Open Access

Using Illumina Infinium HumanMethylation 450K BeadChip to explore genome‑wide DNA methylation profiles in a human hepatocellular carcinoma cell line

  • Authors:
    • Ning Sun
    • Jialin Zhang
    • Chengshuo Zhang
    • Yue Shi
    • Bochao Zhao
    • Ao Jiao
    • Baomin Chen
  • View Affiliations

  • Published online on: September 3, 2018     https://doi.org/10.3892/mmr.2018.9441
  • Pages: 4446-4456
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aberrant DNA methylation is the most common type of epigenetic alteration and is associated with many types of cancer. Although previous studies have provided a few novel DNA methylation markers in hepatocellular carcinoma (HCC), specific DNA methylation patterns and comparisons of the aberrant alterations in methylation between HCC and normal liver cell lines have not yet been reported. Therefore, in the present study the Illumina Infinium HumanMethylation 450K BeadChip was employed to identify the genome‑wide aberrant DNA methylation profiles of Huh7 and L02 cells. Following Bonferroni adjustment, 102,254 differentially methylated CpG sites (covering 26,511 genes) were detected between Huh7 and L02 cells. Of those CpG sites, 62,702 (61.3%) sites were hypermethylated (covering 12,665 genes) and 39,552 (38.7%) sites were hypomethylated (covering 13,846 genes). The results of the present study indicated that 40.3% of the CpG sites were in CpG island regions, 20.7% were in CpG shores and 8.8% were in shelf regions. A total of 57.3% hypermethylated CpG sites and 39.4% of the hypomethylated CpG sites had a |β‑Difference| ≥50%. Within the significant differentially methylated CpG sites, 490 genes were located within 598 differentially methylated regions. Gene Ontology enrichment analysis revealed that 2,107 differentially methylated genes were associated with ‘biological process’, 13,351 differentially methylated genes were associated with ‘molecular function’, and 18,041 differentially methylated genes were associated with ‘cellular component’. Kyoto Encyclopedia of Genes and Genomes pathway‑based analysis revealed 43 signaling pathways that were associated with 5,195 differentially methylated genes. These results demonstrated that aberrant DNA methylation may be a key and common event underlying the tumorigenesis of Huh7 cells. The present study also identified many subsets of hypo‑ or hyper‑methylated CpG sites, genes and signaling pathways, which have an importance in the occurrence and development of HCC.

Introduction

Worldwide, primary liver cancer (PLC) is the second leading cause of cancer-associated mortality in poorly developed countries and the sixth in more developed countries (1). Of the ~782,500 new annual cases of PLC, China accounts for >50% of the associated incidence (2). In 2015, PLC was the fourth most common type of cancer and the third most common cause of cancer-associated mortality in China (3). The majority of PLC cases occurring worldwide are cases of hepatocellular carcinoma (HCC) (1). Many risk factors can induce the development of HCC, including chronic hepatitis B virus or hepatitis C virus infections, chronic alcoholic cirrhosis and high doses of aflatoxin B1 (4). Although a number of studies have identified a few of the molecular alterations associated with the pathogenesis of HCC, the main mechanism underlying HCC is still unclear.

Previous studies have demonstrated that epigenetic alterations are one of the many early events that occur during tumorigenesis (5,6). DNA methylation is the main epigenetic feature in regulating gene transcriptional regulation and preserving genome stability; however, aberrant DNA methylation can lead to the inactivation of tumor suppressor genes or activation of oncogenes, which eventually induces the development of many types of cancer (7,8). A number of studies have also reported alterations in one or several genes at one time; the abnormal methylation of genes, including Ras association domain family member 1 (9), p16, postmeiotic segregation increased 2, MutL homolog 1, MutS homolog 2 (10), Adenomatosis polyposis coli (11) and glutathione S-transferase Pi 1 (12,13), has also been associated with HCC. Shen et al (14) used Illumina Infinium HumanMethylation 27K arrays to analyze 27,578 CpG sites covering 14,495 genes in paired HCC tumor and adjacent non-tumor tissues. The Illumina Infinium HumanMethylation 450K BeadChip represents a significant improvement in the detection of CpG sites (482,421 CpG and 3,091 non-CpG sites), covers 99% of RefSeq genes with multiple sites in annotated promoters (1,500 or 200 bp upstream of the transcription start site), 5′-untranslated regions (UTRs), first exons, gene body, and 3′-UTRs (15). Previously, aberrant DNA hypermethylation of CpG islands was reported to induce the inactivation of tumor suppressor genes (16), which was thought to contribute to tumorigenesis (17). Recently, previous studies have revealed that cancer-associated aberrant DNA methylation not only occurs within CpG islands but may also be detected within CpG shores or CpG shelves (1820).

To the best of our knowledge, no research analyzing the genome-wide DNA methylation status within a HCC cell line using the Illumina Infinium HumanMethylation 450K BeadChip has been conducted. Therefore, in the present study, the Illumina 450K Methylation BeadChip was employed to screen promoter DNA methylation and the expression profiles of methylated genes in a human hepatocellular carcinoma cell line (Huh7 cells) and in a human normal liver cell line (L02 cells). The results may aid the characterization of differentially methylated CpG sites, regions and genes associated with the pathogenesis of HCC, thereby improving our current understanding of the methylation mechanisms underlying the development and progression of HCC.

Materials and methods

Cell culture

The human HCC cell line, Huh7 and the human normal liver cell line, L02, were purchased from the Cell Bank of Type Culture Collection of Chinese Academy of Sciences (Shanghai, China). Huh7 cells were maintained in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), while L02 cells were maintained in RPMI-1640 (Invitrogen; Thermo Fisher Scientific, Inc.). The cell lines were supplemented with 100 U/ml penicillin and 100 g/ml streptomycin in the presence of 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and incubated in a humidified atmosphere containing 5% CO2 at 37°C.

DNA preparation and Illumina Infinium HumanMethylation 450K BeadChip assay

DNA was extracted from the two cell lines (Huh7 and L02) using a QIAamp DNA Micro kit (Qiagen GmbH, Hilden, Germany) according to the manufacturer's protocol. Bisulfite modification of 1 µg DNA was conducted using an EZ DNA Methylation kit (Zymo Research Corp., Irvine, CA, USA) according to the manufacturer's protocol. The Illumina Infinium HumanMethylation 450K BeadChip assay was performed according to Illumina's standard protocol (Illumina, Inc., San Diego, CA, USA). Experiments with Huh7 and L02 cells were performed in triplicate to avoid false positive and false negative results.

Statistical analysis

The methylation data were processed with the Methylation Module of GenomeStudio software (Methylation v1.9; Illumina, Inc.). The methylation levels of the CpG sites were calculated as β-values: β=intensity of the methylated allele (M)/[intensity of the unmethylated allele (U) + M + 100] (15). A t-test, in addition to analysis of variance with Bonferroni correction for multiple comparisons was used to compare differentially methylated CpG sites between Huh7 and L02 cells. The differentially methylated CpG sites were defined as sites with Adjusted P-values of ≤0.05 and |β-Difference|≥0.2. Methylation measures with a detection P>0.05 and CpG coverage <95% were excluded (14). For the selection of candidate CpG sites that had significant differences between Huh7 and L02 cell methylation levels, the following additional filtering criteria were applied: i) Adjusted P≤0.05, which corresponds to a raw P-value of ≤1.06×10−7; ii) for significantly hypermethylated CpG sites, the |β-Difference| in the methylation levels between Huh7 and L02 cells was >20%, and the mean methylation level for L02 was <25%; and iii) for significantly hypomethylated CpG sites, the methylation level |β-Difference| between L02 and Huh7 cells was >20%, and the mean methylation level for Huh7 cells was <25% (14).

Functional annotation of differentially methylated genes

The genes for which the CpG sites corresponded with differential methylation levels were determined using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway (www.genome.jp/kegg/) and Gene Ontology (GO; www.geneontology.org/) databases to analyze the pathway and enrichment information of these genes.

Results

Global DNA methylation in Huh7 and LO2 cells

Following a t-test with Bonferroni correction for multiple comparisons, 102,254 differentially methylated CpG sites (covering 26,511 genes) were detected between Huh7 and L02 cells. Within these CpG sites, 62,702 (61.3%) sites were hypermethylated (covering 12,665 genes), and 39,552 (38.7%) sites were hypomethylated (covering 13,846 genes). The results suggested that aberrant DNA methylation may a very common event in Huh7 cells, and alterations in hypermethylation were more frequently observed than hypomethylation. Figs. 1 and 2 present hierarchical cluster analysis of the differentially methylated CpG sites and genes that distinguish Huh7 from L02 cells.

In addition, the results revealed that there were 41,178 (40.3%) CpG sites located in CpG island regions, 21,150 (20.7%) CpG sites were within CpG shores, 9,030 (8.8%) CpG sites were in shelves, and 30,896 (30.2%) CpG sites were in open sea (Table I). Furthermore, the results also demonstrated that within different regions, the distribution of hypo- or hypermethylated CpG sites differed: In CpG island regions, 19,462 (47.3%) CpG sites were hypermethylated and 21,716 (52.7%) CpG sites were hypomethylated. In CpG shore regions, 13,729 (64.9%) CpG sites were hypermethylated and 7,421 (35.1%) CpG sites were hypomethylated. In shelf regions, 7,340 (81.3%) CpG sites were hypermethylated and 1,690 (18.7%) CpG sites were hypomethylated.

Table I.

Distribution of all of the differentially methylated CpG sites.

Table I.

Distribution of all of the differentially methylated CpG sites.

TypeAll methylated CpG sites, n (%)Hypermethylated CpG sites, n (%)Hypomethylation CpG sites, n (%)
CpG island41,178 (40.3)19,462 (47.3)21,716 (52.7)
CpG shores21,150 (20.7)13,729 (64.9)7,421 (35.1)
CpG shelves9,030 (8.8)7,340 (81.3)1,690 (18.7)
Open sea30,896 (30.2)22,171 (71.8)8,725 (28.2)
Total102,25462,70239,552

Frequency distribution of differentially methylated CpG sites between Huh7 and L02 cells. In the results of the present study, 35,937 (57.3%) hypermethylated CpG sites and 15,587 (39.4%) hypomethylated CpG sites were observed to have an |β-Difference|≥50%. A total of 18,529 (29.5%) of hypermethylated CpG sites and 14,177 (35.9%) hypomethylated CpG sites had an |β-Difference|≥30% but <50%. A total of 8,236 (13.1%) hypermethylated CpG sites and 9,788 (24.7%) of hypomethylated CpG sites had an |β-Difference|<30% but ≥20% (Table II). Collectively, these results revealed that DNA aberrant hypermethylation in Huh7 cells was more frequent than in L02 cells, which may serve a potential role in genomic instability.

Table II.

Frequency distribution of all of the differentially methylated CpG sites in Huh7 and L02 cells by methylation status.

Table II.

Frequency distribution of all of the differentially methylated CpG sites in Huh7 and L02 cells by methylation status.

|β-difference|, %Hypermethylated CpG sites, n (%)Cumulative, %Hypomethylated CpG sites, n (%)Cumulative, %Total CpG sites, n (%)Cumulative, %
≥6025,585 (40.8)40.810,796 (27.3)27.336,381 (35.6)35.6
50≤x<6010,352 (16.5)57.34,791 (12.1)39.415,134 (14.8)50.4
40≤x<509,681 (15.4)72.76,159 (15.6)54.015,840 (15.5)65.9
30≤x<408,848 (14.1)86.88,018 (20.3)75.316,866 (16.5)82.4
20≤x<308,236 (13.1)100.09,788 (24.7)100.018,024 (17.6)100.0
Total62,70239,552102,254
Significant differentially methylated CpG sites and genes

To reduce the potential impact of an extreme β value on methylation differences, the present study applied stringent criteria to select potentially biologically important CpG sites (14). A total of 5,285 significantly hypermethylated CpG sites (covering 3,222 genes) and 2,659 significantly hypomethylated CpG sites (covering 2,204 genes) were observed. For the significantly hypermethylated CpG sites, there were 1,544 sites in CpG islands, 1,137 sites in CpG shores, 655 sites within CpG shelves and 1,949 sites in open sea regions. By contrast, for the significantly hypomethylated CpG sites, there were 1,201 sites in CpG islands, 632 sites in CpG shores, 133 sites in CpG shelves and 693 sites in open sea regions (Table III). The top 20 differentially hypermethylated and hypomethylated sites and genes are presented in the Tables IV and V.

Table III.

Distribution of genomic regions for significant differentially methylated CpG sites in Huh7 cells when compared with L02 cells.

Table III.

Distribution of genomic regions for significant differentially methylated CpG sites in Huh7 cells when compared with L02 cells.

TypeHypermethylated CpG sites, nHypomethylated CpG sites, n
CpG island1,5441,201
CpG shores1,137   632
CpG shelves   655   133
Open sea1,949   693
Total5,2852,659

Table IV.

Top 20 significant hypermethylated CpG sites and genes within differentially methylated regions in Huh7 cells when compared with L02 cells.

Table IV.

Top 20 significant hypermethylated CpG sites and genes within differentially methylated regions in Huh7 cells when compared with L02 cells.

CpG sitesAdjust P-value|β-difference|Mean HuhMean L02Hypermethylated genes
cg11058366 2.09×10−60.9240.9340.010ERBB4
cg13245152 1.69×10−60.8630.8760.013PAX6
cg25758545 1.69×10−60.8630.8760.013SALL4
cg14950829 1.69×10−60.9250.9400.015PCDH8
cg09260089 1.69×10−60.9520.9680.016NKX6-2
cg11459773 1.69×10−60.8760.8910.015BCL3
cg12989574 1.69×10−60.9650.9830.018GPC6
cg03129384 1.69×10−60.9560.9740.018FAM196A; DOCK1
cg03396151 1.69×10−60.9290.9470.018MEIS2
cg04556126 1.69×10−60.9200.9380.018ZIC4
cg20317123 1.69×10−60.9470.9660.019TCF21
cg21062760 1.69×10−60.8760.8940.018ZBTB32
cg09454560 1.99×10−60.6240.6360.013LRFN2
cg12090740 1.69×10−60.8920.9110.020BCL2
cg24249411 1.69×10−60.8870.9070.020BDNF
cg00057722 1.69×10−60.9290.9500.021
cg08640046 1.69×10−60.8100.8280.018
cg03283124 2.03×10−60.8980.9200.021PCDH9
cg13087076 1.69×10−60.8900.9120.021DYDC2
cg25453154 2.03×10−60.8200.8390.020ZCCHC24

[i] Data are presented to 3 decimal places.

Table V.

Top 20 significant hypomethylated CpG sites and genes within differentially methylated regions in Huh7 cells when compared with L02 cells.

Table V.

Top 20 significant hypomethylated CpG sites and genes within differentially methylated regions in Huh7 cells when compared with L02 cells.

CpG sitesAdjust P-value|β-difference|Mean HuhMean L02Hypomethylated gene
cg10739344 1.70×10−60.8940.0190.913WDR76
cg00618865 1.69×10−60.9450.0230.967PLXND1
cg16267343 1.69×10−60.8980.0240.922NPR3
cg00138041 1.69×10−60.9430.0290.972PRDM8
cg01529365 1.69×10−60.9300.0310.961
cg09564253 1.69×10−60.9020.0310.933LASP1
cg08176368 1.69×10−60.9260.0330.959MMP9
cg08812555 2.05×10−60.7840.0290.813DKK1
cg25612391 1.77×10−60.7410.0280.769SLC25A42
cg22417879 1.70×10−60.9080.0350.943SDCBP2
cg15019790 1.69×10−60.9240.0360.960SIX2
cg07407787 1.69×10−60.9220.0360.958ARSG; SLC16A6
cg08361684 1.69×10−60.9110.0380.949FJX1
cg16195157 1.69×10−60.9000.0380.938DNAJB1
cg15842502 1.69×10−60.9130.0400.953RB1
cg13848566 1.95×10−60.9340.0410.975GAS1
cg27454412 1.81×10−60.7810.0340.815C7orf50
cg02152578 1.69×10−60.9310.0410.972AHCYL1
cg13355248 1.69×10−60.7920.0380.829NPTX1
cg16443866 1.69×10−60.8780.0420.920STC2

[i] Data are presented to 3 decimal places.

Significant differentially methylated regions (DMRs)

The results of the present study revealed that 390 significantly hypermethylated CpG sites (covering 287 genes) and 208 significantly hypomethylated CpG sites (covering 203 genes) were in DMRs. For the significantly hypermethylated CpG sites, 64 sites were in cancer-specific (c)-DMRs, 125 sites were in reprogramming-specific (r)-DMRs and 201 sites were in DMRs; for the significantly hypomethylated CpG sites, 30 were located within cDMRs, 74 were located within rDMRs and 104 were located within DMRs (Table VI; Fig. 3).

Table VI.

Significant differentially methylated regions.

Table VI.

Significant differentially methylated regions.

DMRsSignificantly hypermethylated sites (n)Significantly hypermethylated sites (n)
cDMRs  64  30
Island  16  4
Shores  36  18
Shelves  5  3
Open sea  7  5
rDMRs125  74
Island  38  13
Shores  70  50
Shelves  7  3
Open sea  10  8
DMRs201104
Island179  70
Shores  13  21
Shelves  3  0
Open sea  6  13
Total390208

[i] DMRs, differentially methylated regions; cDMRs, cancer-specific-DMRs; rDMRs, reprogramming-specific-DMRs.

GO enrichment and KEGG pathway analysis

GO enrichment and the KEGG Pathway database were employed to analyze information regarding the differentially methylated genes. The results of GO enrichment revealed that there were 2,107 differentially methylated genes associated with ‘biological process’, and the most enriched groups included negative regulators of cell proliferation, negative regulators of transcription such as RNA polymerase II promoter, and synaptic transmission. A total of 13,351 differentially methylated genes were associated with ‘molecular function’, and the most enriched groups included protein binding, DNA binding and metal ion binding. A total of 18,041 differentially methylated genes were associated with ‘cellular component’, and the most enriched groups included the nucleus, cytoplasm and cytosol (Fig. 4). The top 20 significant differentially methylated genes obtained from GO enrichment analysis are listed in Table VII.

Table VII.

Top 20 significant differentially methylated genes in Gene Ontology enrichment.

Table VII.

Top 20 significant differentially methylated genes in Gene Ontology enrichment.

GO enrichmentTop 20 significantly hypermethylated genes in GO enrichmentTop 20 significantly hypomethylated genes in GO enrichment
Biological process
  Positive regulation of protein phosphorylationERBB4
  Negative regulation of cell proliferationERBB4
  Epidermal growth factor receptor signalingERBB4
pathway
  Synaptic transmissionPCDH8
  Negative regulation of transcription from RNASALL4; NKX6-2;
polymerase II promoterMEIS2; TCF21; ZBTB32;
  Fibroblast growth factor receptor signaling pathwayERBB4
Molecular function
  Protein bindingERBB4; PAX6; BCL3; DOCK1; ZBTB32; BCL2PLXND1; LASP1; MMP9; DKK1; DNAJB1; RB1; GAS1; AHCYL1
  ATP bindingERBB4
  Sequence-specific DNA binding transcription factor activityPAX6; NKX6-2; BCL3SIX2; RB1
  DNA bindingPAX6; SALL4; BCL3; ZIC4; ZBTB32PRDM8; RB1
  Metal ion bindingSALL4; ZIC4; ZBTB3PRDM8; ARSG; NPTX1
  Protein heterodimerization activityBCL2NPR3; SDCBP2
  Sequence-specific DNA bindingBCL2SIX2
  Protein kinase bindingPAX6
  Transcription regulatory region DNA bindingERBB4; TCF21
  Protein complex bindingSIX2
  Protein dimerization activityTCF21
  Identical protein bindingBCL2MMP9; RB1
  Protein homodimerization activityERBB4; BCL2SDCBP2
  Protein C-terminus bindingSDCBP2
Cellular component
  NucleusERBB4; PAX6; SALL4; NKX6-2; BCL3; DOCK1; MEIS2; ZIC4; TCF21; ZBTB32; BCL2PRDM8; SIX2; RB1
  CytosolERBB4
  CytoplasmERBB4; PAX6; SALL4; BCL3; DOCK1; BCL2; BDNFSDCBP2; DNAJB1
  NucleoplasmERBB4; ZBTB32RB1
  Golgi apparatusSTC2
  Perinuclear region of cytoplasmBCL3; BDNFNPTX1
  MitochondrionERBB4; BCL2SLC25A42
  NucleolusERBB4; PAX6DNAJB1; RB1
  Transcription factor complexLRFN2
  Cell junctionPCDH
  MembraneERBB4; DOCK1; BCL2SLC16A6
  Nuclear chromatinPAX6

[i] GO, Gene Ontology.

KEGG Pathway-based analyses revealed that 43 signaling pathways involved 5,195 differentially methylated genes, and these genes were significantly enriched in specific pathways, including the cancer, metabolic, mitogen-activated protein kinase (MAPK), calcium, Wnt, hepatitis C, Erb-B2 receptor tyrosine kinase (ErbB), transforming growth factor (TGF)-β, vascular endothelial growth factor (VEGF), p53 and Notch signaling pathways (Table VIII).

Table VIII.

Kyoto encyclopedia of genes and genomes pathway analysis of differentially methylated genes.

Table VIII.

Kyoto encyclopedia of genes and genomes pathway analysis of differentially methylated genes.

PathwayNumber of differentially methylated genes (n)
Pathways in cancer309
Focal adhesion186
MAPK signaling pathway251
Wnt signaling pathway143
Axon guidance120
TGF-β signaling pathway  81
Basal cell carcinoma  55
Regulation of actin cytoskeleton191
Colorectal cancer  61
Adherens junction  71
Chronic myeloid leukemia  71
ECM-receptor interaction  81
Endocytosis186
Pyrimidine metabolism  97
Non-small cell lung cancer  57
Hedgehog signaling pathway  54
Neurotrophin signaling pathway116
Glioma  63
Endometrial cancer  52
VEGF signaling pathway  71
ErbB signaling pathway  80
Small cell lung cancer  80
Lysosome110
Metabolic pathways964
Calcium signaling pathway159
Purine metabolism150
Ubiquitin mediated proteolysis128
Insulin signaling pathway128
Notch signaling pathway  45
Protein processing in endoplasmic reticulum156
RNA polymerase  32
Hepatitis C116
Renal cell carcinoma  61
Aminoacyl-tRNA biosynthesis  42
B cell receptor signaling pathway  69
Thyroid cancer  29
Melanoma  65
Oocyte meiosis103
Adipocytokine signaling pathway  64
Melanogenesis  94
Vascular smooth muscle contraction115
Selenocompound metabolism  26
p53 signaling pathway  63

Discussion

DNA methylation is the main epigenetic modification and regulator of gene expression in humans. Aberrant changes in genomic methylation patterns have been observed in many cancer cell lines; these are regarded as the major type of molecular aberration in malignancies (7,8). Previous studies have evaluated aberrant DNA methylation in HCC by analyzing tumor tissues and adjacent non-tumor tissues (14,2123). Their main aims were to identify novel potential biomarkers for the diagnosis of HCC or to study the associations between methylation and cirrhosis-associated HCC; the study design and enrolment criteria differed when selecting various patients for study. In addition, variations in ethnicity and the effects of the cutting edge of adjacent non-tumor tissues may lead to differing results observed across the different studies. Although previous studies have indicated a few novel DNA methylation markers that are associated with HCC, specific DNA methylation patterns associated with the progression of HCC and alterations in methylation between HCC and normal liver cells have yet to be identified. In the present study, Illumina Infinium HumanMethylation 450K BeadChip was used to identify global DNA methylation profiles in Huh7 and L02 cells.

In the present study, a total of 102,254 differentially methylated CpG sites were detected across the whole-genome of Huh7 and L02 cells; more hypermethylated CpG sites (62,702; 61.3%) were observed than hypomethylated (39,552; 38.7%) CpG sites. The results indicated that within Huh7 cells, aberrant DNA methylation was a very common event and that hypermethylation of CpG sites occurred more frequently than hypomethylation. In addition, stringent criteria were employed to select the significantly differentially methylated CpG sites, genes and DMRs. Finally, 5,285 (66.5%) significantly hypermethylated and 2,659 (33.5%) hypomethylated CpG sites were identified. It has been reported that, in many types of diseases including cancers, aberrant DNA methylation is a common event, particularly aberrant DNA methylation of CpG islands or within promoter regions, which are associated with tumor suppressor gene inactivation or oncogene activation (16). The results of the present study indicated that within a CpG island, a greater number of significantly hypermethylated CpG sites (1,544) were observed than significantly hypomethylated CpG sites (1,201). This result is consistent with previous HCC genome-wide methylation studies (14,2427). Yates et al (18) and Dudziec et al (19) demonstrated that aberrant DNA methylation occurs in CpG islands, but can also be detected in the regions adjacent to CpG islands, CpG shores and CpG shelves (15), and may lead to tumorigenesis (20,28,29). The present study also demonstrated these points; significantly hypermethylated CpG sites in the CpG shores regions were more abundant than significantly hypomethylated CpG sites (1,137 to 632). In addition, in the CpG shelf regions, the significantly hypermethylated CpG sites were more frequent than significantly hypomethylated CpG sites (655 to 133).

DMRs are stretches of DNA in the genome. Varied DNA methylation patterns are seen between different organisms, and adjacent sites or a group of sites in proximity to each other tend to have different methylation patterns between different diseases (30). DMRs are associated with many diseases including several types of cancer (31). There are also many types of DMRs: Tissue-specific DMRs, cDMRs, rDMRs, imprinting-specific DMRs and aging-specific DMRs (20). In the present study, there were 390 differentially hypermethylated CpG sites located within DMRs, 233 (59.7%) were in island regions, 119 (30.5%) were in shore regions, and 15 (3.8%) were in shelf regions. In addition, there were 208 differentially hypomethylated CpG sites located within DMRs, 87 (41.8%) were in CpG island regions, 89 (42.8%) were in shore regions and 6 (2.9%) were in shelf regions. These results indicated that within HCC cells, aberrant DNA methylation may occur within CpG shore regions, which can also cause DNA transcriptional silencing and inactivation of gene function. Hepatocarcinogenesis was also associated with genomic instability and inactivation of gene function; the results of the present study concerning DMRs suggests that aberrant methylation within these sites may be an important epigenetic mechanism associated with hepatocarcinogenesis. These results may provide more information regarding the associations between HCC and aberrant DNA methylation.

Furthermore, the present study listed the top 20 significantly hyper- and hypo-methylated CpG sites, and genes in DMRs within Huh7 cells compared with L02 cells. The top 20 significantly hypermethylated genes, which were high-ranking with notable differences in the absolute value of β-difference, included the following: ERBB4, paired box 6, splat like transcription factor 4, protocadherin (PCDH)-8, NK2 homeobox 6, B-cell lymphoma (BCL)-3, glypican 6, family with sequence similarity 196 member A, dedicator of cytokinesis 1, Meis homeobox 2, Zic family member 4, transcription factor 21, zinc finger and BTB domain containing 32, leucine rich repeat and fibronectin type III domain containing 2, BCL2, PCDH9, DPY30 domain-containing protein 2, zinc finger CCHC-type containing 24, brain-derived neurotrophic factor, cg00057722 and cg08640046. The functional role of these genes in HCC requires further study. The top 20 significant differentially hyper- and hypo-methylated genes from GO enrichment were also listed. These genes, which were located within DMRs, were mainly associated with ‘cell differentiation development’, ‘transcription factor activity’, ‘sequence-specific DNA binding’, ‘cellular development process’ and ‘cell junction’.

Additionally, through GO enrichment analysis, the present study revealed that aberrant DNA methylation in HCC was associated with cell differentiation and proliferation, and through KEGG pathway analysis, 43 signaling pathways associated with HCC were identified, including pathways in cancer, MAPK signaling, Wnt signaling, VEGF signaling and p53 signaling pathways. Previous studies have demonstrated that aberrant DNA hypermethylation can downregulate the expression of cell cycle inhibitors, p16INK4A, p53 and factors involved in TGF-β/mothers against decapentaplegic signaling (32,33). Thus far, researchers have revealed that the inactivation of Wnt pathway-associated antagonists is linked to the aberrant DNA hypermethylation of some genes (34,35). Activation of the ERB receptor and MAPK signaling pathways, as well as the regulation of epigenetic proteins that were previously demonstrated to promote cancer growth and metastasis, have been reported to be possible candidate targets for anticancer treatment in multiple types of cancer, including HCC (32,36).

In addition, HCC cells may escape or become tolerant to chemotherapy via various mechanisms, therefore, identifying novel drugs is very important for the future therapy of HCC. The application of inhibitors of DNA methylated drugs in the treatment of cancer has gradually attracted the attention of researchers (37), including 5-azacytidine (5-aza-C), decitabine (5-aza-2′-deoxycytidine, 5-aza-dC), 1-β-D-arabinofuranosyl-5-azacytosine, dihydro-5-azacytosine (38), SGI-110 (previously known as S110), a dinucleotide of 5-aza-2′-deoxycytidine and deoxyguanosine, containing 5-azaCdR moiety, which has been revealed to be very effective in inhibiting DNA methylation, though its stability and cytotoxicity are comparable to that of decitabine (39), and a non-nucleoside DNA methyltransferase inhibitor, SGI-1027 (40,41). To the best of our knowledge, there have been only a few studies investigating the effects of demethylation agents on HCC in vitro.

In conclusion, the present study detected genome-wide DNA methylation patterns occurring in Huh7 cells, and identified numerous differentially hypo- and hypermethylated CpG sites, genes, DMRs and signaling pathways associated with HCC. Additionally, the diversity in methylation within Huh7 cells was also observed. The results of the present study may provide important information regarding the molecular mechanisms underlying methylation in Huh7 cells, which may be useful in future research into the underlying mechanisms associated with HCC. In addition, HCC cells may escape or develop tolerance to chemotherapy via various mechanisms, therefore, identifying novel drugs is very important for future therapies of HCC. The application of inhibitors of DNA methylation for the treatment of cancer has gradually attracted more attention within the field (37), and there have been a few studies investigating the effects of demethylation agents in HCC in vitro. The results of the present study may provide a useful basis for future research into effective HCC therapies.

Acknowledgements

Not applicable.

Funding

The present study was supported by The Science and Technology project of Shenyang (grant no. F13-212-9-00).

Availability of data and materials

The analyzed datasets generated during this study are available from the corresponding author on reasonable request.

Authors' contributions

JZ conceived and designed the study. NS, CZ, YS, BZ and BC performed the experiments. NS and AJ analyzed the data. NS wrote the 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.

Glossary

Abbreviations

Abbreviations:

PLC

primary liver cancer

HCC

hepatocellular carcinoma

DMR

differentially methylated regions

GO

Gene Ontology

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Zuo TT, Zheng RS, Zhang SW, Zeng HM and Chen WQ: Incidence and mortality of liver cancer in China in 2011. Chin J Cancer. 34:562015. View Article : Google Scholar : PubMed/NCBI

3 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Tanaka M, Katayama F, Kato H, Tanaka H, Wang J, Qiao YL and Inoue M: Hepatitis B and C virus infection and hepatocellular carcinoma in China: A review of epidemiology and control measures. J Epidemiol. 21:401–416. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Panayiotidis MI: Cancer epigenetics as biomarkers of clinical significance. Cancer Lett. 342:168–169. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Udali S, Guarini P, Moruzzi S, Ruzzenente A, Tammen SA, Guglielmi A, Conci S, Pattini P, Olivieri O, Corrocher R, et al: Global DNA methylation and hydroxymethylation differ in hepatocellular carcinoma and cholangiocarcinoma and relate to survival rate. Hepatology. 62:496–504. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Jones PA and Takai D: The role of DNA methylation in mammalian epigenetics. Science. 293:1068–1070. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Esteller M: Epigenetics in cancer. N Engl J Med. 358:1148–1159. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Jain S, Xie L, Boldbaatar B, Lin SY, Hamilton JP, Meltzer SJ, Chen SH, Hu CT, Block TM, Song W and Su YH: Differential methylation of the promoter and first exon of the RASSF1A gene in hepatocarcinogenesis. Hepatol Res. 45:110–123. 2015. View Article : Google Scholar

10 

Hinrichsen I, Kemp M, Peveling-Oberhag J, Passmann S, Plotz G, Zeuzem S and Brieger A: Promoter methylation of MLH1, PMS2, MSH2 and p16 is a phenomenon of advanced-stage HCCs. PLoS One. 9:e844532014. View Article : Google Scholar : PubMed/NCBI

11 

Xu B, Nie Y, Liu X, Feng S, Yang Z, Wang Z, Zheng Q and Luo X: Quantitative analysis of APC promoter methylation in hepatocellular carcinoma and its prognostic implications. Oncol Lett. 7:1683–1688. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Jain S, Chen S, Chang KC, Lin YJ, Hu CT, Boldbaatar B, Hamilton JP, Lin SY, Chang TT, Chen SH, et al: Impact of the location of CpG methylation within the GSTP1 gene on its specificity as a DNA marker for hepatocellular carcinoma. PLoS One. 7:e357892012. View Article : Google Scholar : PubMed/NCBI

13 

Qu Z, Jiang Y, Li H, Yu DC and Ding YT: Detecting abnormal methylation of tumor suppressor genes GSTP1, P16, RIZ1, and RASSF1A in hepatocellular carcinoma and its clinical significance. Oncol Lett. 10:2553–2558. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Shen J, Wang S, Zhang YJ, Wu HC, Kibriya MG, Jasmine F, Ahsan H, Wu DP, Siegel AB, Remotti H and Santella RM: Exploring genome-wide DNA methylation profiles altered in hepatocellular carcinoma using Infinium HumanMethylation 450 BeadChips. Epigenetics. 8:34–43. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Bibikova M, Barnes B, Tsan C, Ho V, Klotzle B, Le JM, Delano D, Zhang L, Schroth GP, Gunderson KL, et al: High density DNA methylation array with single CpG site resolution. Genomics. 98:288–295. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Esteller M: Epigenetic gene silencing in cancer: The DNA hypermethylome. Hum Mol Genet. 16:R50–R59. 2007. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Yates DR, Rehman I, Meuth M, Cross SS, Hamdy FC and Catto JW: Methylational urinalysis: A prospective study of bladder cancer patients and age stratified benign controls. Oncogene. 25:1984–1988. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Dudziec E, Miah S, Choudhry HM, Owen HC, Blizard S, Glover M, Hamdy FC and Catto JW: Hypermethylation of CpG islands and shores around specific microRNAs and mirtrons is associated with the phenotype and presence of bladder cancer. Clin Cancer Res. 17:1287–1296. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, et al: The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 41:178–186. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Nishida N, Kudo M, Nagasaka T, Ikai I and Goel A: Characteristic patterns of altered DNA methylation predict emergence of human hepatocellular carcinoma. Hepatology. 56:994–1003. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Shen J, Wang S, Zhang YJ, Kappil M, Wu HC, Kibriya MG, Wang Q, Jasmine F, Ahsan H, Lee PH, et al: Genome-wide DNA methylation profiles in hepatocellular carcinoma. Hepatology. 55:1799–1808. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Udali S, Guarini P, Ruzzenente A, Ferrarini A, Guglielmi A, Lotto V, Tononi P, Pattini P, Moruzzi S, Campagnaro T, et al: DNA methylation and gene expression profiles show novel regulatory pathways in hepatocellular carcinoma. Clin Epigenetics. 7:432015. View Article : Google Scholar : PubMed/NCBI

24 

Gao W, Kondo Y, Shen L, Shimizu Y, Sano T, Yamao K, Natsume A, Goto Y, Ito M, Murakami H, et al: Variable DNA methylation patterns associated with progression of disease in hepatocellular carcinomas. Carcinogenesis. 29:1901–1910. 2008. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Ammerpohl O, Pratschke J, Schafmayer C, et al: Distinct DNA methylation patterns in cirrhotic liver and hepatocellular carcinoma. Int J Cancer. 130:1319–1328. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Kohles N, Nagel D, Jüngst D, Durner J, Stieber P and Holdenrieder S: Prognostic relevance of oncological serum biomarkers in liver cancer patients undergoing transarterial chemoembolization therapy. Tumour Biol. 33:33–40. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, Herb B, Ladd-Acosta C, Rho J, Loewer S, et al: Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet. 41:1350–1353. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Ogoshi K, Hashimoto S, Nakatani Y, Qu W, Oshima K, Tokunaga K, Sugano S, Hattori M, Morishita S and Matsushima K: Genome-wide profiling of DNA methylation in human cancer cells. Genomics. 98:280–287. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Rakyan VK, Down TA, Balding DJ and Beck S: Epigenome-wide association studies for common human diseases. Nat Rev Genet. 12:529–541. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Weber M, Davies JJ, Wittig D, Oakeley EJ, Haase M, Lam WL and Schübeler D: Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nat Genet. 37:853–862. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Calvisi DF, Pascale RM and Feo F: Dissection of signal transduction pathways as a tool for the development of targeted therapies of hepatocellular carcinoma. Rev Recent Clin Trials. 2:217–236. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Kuo KK, Jian SF, Li YJ, Wan SW, Weng CC, Fang K, Wu DC and Cheng KH: Epigenetic inactivation of transforming growth factor-beta1 target gene HEYL, a novel tumor suppressor, is involved in the P53-induced apoptotic pathway in hepatocellular carcinoma. Hepatol Res. 45:782–793. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Umer M, Qureshi SA, Hashmi ZY, Raza A, Ahmad J, Rahman M and Iqbal M: Promoter hypermethylation of Wnt pathway inhibitors in hepatitis C virus-induced multistep hepatocarcinogenesis. Virol J. 11:1172014. View Article : Google Scholar : PubMed/NCBI

35 

Ding SL, Yang ZW, Wang J, Zhang XL, Chen XM and Lu FM: Integrative analysis of aberrant Wnt signaling in hepatitis B virus-related hepatocellular carcinoma. World J Gastroenterol. 21:6317–6328. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Stefanska B, Cheishvili D, Suderman M, Arakelian A, Huang J, Hallett M, Han ZG, Al-Mahtab M, Akbar SM, Khan WA, et al: Genome-wide study of hypomethylated and induced genes in patients with liver cancer unravels novel anticancer targets. Clin Cancer Res. 20:3118–3132. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Yan W, Herman JG and Guo M: Epigenome-based personalized medicine in human cancer. Epigenomics. 8:119–133. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Ghoshal K and Bai S: DNA methyltransferases as targets for cancer therapy. Drugs Today (Barc). 43:395–422. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Yoo CB, Jeong S, Egger G, Liang G, Phiasivongsa P, Tang C, Redkar S and Jones PA: Delivery of 5-aza-2′-deoxycytidine to cells using oligodeoxynucleotides. Cancer Res. 67:6400–6408. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Datta J, Ghoshal K, Denny WA, Gamage SA, Brooke DG, Phiasivongsa P, Redkar S and Jacob ST: A new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation. Cancer Res. 69:4277–4285. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Gros C, Fleury L, Nahoum V, Faux C, Valente S, Labella D, Cantagrel F, Rilova E, Bouhlel MA, David-Cordonnier MH, et al: New insights on the mechanism of quinoline-based DNA Methyltransferase inhibitors. J Biol Chem. 290:6293–6302. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 18 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun N, Zhang J, Zhang C, Shi Y, Zhao B, Jiao A and Chen B: Using Illumina Infinium HumanMethylation 450K BeadChip to explore genome‑wide DNA methylation profiles in a human hepatocellular carcinoma cell line. Mol Med Rep 18: 4446-4456, 2018
APA
Sun, N., Zhang, J., Zhang, C., Shi, Y., Zhao, B., Jiao, A., & Chen, B. (2018). Using Illumina Infinium HumanMethylation 450K BeadChip to explore genome‑wide DNA methylation profiles in a human hepatocellular carcinoma cell line. Molecular Medicine Reports, 18, 4446-4456. https://doi.org/10.3892/mmr.2018.9441
MLA
Sun, N., Zhang, J., Zhang, C., Shi, Y., Zhao, B., Jiao, A., Chen, B."Using Illumina Infinium HumanMethylation 450K BeadChip to explore genome‑wide DNA methylation profiles in a human hepatocellular carcinoma cell line". Molecular Medicine Reports 18.5 (2018): 4446-4456.
Chicago
Sun, N., Zhang, J., Zhang, C., Shi, Y., Zhao, B., Jiao, A., Chen, B."Using Illumina Infinium HumanMethylation 450K BeadChip to explore genome‑wide DNA methylation profiles in a human hepatocellular carcinoma cell line". Molecular Medicine Reports 18, no. 5 (2018): 4446-4456. https://doi.org/10.3892/mmr.2018.9441