0019439Aromatic compound catabolic process 2.70×10−2EPHX2, IDO2, KMO, PON3
0016054Organic acid catabolic process 2.18×10−2ASPA, GOT1, ACADS, IDO2, KMO, UROC1, PON3
0010817Regulation of hormone levels 4.64×10−2ALDH8A1, SHBG, LY6E, CRHBP, CYP26A1, SRD5A1, BCO2
0009725Response to hormone stimulus 2.54×10−2PRSS8, FOS, GOT1, CCL2, HMGCS2, ACADS, FBP1, WFDC1, CA2, NPY1R, GHR
0009719Response to endogenous stimulus 4.10×10−2PRSS8, FOS, GOT1, CCL2, HMGCS2, ACADS, FBP1, WFDC1, CA2, NPY1R, GHR
0009611Response to wounding 1.02×10−3C7, CCL2, HPS5, EPHX2, CHST4, C1R, CFP, C8B, FOS, LPA, PLSCR4, FCN3, KLKB1, IL1RAP, PROZ, VNN1, NGFR
0006959Humoral immune response 2.46×10−2CFP, C8B, C7, CCL2, FCN3, C1R
0006956Complement activation 2.70×10−2CFP, C8B, C7, FCN3, C1R
0006955Immune response 6.96×10−3C7, CCL2, CHST4, C1R, VIPR1, CXCL12, CD1D, GCH1, CFP, C8B, FCN3, HAMP, IL1RAP, VNN1
0006954Inflammatory response 5.26×10−3CFP, C8B, FOS, C7, CCL2, FCN3, KLKB1, IL1RAP, EPHX2, VNN1, C1R, CHST4
0006952Defense response 1.98×10−2C7, CCL2, EPHX2, CHST4, C1R, CD1D, GCH1, CFP, C8B, FOS, FCN3, HAMP, KLKB1, IL1RAP, VNN1
0006575Cellular amino acid derivative metabolic process 2.47×10−2GGT5, LY6E, IDO2, VNN1, KMO, BBOX1, GHR, GCH1
0002526Acute inflammatory response 3.37×10−3CFP, C8B, C7, FCN3, KLKB1, EPHX2, VNN1, C1R

B, Upregulated/hypomethylated genes

GO IDGO termFDRGenes

0051726Regulation of cell cycle 3.52×10−2TP53BP2, NUSAP1, SFN, CDKN3, UBE2C
0051301Cell division 4.37×10−2RAD21, NUSAP1, NDC80, PARD3B, CEP55, UBE2C, CDCA3
0022616DNA strand elongation 4.65×10−2RFC4, FEN1
0022403Cell cycle phase 3.99×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, CDKN3, UBE2C, CDCA3
0022402Cell cycle process 2.27×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, CDKN3, UBE2C, CDCA3
0007067Mitosis 3.63×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, UBE2C, CDCA3
0007049Cell cycle 3.98×10−2RAD21, TP53BP2, E2F8, NUSAP1, NDC80, AURKA, PARD3B, CEP55, CDKN3, UBE2C, CDCA3, MCM6
0006271DNA strand elongation during DNA replication 4.22×10−2RFC4, FEN1
0006261DNA-dependent DNA replication 4.56×10−2RFC4, MCM4, FEN1, MCM6
0006260DNA replication 4.15×10−2RFC4, RNASEH2A, MCM4, FEN1, MCM6

B, Upregulated/hypomethylated genes

GO IDGO termFDRGenes

000028Nuclear division 3.63×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, UBE2C, CDCA3
0000279M phase 4.61×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, UBE2C, CDCA3
0000278Mitotic cell cycle 3.06×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, CDKN3, UBE2C, CDCA3
0000087M phase of mitotic cell cycle 2.68×10−2RAD21, NUSAP1, NDC80, AURKA, CEP55, UBE2C, CDCA3

[i] FDR, false discovery rate; GO, Gene Ontology.

Table II.

KEGG pathway enrichment for methylation-related differentially expressed genes.

Table II.

KEGG pathway enrichment for methylation-related differentially expressed genes.

A, Downregulated/hypermethylated genes

KEGG IDKEGG pathwayP-valueGenes
hsa04610Complement and coagulation cascades 3.13×10−3C8B, C7, KLKB1, C1R
hsa00250Alanine, aspartate and glutamate metabolism 3.81×10−3ASPA, GOT1, ASS1
hsa00380Tryptophan metabolism 6.04×10−3AADAT, IDO2, KMO
hsa00460Cyanoamino acid metabolism 6.82×10−3GBA3, GGT5
hsa00830Retinol metabolism 1.02×10−2CYP4A11, CYP26A1, RDH16
hsa04060Cytokine-cytokine receptor interaction 2.67×10−2CCL2, IL1RAP, NGFR, CXCL12, GHR
hsa00270Cysteine and methionine metabolism 2.91×10−2GOT1, BHMT
hsa00620Pyruvate metabolism 3.33×10−2LDHD, ACOT12
hsa00071Fatty acid metabolism 3.33×10−2CYP4A11, ACADS
hsa00983Drug metabolism 3.53×10−2NAT2, UPP2
hsa04115p53 signaling pathway 4.98×10−2GADD45B, IGFBP3

B, Upregulated/hypomethylated genes

hsa03030DNA replication 1.70×10−5RFC4, RNASEH2A, MCM4, FEN1, MCM6

hsa04110Cell cycle 1.79×10−2RAD21, SFN, MCM4, MCM6
hsa04120Ubiquitin mediated proteolysis 4.67×10−2UBE2C, UBE2Q1
hsa05200Pathways in cancer 4.79×10−2LAMC1, CTNNA1

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

PPI network

The STRING database identified interaction relationships in 105 out of the 185 methylation-related DEGs (68 downregulated and 37 upregulated). The 211 interaction relationship pairs among the DEGs were used to construct a PPI network (Fig. 5A); seven of the previously identified human oncogenes were included (downregulated, ID1, FOS and EPCAM; upregulated, AURKA, CDKN3, UBE2C and ERBB3), as no interactions with other DEGs were identified for ID2 and PLAC8.

Two human oncogenes, FOS and CDKN3, were indicated as hub genes of the PPI network as they were shared and ranked in the top 15 for 4 topological characteristics (Table III). In addition, AURKA and UBE2C ranked top 5 in ‘degree’. ID1 was one of the top 10 genes in ‘CC’ and ‘APL’.

Table III.

Top 15 genes based on each topological characteristic.

Table III.

Top 15 genes based on each topological characteristic.

NodeDegreeNodeCloseness centralityNodeAverage path lengthNodeBetweenness Centrality
NDC8018FAT11.00FAT11.00FAT11.00
CDKN3a16SMAD51.00SMAD51.00FOS0.52
AURKAa16CBFA2T31.00CBFA2T31.00CDKN3a0.40
UBE2Ca15SPP21.00SPP21.00FEN10.36
NUSAP115FNIP11.00FNIP11.00MTR0.35
RFC414WFDC11.00WFDC11.00LPL0.25
KIF4A14ECM11.00ECM11.00LPA0.18
CEP5514SLC22A11.00SLC22A11.00IGFBP30.17
FEN113SLC10A11.00SLC10A11.00TXNRD10.16
ATAD213ID11.00ID11.00SHBG0.16
MCM413RRAGD1.00RRAGD1.00ASS10.16
FOS11CTNNA10.67CTNNA11.50CCL20.15
MCM611LAMC10.67LAMC11.50ACADS0.15
RNASEH2A11CDKN3a0.28FOS3.54APOF0.14
DEPDC18FOS0.28CDKN3a3.54CANX0.14

a Potential hub gene.

Subsequently, four highly connected PPI sub-modules (Fig. 5B) were extracted from the overall PPI network using MCODE. BinGO enrichment analysis demonstrated that the genes in module 1 (MCODE score=12.81) were involved in mitotic cell cycle (AURKA, CDKN3 and UBE2C); the genes in module 2 (MCODE score=5.067) were associated with detoxification of copper ions; the genes in module 3 (MCODE score=3.771) participated in the regulation of cell migration; and the genes in module 4 (MCODE score=3.41) were associated with carboxylic acid metabolic process (Table IV).

Table IV.

GO enrichment for genes in modules.

Table IV.

GO enrichment for genes in modules.

A, Module 1

GO ID PcorrGO termGenes in test set
48015 1.86×10−6 Phosphoinositide-mediated signalingFEN1, RFC4, UBE2C, NDC80, AURKA
278 3.01×10−5Mitotic cell cycleUBE2C, NUSAP1, NDC80, CEP55, AURKA, CDKN3
6260 3.01×10−5DNA replicationFEN1, RNASEH2A, RFC4, MCM4, MCM6
22403 3.65×10−5Cell cycle phaseUBE2C, NUSAP1, NDC80, CEP55, AURKA, CDKN3
280 3.65×10−5Nuclear divisionUBE2C, NUSAP1, NDC80, CEP55, AURKA
7067 3.65×10−5mitosisUBE2C, NUSAP1, NDC80, CEP55, AURKA
87 3.65×10−5M phase of mitotic cell cycleUBE2C, NUSAP1, NDC80, CEP55, AURKA
7049 4.09×10−5Cell cycleUBE2C, NUSAP1, MCM6, NDC80, CEP55, AURKA, CDKN3
51301 1.03×10−4Cell divisionUBE2C, NUSAP1, NDC80, CEP55, AURKA
22402 1.03×10−4Cell cycle processUBE2C, NUSAP1, NDC8, CEP55, AURKA, CDKN3
279 1.35×10−4M phaseUBE2C, NUSAP1, NDC80, CEP55, AURKA
35556 4.36×10−3Intracellular signal transductionFEN1, RFC4, UBE2C, NDC80, AURKA
6996 5.42×10−3Organelle organizationUBE2C, KIF4A, NUSAP1, NDC80, CEP55, AURKA
34645 1.06×10−2Cellular macromolecule biosynthetic processFEN1, RNASEH2A, RFC4, MCM4, MCM6
9059 1.08×10−2Macromolecule biosynthetic processFEN1, RNASEH2A, RFC4, MCM4, MCM6
23034 1.41×10−2Intracellular signaling pathwayFEN1, RFC4, UBE2C, NDC80, AURKA
90304 2.84×10−2Nucleic acid metabolic processFEN1, RNASEH2A, RFC4, MCM4, MCM6
44249 4.55×10−2Cellular biosynthetic processFEN1, RNASEH2A, RFC4, MCM4, MCM6

B, Module 2

GO ID PcorrGO termGenes in test set

10273 9.41×10−3Detoxification of copper ionMT2A
10038 9.41×10−3Response to metal ionMT2A, MT1X
10035 1.12×10−2Response to inorganic substanceMT2A, MT1X
6882 1.12×10−2Cellular zinc ion homeostasisMT2A
55069 1.12×10−2Zinc ion homeostasisMT2A
6878 1.12×10−2Cellular copper ion homeostasisMT2A
55070 1.12×10−2Copper ion homeostasisMT2A
7263 1.37×10−2Nitric oxide mediated signal transductionMT2A
46688 1.48×10−2Response to copper ionMT2A

C, Module 3

GO ID PcorrGO termGenes in test set

30334 1.28×10−2Regulation of cell migrationCXCL12, IGFBP3, THY1
51270 1.28×10−2Regulation of cellular component movementCXCL12, IGFBP3, THY1
40012 1.28×10−2Regulation of locomotionCXCL12, IGFBP3, THY1
42325 3.08×10−2Regulation of phosphorylationGHR, IGFBP3, THY1
19220 3.08×10−2Regulation of phosphate metabolic processGHR, IGFBP3, THY1
51174 3.08×10−2Regulation of phosphorus metabolic processGHR, IGFBP3, THY1
45595 3.08×10−2Regulation of cell differentiationGHR, IGFBP3, THY1
7155 3.56×10−2Cell adhesionCXCL12, THY1, IGFALS
22610 3.56×10−2Biological adhesionCXCL12, THY1, IGFALS
32879 3.64×10−2Regulation of localizationCXCL12, IGFBP3, THY1
50793 3.69×10−2Regulation of developmental processGHR, IGFBP3, THY1
48522 3.81×10−2Positive regulation of cellular processGHR, CXCL12, IGFBP3, THY1
48518 4.28×10−2Positive regulation of biological processGHR, CXCL12, IGFBP3, THY1
51239 4.29×10−2Regulation of multicellular organismal processGHR, IGFBP3, THY1
10646 4.53×10−2Regulation of cell communicationGHR, IGFBP3, THY1
7166 4.93×10−2Cell surface receptor linked signaling pathwayGHR, CXCL12, THY1

D, Module 4

GO ID PcorrGO termGenes in test set

19752 7.81×10−8Carboxylic acid metabolic processBHMT, GOT1, MTR, ALDH8A1, ACADS, ASPA, ASS1
43436 7.81×10−8Oxoacid metabolic processBHMT, GOT1, MTR, ALDH8A1, ACADS, ASPA, ASS1
6082 7.81×10−8Organic acid metabolic processBHMT, GOT1, MTR, ALDH8A1, ACADS, ASPA, ASS1
42180 7.81×10−8Cellular ketone metabolic processBHMT, GOT1, MTR, ALDH8A1, ACADS, ASPA, ASS1
6520 2.23×10−6Cellular amino acid metabolic processBHMT, GOT1, MTR, ASPA, ASS1
44106 6.48×10−6Cellular amine metabolic processBHMT, GOT1, MTR, ASPA, ASS1
44281 1.21×10−5Small molecule metabolic processBHMT, GOT1, MTR, ALDH8A1, ACADS, ASPA, ASS1
6519 1.21×10−5Cellular amino acid and derivative metabolic processBHMT, GOT1, MTR, ASPA, ASS1
9308 1.89×10−5Amine metabolic processBHMT, GOT1, MTR, ASPA, ASS1
44283 2.33×10−5Small molecule biosynthetic processBHMT, GOT1, MTR, ALDH8A1, ASS1
44237 1.82×10−3Cellular metabolic processBHMT, GOT1, TXNRD1, MTR, ALDH8A1, ACADS, ASPA, ASS1
8152 5.73×10−3Metabolic processBHMT, GOT1, TXNRD1, MTR, ALDH8A1, ACADS, ASPA, ASS1
44249 5.73×10−3Cellular biosynthetic processBHMT, GOT1, MTR, ALDH8A1, ASS1
9058 6.30×10−3Biosynthetic processBHMT, GOT1, MTR, ALDH8A1, ASS1
34641 9.55×10−3Cellular nitrogen compound metabolic processBHMT, GOT1, MTR, ASPA, ASS1
6807 1.19×10−2Nitrogen compound metabolic processBHMT, GOT1, MTR, ASPA, ASS1
44238 1.71×10−2Primary metabolic processBHMT, GOT1, MTR, ALDH8A1, ACADS, ASPA, ASS1

[i] GO, Gene Ontology; Pcorr, corrected P-value.

Validation of the selected methylation-mediated DEGs

Based on the enrichment and PPI analyses, it was hypothesized that downregulated/hypermethylated FOS and ID1 and upregulated/hypomethylated CDKN3, AURKA and UBE2C may be important human oncogenes for HCV-positive HCC. To further confirm their expression and methylation levels, the mRNA and methylation sequencing data of 58 HCV-HCC tissues and 50 normal controls were obtained from the TCGA database. The results demonstrated that the transcriptional expression and methylation levels of FOS, CDKN3 and AURKA in TCGA sequencing data (Fig. 6B) were consistent with the microarray data (Fig. 6A). However, the methylation level of UBE2C was not significantly different between HCV-positive HCC and normal control TCGA samples, although its expression level was consistent between TCGA sequencing data and our used microarray data (Fig. 6). The methylation level of ID1 had a detection value of 0 in the TCGA and thus comparison was not performed.

In addition, the HPA database was used to confirm the protein expression level of the genes in HCC by immunohistochemistry. Protein expression levels of AURKA in HCC tissues were higher, whereas protein expression levels of FOS in HCC tissues were lower compared with normal hepatocytes (Fig. 7). There was no immunohistochemical result for CDKN3 in the HPA database and no difference was observed in UBE2C and ID1 protein expression levels between HCC and normal control tissues.

Discussion

Through comprehensive analysis and validation, results from the present study indicated that AURKA and FOS may be crucial genes involved in HCV-positive HCC by participating in the cell cycle process and inflammatory response. HCV may upregulate the expression of AURKA and downregulate FOS by changes in DNA methylation.

HCV stimulates excessive cell proliferation in hepatocytes by dysregulating the cell cycle, which induces the development of HCC (25,26). Several positive cell cycle regulators (such as cyclin D1, cyclin E and Rb/p105) have been identified to be upregulated, whereas negative regulators [such as cyclin-dependent kinase 4 (CDK4), CDK6, p21Cip1, p27Kip1 and p57Kip2) are downregulated in patients with HCV-positive HCC compared with patients with chronic hepatitis C with or without liver cirrhosis (27). AURKA, which is located on chromosome 20q13.2, encodes a serine/threonine kinase involved in the assembly and maintenance of the mitotic spindle (28). Thus, AURKA is speculated to be a crucial gene for the regulation of cell cycle and carcinogenesis in several cancer types, including HCC (29). Yang et al demonstrated that knockdown of AURKA suppressed the growth of ovarian cancer cells by reducing centrosome amplification, malformation of mitotic spindles, and chromosome aberration (30). Additionally, restoring the expression of p21 and pRb attenuated the effects of AURKA silencing on cell cycle progression (30). Using RNA microarray and reverse transcriptase-quantitative PCR analysis, Zhou et al reported that AURKA was significantly upregulated in human urothelial carcinoma compared with normal urothelium (31) and demonstrated that AURKA inhibitor MLN8237 induced cell-cycle arrest, aneuploidy, mitotic spindle failure and apoptosis in human bladder cancer cells, which arrested tumor growth (31). Li et al also used the MLN8237 to demonstrate that AURKA regulated cell cycle in breast cancer cells by modulating the p53/p21/cell division control 2/cyclin B1 pathway (32). Similarly, the verification experiments demonstrated that AURKA inhibitor alisertib arrested HCC cells in G2/M phase and induced an accumulation of aneuploidy by regulating the expression of key cell cycle regulators such as cyclin B1 (33,34). The present study demonstrated that AURKA was highly expressed at the mRNA and protein levels in HCV-positive HCC. In addition to the cell cycle, a recent study has suggested that AURKA contributes to tumor migration, invasion, epithelial mesenchymal transition and cancer stem cell behaviors, which also have been preliminarily validated in HCC (35), but not HCV-related HCC. Thus, further investigation of the roles of AURKA in HCV-related HCC remains necessary.

DNA methylation is an important mechanism for regulating gene expression epigenetically. Hypermethylation of genes is associated with reduced expression, whereas hypomethylation is associated with increased expression. Thus, high expression of AURKA in HCV-positive HCC was predicted to be due to hypomethylation, which was validated by the microarray and TCGA data. This conclusion agreed with a previous study on esophageal cancer, in which AURKA methylation and human papillomavirus infection was higher in precancer, esophagitis and normal tissues compared with cancer tissues (36). However, further experiments are needed to confirm the effects of HCV on the methylation of AURKA and the development of HCC.

FOS is a member of the fos family of transcription factors, which has been extensively demonstrated to be a pro-oncogenic gene and promote proliferation, invasion and metastasis of cancer through AP-1-related mechanisms, including HCV-positive HCC (37). However, in the present study, FOS was downregulated and hypermethylated. This may indicate that FOS may be a dual-function gene, which has been identified in other cancers, such as ovarian cancer (38) and pancreatic cancer (39). Alternatively, the results may be negative due to the small sample size. Further studies with larger sample sizes are needed to confirm role of FOS in HCV-positive HCC.

There were certain limitations to the present study. First, although the known microarray and TCGA sequencing data were included to confirm the expression and methylation levels of crucial genes, the sample size associated with HCV-positive HCC was small. Therefore, more clinical samples need to be collected to further confirm their expression levels. Second, although the present study has suggested that the expression levels of AURKA and FOS may be regulated by methylation, additional in vitro and in vivo experiments using a methylation inhibitor, such as 5-azacytidine, are essential to verify these results. Third, HCV infection-related in vitro and in vivo experiments (accompanied with overexpression or knockdown of genes) are also needed to demonstrate the functional roles of AURKA and FOS in cell proliferation, apoptosis, migration and invasion.

The results of the present study preliminarily indicate that aberrantly methylated AURKA and FOS may be potential therapeutic targets for treatment of HCV-positive HCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The sequencing datasets GSE19665 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE19665), GSE62232 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE62232) and GSE60753 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE60753) were downloaded from the GEO database in NCBI. The mRNA and microRNA sequencing data were obtained from The Cancer Genome Atlas (https://tcga-data.nci.nih.gov).

Authors' contributions

ZM and XH conceived and designed the study; YL and ZH performed the acquisition of data; ZM and YL conducted the statistical analysis. WL and ZH were involved in the interpretation of the data. ZM drafted the manuscript. XH 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 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Jing L, Huang L, Yan J, Qiu M and Yan Y: Liver resection for hepatocellular carcinoma: Personal experiences in a series of 1330 consecutive cases in China. ANZ J Surg. 88:E713–E717. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Petruzziello A: Epidemiology of Hepatitis B Virus (HBV) and Hepatitis C Virus (HCV) related hepatocellular carcinoma. Open Virol J. 12:26–32. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Moore MS, Bocour A, Tran OC, Qiao B, Schymura MJ, Laraque F and Winters A: Effect of hepatocellular carcinoma on mortality among individuals with Hepatitis B or Hepatitis C infection in New York City, 2001–2012. Open Forum Infect Dis. 5:ofy1442018. View Article : Google Scholar : PubMed/NCBI

5 

Kiran M, Chawla YK and Kaur J: Methylation profiling of tumor suppressor genes and oncogenes in hepatitis virus-related hepatocellular carcinoma in northern India. Cancer Genet Cytogenet. 195:112–119. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Zekri AR, Bahnasy AA, Shoeab FE, Mohamed WS, El-Dahshan DH, Ali FT, Sabry GM, Dasgupta N and Daoud SS: Methylation of multiple genes in hepatitis C virus associated hepatocellular carcinoma. J Adv Res. 5:27–40. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Ramadan RA, Zaki MA, Awad AM and El-Ghalid LA: Aberrant methylation of promoter region of SPINT2/HAI-2 gene: An epigenetic mechanism in hepatitis C virus-induced hepatocarcinogenesis. Genet Test Mol Biomarkers. 19:399–404. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Takagi K, Fujiwara K, Takayama T, Mamiya T, Soma M and Nagase H: DNA hypermethylation of zygote arrest 1 (ZAR1) in hepatitis C virus positive related hepatocellular carcinoma. Springerplus. 2:1502013. View Article : Google Scholar : PubMed/NCBI

9 

Tsunedomi R, Iizuka N, Yoshimura K, Iida M, Tsutsui M, Hashimoto N, Kanekiyo S, Sakamoto K, Tamesa T and Oka M: ABCB6 mRNA and DNA methylation levels serve as useful biomarkers for prediction of early intrahepatic recurrence of hepatitis C virus-related hepatocellular carcinoma. Int J Oncol. 42:1551–1559. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Mileo AM, Mattarocci S, Matarrese P, Anticoli S, Abbruzzese C, Catone S, Sacco R, Paggi MG and Ruggieri A: Hepatitis C virus core protein modulates pRb2/p130 expression in human hepatocellular carcinoma cell lines through promoter methylation. J Exp Clin Cancer Res. 34:1402015. View Article : Google Scholar : PubMed/NCBI

11 

Quan H, Zhou F, Nie D, Chen Q, Cai X, Shan X, Zhou Z, Chen K, Huang A, Li S and Tang N: Hepatitis C virus core protein epigenetically silences SFRP1 and enhances HCC aggressiveness by inducing epithelial-mesenchymal transition. Oncogene. 33:2826–2835. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Deng YB, Nagae G, Midorikawa Y, Yagi K, Tsutsumi S, Yamamoto S, Hasegawa K, Kokudo N, Aburatani H and Kaneda A: Identification of genes preferentially methylated in hepatitis C virus-related hepatocellular carcinoma. Cancer Sci. 101:1501–1510. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Yang Y, Chen L, Gu J, Zhang H, Yuan J, Lian Q, Lv G, Wang S, Wu Y, Yang YC, et al: Recurrently deregulated lncRNAs in hepatocellular carcinoma. Nat Commun. 8:144212017. View Article : Google Scholar : PubMed/NCBI

14 

Hlady RA, Tiedemann RL, Puszyk W, Zendejas I, Roberts LR, Choi JH, Liu C and Robertson KD: Epigenetic signatures of alcohol abuse and hepatitis infection during human hepatocarcinogenesis. Oncotarget. 5:9425–9443. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Liu Y, Sun J and Zhao M: ONGene: A literature-based database for human oncogenes. J Genet Genomics. 44:119–121. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: Protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res 43 (Database Issue). D447–D452. 2015. View Article : Google Scholar

19 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Tang Y, Li M, Wang J, Pan Y and Wu FX: CytoNCA: A cytoscape plugin for centrality analysis and evaluation of protein interaction networks. Biosystems. 127:67–72. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

22 

Huang DW, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Maere S, Heymans K and Kuiper M: BiNGO: A Cytoscape plugin to assess overrepresentation of Gene Ontology categories in Biological Networks. Bioinformatics. 21:3448–3449. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Pontén F, Jirström K and Uhlen M: The human protein atlas-a tool for pathology. J Pathol. 216:387–393. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Irshad M, Gupta P and Irshad K: Molecular basis of hepatocellular carcinoma induced by hepatitis C virus infection. World J Hepatol. 9:1305–1314. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Moustafa S, Karakasiliotis I and Mavromara P: Hepatitis C virus core+1/ARF protein modulates Cyclin D1/pRb pathway and promotes carcinogenesis. J Virol. 92(pii): e02036–17. 2018.PubMed/NCBI

27 

Bassiouny AEE, Nosseir MM, Zoheiry MK, Ameen NA, Abdelhadi AM, Ibrahim IM, Zada S, El-Deen AH and El-Bassiouni NE: Differential expression of cell cycle regulators in HCV-infection and related hepatocellular carcinoma. World J Hepatol. 2:32–41. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Kufer TA, Silljé HH, Körner R, Gruss OJ, Meraldi P and Nigg EA: Human TPX2 is required for targeting Aurora-A kinase to the spindle. J Cell Biol. 158:617–623. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Benten D, Keller G, Quaas A, Schrader J, Gontarewicz A, Balabanov S, Braig M, Wege H, Moll J, Lohse AW and Brummendorf TH: Aurora kinase inhibitor PHA-739358 suppresses growth of hepatocellular carcinoma in vitro and in a xenograft mouse model. Neoplasia. 11:934–944. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Yang G, Chang B, Yang F, Guo X, Cai KQ, Xiao X, Wang H, Sen S, Hung MC, Mills GB, et al: Aurora Kinase A promotes ovarian tumorigenesis through dysregulation of the cell cycle and suppression of BRCA2. Clin Cancer Res. 16:3171–3181. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Zhou N, Singh K, Mir MC, Parker Y, Lindner D, Dreicer R, Ecsedy JA, Zhang Z, The BT, Almasan A and Hansel DE: The investigational Aurora kinase A inhibitor MLN8237 induces defects in cell viability and cell cycle progression in malignant bladder cancer cells in vitro and in vivo. Clin Cancer Res. 19:1717–1728. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Li JP, Yang YX, Liu QL, Pan ST, He ZX, Zhang X, Yang T, Chen XW, Dong W, Qiu JX and Zhou SF: The investigational Aurora kinase A inhibitor alisertib (MLN8237) induces cell cycle G2/M arrest, apoptosis, and autophagy via p38 MAPK and Akt/mTOR signaling pathways in human breast cancer cells. Drug Des Devel Ther. 9:1627–1652. 2015.PubMed/NCBI

33 

Zhu Q, Yu X, Zhou ZW, Zhou C, Chen XW and Zhou SF: Inhibition of Aurora A Kinase by alisertib induces autophagy and cell cycle arrest and increases chemosensitivity in human hepatocellular carcinoma HepG2 cells. Curr Cancer Drug Targets. 17:386–401. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Zhu Q, Yu X, Zhou ZW, Luo M, Zhou C, He ZX, Chen Y and Zhou SF: A quantitative proteomic response of hepatocellular carcinoma Hep3B cells to danusertib, a pan-Aurora kinase inhibitor. J Cancer. 9:2061–2071. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Chen C, Song G, Xiang J, Zhang H, Zhao S and Zhan Y: AURKA promotes cancer metastasis by regulating epithelial-mesenchymal transition and cancer stem cell properties in hepatocellular carcinoma. Biochem Biophys Res Commun. 486:514–520. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Mohiuddin MK, Chava S, Upendrum P, Latha M, Zubeda S, Kumar A, Ahuja YR, Hasan Q and Mohan V: Role of Human papilloma virus infection and altered methylation of specific genes in esophageal cancer. Asian Pac J Cancer Prev. 14:4187–4193. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Watanabe T, Hiasa Y, Tokumoto Y, Hirooka M, Abe M, Ikeda Y, Matsuura B, Chung RT and Onji M: Protein Kinase R modulates c-Fos and c-Jun signaling to promote proliferation of hepatocellular carcinoma with Hepatitis C virus infection. PLoS One. 8:e677502013. View Article : Google Scholar : PubMed/NCBI

38 

Oliveiraferrer L, Rößler K, Haustein V, Schröder C, Wicklein D, Maltseva D, Khaustova N, Samatov T, Tonevitsky A, Mahner S, et al: c-FOS suppresses ovarian cancer progression by changing adhesion. Br J Cancer. 110:753–763. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Guo JC, Li J, Zhao YP, Zhou L, Cui QC, Zhou WX, Zhang TP and You L: Expression of c-fos was associated with clinicopathologic characteristics and prognosis in pancreatic cancer. PLoS One. 10:e01203322015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2019
Volume 20 Issue 3

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
Ma Z, Liu Y, Hao Z, Hua X and Li W: DNA hypermethylation of aurora kinase A in hepatitis C virus‑positive hepatocellular carcinoma. Mol Med Rep 20: 2519-2532, 2019
APA
Ma, Z., Liu, Y., Hao, Z., Hua, X., & Li, W. (2019). DNA hypermethylation of aurora kinase A in hepatitis C virus‑positive hepatocellular carcinoma. Molecular Medicine Reports, 20, 2519-2532. https://doi.org/10.3892/mmr.2019.10487
MLA
Ma, Z., Liu, Y., Hao, Z., Hua, X., Li, W."DNA hypermethylation of aurora kinase A in hepatitis C virus‑positive hepatocellular carcinoma". Molecular Medicine Reports 20.3 (2019): 2519-2532.
Chicago
Ma, Z., Liu, Y., Hao, Z., Hua, X., Li, W."DNA hypermethylation of aurora kinase A in hepatitis C virus‑positive hepatocellular carcinoma". Molecular Medicine Reports 20, no. 3 (2019): 2519-2532. https://doi.org/10.3892/mmr.2019.10487