Open Access

mRNA expression and DNA methylation analysis of the inhibitory mechanism of H2O2 on the proliferation of A549 cells

  • Authors:
    • Yepeng Li
    • Zhongheng Wei
    • Shiqing Huang
    • Bo Yang
  • View Affiliations

  • Published online on: September 23, 2020     https://doi.org/10.3892/ol.2020.12151
  • Article Number: 288
  • Copyright: © Li 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

Reactive oxygen species, particularly hydrogen peroxide (H2O2), can induce proliferation inhibition and death of A549 cells via oxidative stress. Oxidative stress has effect on DNA methylation. Oxidative stress and DNA methylation feature a common denominator: The one carbon cycle. To explore the inhibitory mechanism of H2O2 on the proliferation of lung cancer cells, the present study analysed the mRNA expression and methylation profiles in A549 cells treated with H2O2 for 24 h, as adenocarcinoma is the most common pathological type of lung cancer. The DNA methylation profile was constructed using reduced representation bisulphite sequencing, which identified 29,755 differentially methylated sites (15,365 upregulated and 14,390 downregulated), and 1,575 differentially methylated regions located in the gene promoters were identified using the methylKit. Analysis of the assocaition between gene expression and methylation levels revealed that several genes were downregulated and hypermethylated, including cyclin‑dependent kinase inhibitor 3, denticleless E3 ubiquitin protein ligase homolog, centromere protein (CENP)F, kinesin family member (KIF)20A, CENPA, KIF11, PCNA clamp‑associated factor and GINS complex subunit 2, which may be involved in the inhibitory process of H2O2 on the proliferation of A549 cells.

Introduction

Reactive oxygen species (ROS), whose principal components include superoxide anion (O2), hydrogen peroxide (H2O2) and hydroxyl radical, can be generated in all aerobic cells (1). At normal concentrations, ROS can be regarded as signalling molecules, whereas high concentrations of ROS are cytotoxic, often leading to cell death (2). Lung cancer is one of the most malignant tumours in the respiratory tract, and 85% of lung cancer cases are non-small cell lung cancer (NSCLC) (3). Previous reports have revealed that H2O2 may inhibit the proliferation of A549 lung cancer cells via oxidative stress (4,5). A recent study demonstrated that in lung cancer cells treated with H2O2, MAPK inhibitors (mainly the JNK inhibitor) increased O2 and glutathione depletion, thus leading to cell death (6). Furthermore, oligomeric proanthocyanidins have been reported to protect A549 cells from oxidative stress induced by H2O2 through the Nrf2-antioxidant responsive element signalling pathway (7).

DNA methylation is a biochemical process that occurs with the addition of methyl groups to cytosines adjacent to guanines (CpG) by DNA methyltransferases (8). Although DNA methylation is mainly located in the gene promoter region, it can also occur along the gene sequence, and different locations correspond to different effects on the gene expression level (9,10). DNA methylation can regulate gene expression, as well as maintain the DNA structure and control the transposable elements; therefore, it is often associated with various processes, such as tissue differentiation and disease susceptibility (11,12). Anglim et al (13) identified several useful DNA methylation markers for the early detection of squamous cell lung cancer using DNA methylation profiles. Del Real et al (14) identified two long non-coding RNAs that may promote the pathological process of non-small cell lung cancer by analysing gene expression and methylation profiles.

In the present study, genome-wide analyses of mRNA expression and DNA methylation were systematically performed to evaluate the inhibitory mechanism of H2O2 on the proliferation of A549 cells. Association analysis of differentially expressed genes (DEGs) and differentially methylated regions (DMRs) revealed several genes that may be associated with the inhibitory process of H2O2 on the proliferation of A549 cells. The present study may provide novel insights into the molecular mechanisms underlying the inhibitory effects of H2O2 on the proliferation of A549 cells, and could contribute to the identification of novel prognostic or diagnostic markers for lung cancer.

Materials and methods

Cell culture

A549 lung cancer cells were purchased from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. A549 cells were cultured in RPMI-1640 medium containing 10% FBS (Ausbian, http://www.viansaga.com/ausbian.html), 2 mM L-glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin at 37°C in a 5% CO2 incubator. Cells treated only with culture medium were used as the control group, whereas cells treated with H2O2 for 24 h at 37°C with a final concentration of 200 µM constituted the experimental group. The original images of H2O2-treated and untreated A549 cells were captured using a light microscope. After counting, the cells in both groups were inoculated on corresponding culture plates for further analysis, with ~106 cells used for the gene expression microarray and reduced representation bisulphite sequencing (RRBS).

MTT assay

Cell viability was assessed using the MTT assay. A549 cells were seeded at a density of 4,000 cells/well in a 96-well plate and incubated with H2O2 at 37°C for 24 h. Subsequently, 20 µl MTT aqueous solution (5 mg/ml) was added to each well and the plates were incubated for 4 h at 37°C in a humidified atmosphere containing 5% CO2. The culture medium was aspirated and 100 µl DMSO was added to each well. Finally, ELX800 UV universal microplate reader (Bio-Tek Instruments Inc.) was used to determine the absorbance at 570 nm with a reference wavelength of 630 nm.

Half maximal inhibitory concentration (IC50) calculation

The IC50 value, which is the concentration that inhibited cell viability to 50% of the control, was used to evaluate the inhibitory effect of H2O2 on the proliferation of A549 cells. GraphPad Prism (version 5; GraphPad Software, Inc.) was used to calculate the IC50 value from the best-fit of the Hill slope curve using nonlinear regression analysis: M = 100/1 + 10^[(LogIC50-N) × HillSlope)], where N is the log of the dose, M is the growth inhibition value normalised to the control and HillSlope is the unitless slope factor or Hill slope.

ROS assay

The Fluorometric Intracellular ROS Assay kit (Sigma-Aldrich; Merck KGaA) was used to detect ROS produced by H2O2-treated A549 cells, according to the manufacturer's protocol. The assay was performed in 96-well plate and read using a fluorescence microplate reader resulting in a fluorometric product (l ex = 490/l em = 520 nm) proportional to the amount of ROS present.

Identification of DEGs and RRBS assay

The treated cells were collected by centrifugation at 8,000 × g at 4°C for 1 min separately, the DNA was extracted using the cetyltrimethylammonium bromide method according to the DNA extract kit manufacurer's protocol (Roche Diagnostics) and the RNA was extracted using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.), according to the operating instructions.

RRBS assay

After genomic DNA was extracted from the samples, DNA concentration and integrity were detected using a NanoDrop spectrophotometer (NanoDrop; Thermo Fisher Scientific, Inc.) and 1% agarose gel electrophoresis, respectively. The DNA libraries for bisulphite sequencing were prepared as previously described (15). Briefly, genomic DNA was fragmented into 100–300 bp via sonication with a duration 30 sec at 30 sec intervals for 15 cycles at 20 kHz and 4°C (Covaris M220) and purified using the MiniElute PCR Purification kit (Qiagen, Inc.). The DNA fragments were end-repaired and a single ‘A’ nucleotide was added to the 3′-end of the blunt fragments. Subsequently, the genomic fragments were ligated to methylated sequencing adapters. Fragments with adapters were bisulphite-converted using the Methylation-Gold kit (Zymo Research Corp.). Finally, the converted DNA fragments were amplified via PCR and sequenced using Illumina HiSeq™ 2500 by Shanghai GeneChem Co., Ltd.

Identification of DEGs: Genome-wide expression profiling analysis was performed by Genminix Informatics Co., Ltd. using GeneChip ClarionD Array (Affymetrix; Thermo Fisher Scientific, Inc.). Briefly, total RNA was separately extracted from 10 individual samples using the RNeasy Mini kit (Qiagen, Inc.). Double-stranded cDNA was then synthesized, labeled and hybridized to the gene chip. After hybridization and washing, the slides were scanned with the GeneChip Operating Software version 4.0 (Affymetrix; Thermo Fisher Scientific, Inc.). Raw data extraction and subsequent data processing were performed using the Affymetrix GeneChip Operating Software.

The significant differential analysis was conducted via unpaired Student's t-tests to identify the DEGs between H2O2-treated A549 cells and the control group. Only genes that met the cut-off criteria (adjusted P<0.05 and |Fold Change| >2) were regarded as significantly differentially expressed. Additionally, the DEGs were clustered using the hierarchical clustering method and Euclidean distance was chosen as a measure of the distance between the samples. The clustering was implemented using the heatmap.2 function in the gplot R package version 3.6.3 (https://www.rdocumentation.org/packages/gplots/).

Quantitative gene expression analysis via reverse transcription-quantitative (RT-q)PCR

RT-qPCR assays were performed to validate the microarray data. Total RNA was extracted from A549 cells using TRIzol reagent. Single-strand cDNA was synthesized from 1 mg total RNA using the Prime-Script™ Reagent kit with gDNA Eraser (TransGen Biotech Co., Ltd.). The steps were 70°C for 5 min, 37°C for 5 min, 42°C for 60 min and 70°C for 10 min to end the reaction. To detect the mRNA expression levels of the DEGs, qPCR (TaqMan kit; Thermo Fisher Scientific, Inc.) was conducted on a Q1 PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primer sequences are listed in Table I. The following thermocycling conditions were applied: 95°C for 30 sec, followed by 40 cycles at 95°C for 5 sec, 60°C for 15 sec and 72°C for 10 sec and 72°C for 7 min for final extension. Data were presented as a relative average value ± SEM after normalization with the average value of the housekeeping gene GAPDH. Direct comparison was performed between the control and H2O2-treated groups for the same gene and 2−ΔΔCq method was used for the relative quantification of gene expression (16).

Table I.

Primers used for reverse transcription-quantitative PCR.

Table I.

Primers used for reverse transcription-quantitative PCR.

Primer nameSequence (5′→3′)
(h)CDKN3-138-F ACAGAAGGACGAACCAGTGAG
(h)CDKN3-138-R TTGTATTGAACTGGGCGGCT
(h)CENPF-99-F CGTCCCCGAGAGCAAGTTTAT
(h)CENPF-99-R TGTAGGCAGCCCTTCTTTCC
(h)HIST1H2BM-151-F CGACCATCACTTCGAGGGAG
(h)HIST1H2BM-151-R GTCACGGCGGAACTGTTACT
GAPDH-127-F (h) CCAGGTGGTCTCCTCTGA
GAPDH-127-R (h) GCTGTAGCCAAATCGTTGT

[i] F, forward; R, reverse; h, human; CDKN3, cyclin-dependent kinase inhibitor 3; CENPF, centromere protein F; HIST1H2BM, histone cluster 1 H2B family member M.

Functional enrichment analysis of DEGs

Gene Ontology (GO) analysis, which organises genes into hierarchical categories and identifies the gene regulatory network based on biological processes (BP), molecular functions (MF) and cellular components (CC), was used for the genes and gene products. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was used to identify significant pathways of the genes and enriched gene products. The Database for Annotation, Visualization and Integrated Discovery (DAVID; https:/david.ncifcrf.gov/) was used for GO and KEGG functional annotation of genes with P<0.05.

Analysis of the bisulphite sequencing data

Flexbar version 3.0 software (https://github.com/seqan/flexbar) was used to guarantee the high quality of the sequence reads in three steps: In step 1, the low-quality base calls were trimmed, step 2 removed the adaptor and step 3 filtered the PCR duplication. The absolute methylation level was measured by the enrichment of CpG fractions in the genome after treatment with sodium bisulfite. The microarray reads were aligned to the reference sequence and the methylation called base-by-base in the reads with a coverage >10 using the software BSMAP (whole genome bisulphite sequence MAPing program, http://code.google.com/p/bsmap/) Subsequently, the proportion of cytosine and thymine bases was calculated at CG positions among the bisulphite sequencing reads aligned to the reference sequence as the methylation level. The DMRs were identified using the methylKit R package (from R version 3.6.3).

Association analysis between gene expression levels and DNA methylation

Genes with significant changes in expression (P<0.05) were categorised into four groups based on their changes in gene expression (E) and methylation levels (M): Group 1 corresponds to high methylation and upregulated genes (log2 fold change >0.5; E+ and M+); Group 2 to high methylation and downregulated genes (log2 fold change <1.5; E and M+); Group 3 to low methylation and upregulated genes (log2 fold change >1.5; E+ and M); and Group 4 to low methylation and downregulated genes (log2 fold change ≤1.5; E and M). Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancer-pku.cn) was used to analyse the expression levels and the effects of genes on survival with methylation level changes in patients with lung adenocarcinoma (LUAD). GEPIA is a newly developed interactive web server for analysing the RNA sequencing expression data of 9,736 tumours and 8,587 normal samples from The Cancer Genome Atlas and the Genotype-Tissue Expression projects, using a standard processing pipeline.

Statistical analysis

GraphPad Prism (version 5; GraphPad Software, Inc.) was used for statistical analysis. Data are expressed as the mean ± standard error of the mean (SEM). The mean was obtained from three repeats. Differences between two groups were analysed using unpaired Student's t-tests, whereas ≥3 groups were analysed via one-way ANOVA followed by Tukey's post hoc test. In the Kaplan-Meier analysis log-rank test was conducted to assess the survival, or Cramer-von Mises when crossover occurred. All tests performed were two-sided. P<0.05 was considered to indicate a statistically significant difference.

Results

Inhibitory effects of H2O2 on the proliferation of A549 cells

The inhibitory effect of H2O2 on the proliferation of A549 cells was concentration-dependent (Fig. 1A), with an IC50 of 1.09 mg/l. In addition, the ROS production of A549 cells was markedly increased compared with that of the control group (Fig. 1B). The original images of H2O2-treated and untreated A549 cells were captured using a light microscope (Fig. 1C and D) further confirmed that H2O2 inhibited the proliferation of A549 cells.

Analysis of the mRNA expression profiles

A total of 1,026 DEGs, 261 upregulated and 765 downregulated, were identified in H2O2-treated A549 cells compared with the control group (Fig. 2A). The heatmap of hierarchical clustering of the DEGs is shown in Fig. 2B. There were 261 upregulated and 766 downregulated genes between H2O2-treated A549 cells and the control group. The online analysis tool DAVID was used to identify statistically significant enriched GO terms for the DEGs between the H2O2-treated and control groups (Fig. 2C), indicating that the DEGs were mainly enriched in BP, such as ‘cell cycle’, ‘cell cycle process’ and ‘mitotic cell cycle’. Regarding CC, they were enriched in ‘chromosome’, ‘nuclear chromosome’ and ‘chromosomal region’. In addition, MF analysis revealed that the genes were enriched in ‘protein dimerization activity’, ‘protein heterodimerization activity’, ‘adenyl nucleotide binding’ and ‘macromolecular complex binding’. KEGG analysis revealed that the DEGs were enriched in ‘systemic lupus erythematosus’, ‘cell cycle’, ‘DNA replication’ and ‘homologous recombination’ (Fig. 2D). qPCR was used to verify the expression levels of DEGs in A549 cells (Fig. 3). Association analysis of the gene expression and methylation levels revealed that several genes were downregulated and hypermethylated, including cyclin-dependent kinase inhibitor 3, denticleless E3 ubiquitin protein ligase homolog, centromere protein (CENP)F, kinesin family member (KIF)20A, CENPA, KIF11, PCNA clamp associated factor (PAF) and GINS complex subunit 2. Therefore, these genes were chosen to be verified by qPCR. Consistent with the gene expression microarray, the expression levels of cyclin-dependent kinase inhibitor 3 (CDKN3; NCBI_1033) and centromere protein (CENP)F (NCBI_1063) were significantly downregulated in H2O2-treated A549 cells compared with those in untreated cells (Fig. 3). The expression levels of H2B clustered histone 14 (H2BC14 NCBI_8342) were decreased slightly in H2O2-treated A549 cells, which was also consistent with the microarray results.

Analysis of the DNA methylation expression profiles

RRBS was used to annotate differentially methylated sites between H2O2-treated A549 cells and the control group. The CpGs were grouped based on the genomic position, with a similar distribution of CpGs in both groups (Fig. 4A). Additionally, the average methylation level was calculated, with a similar distribution in both groups (Fig. 4B). In total, 29,755 differentially methylated CpG sites were identified; 15,365 of them were more methylated and 14,390 were less methylated in the A549 cells treated with H2O2 compared with in the untreated cells. Among the differentially methylated CpG sites, the top 20 were annotated (Table II). The differences in methylation levels in the same region between two samples was calculated to identify the DMRs. The top 10 DMRs were annotated (Table III), suggesting that there was a difference in the methylation levels between the two groups. In addition, the analysis at the region level revealed that 1,575 DMRs occurred in the gene promoters. GO analysis revealed that the genes were enriched in the terms ‘extracellular space’, ‘extracellular matrix’, ‘neuron protein’ and ‘perikaryon’ (Table IV). KEGG pathway analysis revealed that the genes were enriched in ‘hsa04723: Retrograde endocannabinoid signalling’, ‘hsa05032: morphine addiction’ and ‘hsa04726: Serotonergic synapse’ (Table V).

Table II.

Top 20 hypermethylated and top 20 hypomethylated CpG sites between the H2O2-treated and control groups.

Table II.

Top 20 hypermethylated and top 20 hypomethylated CpG sites between the H2O2-treated and control groups.

ChromosomePositionStrandP-valueq valuemeth.diff (%)GeneGene typeFunction element
chr1778452227 2.56×10−24 1.99×10−2083.53NANAIntergenic
chr9140951580 1.95×10−21 6.20×10−1881.25CACNA1BProtein_codingGene
chr11973547+ 8.04×10−28 1.89×10−2379.07AP2A2Protein_codingGene
chr1241588198 7.38×10−19 1.04×10−1578.81RP11-527D7.1lincRNAGene
chr626210354+ 1.60×10−29 7.75×10−2577.46NANAIntergenic
chr1544068996+ 9.65×10−15 3.34×10−1276.54RP11-296A16.1Protein_codingGene
chr1119893199+ 1.90×10−17 1.70×10−1476.37NAV2Protein_codingGene
chr1665732093+ 5.75×10−14 1.50×10−1176.28NANAIntergenic
chr895654198+ 1.25×10−21 4.43×10−1875.61ESRP1Protein_codingGene
chrX133119214+ 3.80×10−18 4.25×10−1574.86GPC3Protein_codingGene
chrX153734771+ 2.39×10−29 1.04×10−2473.91FAM3AProtein_codingGene
chr7157697656+ 7.23×10−25 6.57×10−2173.88PTPRN2Protein_codingGene
chr1164659131 5.57×10−22 2.16×10−1873.82MIR194-2lincRNAGene
chr4131947017 9.96×10−17 6.84×10−14NANANAIntergenic
chr1778452271 3.26×10−19 5.17×10−1673.02NANAIntergenic
chr2061983851 7.26×10−15 2.61×10−1272.55CHRNA4Protein_codingGene
chr2467136 2.11×10−18 2.56×10−1572.25NANAIntergenic
chr7426119+ 2.46×10−12 3.61×10−1071.92NANAIntergenic
chr88632457 2.46×10−25 2.50×10−2171.11RP11-211C9.1lincRNAGene
chr1544069009+ 4.02×10−12 5.46×10−1070.47RP11-296A16.1Protein_codingGene
chr1634622413+ 8.19×10−20 1.57×10−16−87.5RP11-488I20.3lincRNAGene
chr332858837+ 3.20×10−28 8.63×10−24−83.04TRIM71Protein_codingPromoter
chr1634622419+ 2.00×10−17 1.77×10−14−81.25RP11-488I20.3lincRNAGene
chr9136150995+ 1.11×10−15 5.36×10−13−81.09ABO Processed_transcriptPromoter
chr1562110720 1.23×10−21 4.39×10−18−80.31NANAIntergenic
chr338071105+ 3.97×10−30 2.34×10−25−80.22PLCD1Protein_codingGene
chr191323858 5.41×10−28 1.34×10−23−79.82MUM1Protein_codingGene
chr1579092872 3.36×10−16 1.94×10−13−78.5ADAMTS7Protein_codingGene
chr1947979937 9.68×10−21 2.47×10−17−76.98KPTNProtein_codingGene
chr1939342337 3.42×10−22 1.41×10−18−76.4HNRNPLProtein_codingGene
chr5179228528 1.87×10−20 4.36×10−17−75.95MGAT4BProtein_codingGene
chr1216275907+ 1.71×10−18 2.16×10−15−75.06USH2AProtein_codingGene
chr3109051010 1.61×10−18 2.05×10−15−74.89DPPA4Protein_codingGene
chr481111777+ 1.42×10−19 2.50×10−16−73.33PRDM8Protein_codingGene
chr313816124+ 1.33×10−18 1.73×10−15−72.31NANAIntergenic
chr1079470698 1.63×10−20 3.87×10−17−72.08NANAIntergenic
chr14104940386+ 4.22×10−19 6.40×10−16−71.71NANAIntergenic
chr10123389019+ 4.70×10−16 2.57×10−13−71.57NANAIntergenic
chr1660247513 1.19×10−14 3.98×10−12−71.48NANAIntergenic
chr153098973+ 2.36×10−15 1.01×10−12−70.82FAM159AProtein_codingPromoter

[i] meth.diff, methylation difference; NA, not available.

Table III.

Top 10 hypermethylated and top 10 hypomethylated CpG regions between the H2O2-treated and control groups.

Table III.

Top 10 hypermethylated and top 10 hypomethylated CpG regions between the H2O2-treated and control groups.

ChromosomeStartEndP-valuemeth.diff (%)GeneGene typeFunction element
chr1035580013559000 3.29×10−5174.40NANAIntergenic
chr4110000000110000000 8.88×10−2770.70CCDC109BProtein_codingGene
chr166829300168294000 6.80×10−2468.70PLA2G15Protein_codingGene
chr158538300185384000 5.03×10−2167.80ALPK3Protein_codingGene
chr1776960017697000 1.36×10−2666.90DNAH2Protein_codingGene
chr51062000110621000 9.08×10−3665.00ANKRD33BProtein_codingGene
chr56863100168632000 3.05×10−2764.20CCDC125Protein_codingPromoter
chr191302500113026000 9.50×10−1963.50GCDHProtein_codingGene
chr206204400162045000 6.57×10−2861.20KCNQ2Protein_codingGene
chr1167050016706000 1.19×10−8960.10MRPL17Protein_codingPromoter
chr1711120011113000 1.15×10−20−72.40 Protein_codingGene
chr9126000000126000000 4.44×10−18−69.00DENND1AProtein_codingGene
chr158622000186221000 4.86×10−17−65.60AKAP13Protein_codingGene
chr116606100166062000 7.37×10−35−65.20TMEM151AProtein_codingGene
chr157241200172413000 1.35×10−20−65.10SENP8Protein_codingGene
chr16732300167324000 5.58×10−22−64.70WDR78Protein_codingGene
chr223757200137573000 2.11×10−17−64.10RP1-151B14.6AntisenseGene
chr7105000000105000000 2.25×10−23−63.80RINT1Protein_codingGene
chr166573100165732000 1.97×10−17−62.50NANAIntergenic
chr205047100150472000 3.02×10−16−61.70RP5-1112F19.2lincRNAGene

[i] meth.diff, methylation difference; NA, not available.

Table IV.

GO analysis of the genes with differentially methylated regions in the promoter of genes.

Table IV.

GO analysis of the genes with differentially methylated regions in the promoter of genes.

CategoryTermCountGenes
GOTERM_CC_DIRECT GO:0005615~extracellular space88GDF3, PXDN, NOG, SLURP1, LYPD3, IL16, KIAA0556, GDF6, LTBP4, EDN2, CRHBP, TNFSF14, TNFSF13, DLK1, MCF2L, MMRN2, SCT, GPC5, AZGP1, APOE, CETP, CFD, ACTN4, IL27, ZNF649, C10ORF99, CST1, MFGE8, CBR3, SSPO, CTSV, PROC, RETN, CBLN4, AMH, CHGA, THBD, GNB2, SERPINF1, F3, SERPINB8, CPXM1, ULBP2, NPPC, C1QL4, UBB, WNT9A, ADAMTS5, TF, RBP4, MFNG, PODN, PRTN3, NDP, CLU, DSCAML1, PF4, CXCL6, NRN1, CCL5, CHIT1, ADCYAP1, CPZ, AGT, LEFTY2, TFF2, VWC2L, ENTPD6, HGFAC, OLFM1, BMP4, FLRT3, BGLAP, PRSS57, HSPG2, TNFSF9, FRZB, KRT35, CLEC11A, MUC4, NBL1, TSLP, DKK3, PPIA, KRT78, LIPG, NRN1L, METRNL
GOTERM_CC_DIRECT GO:0031012~extracellular matrix26FGFR2, HIST1H4L, PXDN, PRTN3, LTBP4, NDP, ADAMTSL5, CLU, HSPG2, MMP17, MFGE8, EMILIN3, NDNF, COL5A1, RPS3, MMRN2, LAMA2, SERPINF1, APOE, F3, LEFTY2, HIST1H4E, ADAMTS10, TGFB1I1, B4GALT7, HSPA9
GOTERM_CC_DIRECTGO:0043005~neuron projection22GDI1, ACTN4, SLC6A12, ADCYAP1R1, SLC6A4, WRN, ATP13A2, SHANK2, CTSV, SHANK3, TRPM2, SLC32A1, ATP2B4, P2RX1, HDAC1, ARPC2, BCL11B, CHRNB4, ABAT, CYGB, NSMF, UBB
GOTERM_CC_DIRECT GO:0043204~perikaryon12SLC8A3, MAPK1, BGLAP, CRHBP, EFNA2, CNTNAP2, NSMF, NTSR1, CTSV, OLFM1, TRPM2, NEURL1
GOTERM_CC_DIRECT GO:0048471~perinuclear region of cytoplasm41SLC8A3, NANOS3, TF, MAL2, HIP1R, ABCD1, SLC39A13, SLC39A12, CLU, TRAPPC2L, CABP7, PLVAP, NXT2, SLK, NMRAL1, ARHGAP1, MMD2, NDRG2, EHD2, C9ORF24, CCAR1, DLG1, ACTN4, LDB3, CIDEB, ACKR3, TPD52L2, KAT5, PRKCD, MT1X, CHGA, SERPINF1, HDAC1, GNB2, VAMP8, TPPP, ATP9A, CDK2AP1, MEX3D, ABL1, NEURL1

[i] GO, Gene Ontology.

Table V.

KEGG pathway analysis of the genes with differentially methylated regions in the promoter of genes.

Table V.

KEGG pathway analysis of the genes with differentially methylated regions in the promoter of genes.

KEGG pathwayCountP-valueGenes
hsa04723:Retrograde endocannabinoid signaling13 7.62×10−4GABRD, GABRG2, GABRG3, CACNA1S, KCNJ3, GRM1, SLC32A1, GNG8, GNGT1, MAPK1, GNB2, FAAH, GNG4
hsa05032:Morphine addiction12≤0.001GNG8, GABRD, SLC32A1, GNGT1, GABRG2, GABRG3, GNB2, PDE1C, GRK4, PDE4D, GNG4, KCNJ3
hsa04726:Serotonergic synapse13≤0.001MAOA, SLC6A4, RAF1, CACNA1S, KCNJ3, GNG8, MAPK1, GNGT1, ALOX15, HTR1B, GNB2, GNG4, HTR2A
hsa04724:Glutamatergic synapse13≤0.001GRIK1, GRM1, KCNJ3, SHANK2, SHANK3, GNG8, GRM4, MAPK1, GNGT1, GNB2, PPP3CC, GNG4, SLC1A1
hsa04727:GABAergic synapse10≤0.001GNG8, GABRD, SLC32A1, GNGT1, GABRG2, GABRG3, GNB2, ABAT, GNG4, CACNA1S
hsa05020:Prion diseases5≤0.001C1QA, MAPK1, NCAM2, C1QB, CCL5

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

Association analysis of gene expression and methylation levels

The association analysis indicated that genes with a high methylation difference always resulted in a difference in the gene expression levels. Regarding hypermethylation located in the gene body, the top genes that showed greatest difference in methylation between groups included H2BC14 (NCBI_8342), CDKN3 (NCBI_1033), denticleless E3 ubiquitin protein ligase homolog (DTL; NCBI_51514) and CENPF (NCBI_1063) (data not shown). Consistent with the present results, all genes except H2BC14 were upregulated in LUAD tumour tissues compared with normal tissues when analysing the gene expression profiling data using the online tool GEPIA (Fig. 5A). Furthermore, the gene expression levels increased with increasing stages of LUAD (Fig. 5B), and low expression levels were associated with improved survival compared with high expression levels (Fig. 5C). Regarding the hypermethylation in the promoter of genes, the top genes showing the most notable difference in methylation included kinesin family member (KIF)20A (NCBI_10112), CENPA (NCBI_1058), KIF11 (NCBI_3832), PCNA clamp-associated factor (PAF, NCBI_9768) and GINS complex subunit 2 (GINS2; NCBI_51659) (data not shown), which exhibited the same trends as aforementioned in LUAD (Fig. 6). The findings showed that genes that were downregulated in H2O2-treated cells were more highly expressed in tumour samples with poor prognosis. The biological function of these genes requires further study.

Although the gene expression and methylation levels were in accordance, there were some differences between the H2O2-treated and the control group (Fig. 7A and B). Integrated analysis revealed that the total expression levels of the genes decreased as the methylation levels increased. Upstream of genes, the high methylation levels decreased gene expression levels compared with low methylation levels (Fig. 7C). Furthermore, the methylation and gene expression levels in both study groups were associated. Consistent with a previous report (9), most genes with low expression levels had high methylation levels (Fig. 7D).

Discussion

In the present study, genome-wide analyses of mRNA expression and DNA methylation profiles were employed to explore the inhibitory mechanism of H2O2 on the proliferation of A549 lung cancer cells. Several genes, including CDKN3, DTL, CENPF, KIF20A, CENPA, KIF11, PAF and GINS2, were downregulated and hypermethylated in H2O2-treated A549 cells compared with in untreated cells, suggesting that they may have important roles in the inhibitory process of H2O2 on the proliferation of A549 cells.

Increasing evidence has revealed that gene expression can be affected by epigenetic regulatory mechanisms, with DNA methylation at CpG islands occurring around the genomic promoter regions (17,18). Hypermethylation can repress transcription and downregulate gene expression by altering the chromatin framework, thereby leading to cancer initiation and progression (19). Consistent with a previous report (19), the present study identified several genes associated with the progression of lung cancer that were downregulated and hypermethylated in the gene bodies in H2O2-treated cells, including CDKN3, DTL and CENPF. The expression levels of these genes increased with the progression of LUAD, and low expression levels were associated with an improved survival, further proving their critical roles in the pathological processes of lung cancer. Although the expression levels of HIST1H2BM were not significantly different between LUAD and normal control samples, this gene was identified as a candidate epigenetic biomarker in LUAD through genome-wide analysis comparing the methylation patterns (20). Another study obtained similar results, revealing that increased expression levels of HIST1H2BM were associated with a poor survival in patients with LUAD and may act as a potential biomarker of drug synergy for the future clinical trials (21). Although in the previous study CDK3 has been reported as a tumour suppressor by maintaining the proper number of centrosomes (22), CDKN3 has also been demonstrated to be a potential poor prognostic marker in LUAD through the systematic analysis of datasets in Lung Cancer Explorer (23). Another report further proved that CDKN3 upregulation, which is mainly caused by the increase of mitotic activity, may be associated with poor survival in patients with LUAD, arguing against CDKN3 as a tumour suppressor (24).

In the present study, genes including KIF20A, CENPA, KIF11, PAF and GINS2, were downregulated in H2O2-treated cells and hypermethylated in the gene promoters. While these genes were also upregulated in LUAD tumour samples and their expression levels increased with the progression of LUAD, suggesting that they may have crucial roles in the development of tumours. Accumulating evidence has identified that KIF20A is a major gene associated with various types of cancer, such as pancreatic cancer (25), gastric cancer (26) and glioma (27). Ni et al (28) identified KIF20A as a potential prognostic biomarker that was associated with the pathogenesis of NSCLC through a series of bioinformatics methods. Gasnereau et al (29) further proved that the expression levels of KIF20A increased during the proliferation of hepatocytes and the occurrence of lung cancer.

Epigenetic mechanisms, such as DNA methylation, are essential for the regulation of gene expression, and aberrant epigenetic alterations can lead to pathological conditions, including cancer (30). DNA methylation close to the promoters of a gene has been reported to repress gene expression (31), whereas the effect of methylation in the gene body is unclear. Xie et al (31) demonstrated that ROS downregulated the expression levels of the AT-rich interaction domain 1A via methylation of its promoter during the pathogenesis of endometriosis, whereas Wang et al (32) revealed that methylation in the inositol-triphosphate 3-kinase A gene body regulated gene expression and may serve as an early diagnostic marker in lung cancer. The results of the present study revealed that in the upstream region of genes, high methylation levels were associated with decreased gene expression compared with a low methylation level, whereas in the gene body or downstream of genes, the methylation level did not markedly affect gene expression. Therefore, it was hypothesized that DNA methylation in the upstream region of genes, particularly near the promoter, may have different mechanisms than in the gene body region. Future research is required to increase the understanding of the mechanisms and critical roles of different methylation regions in various cellular processes.

The present study identified several genes that were downregulated and hypermethylated in A549 cells treated with H2O2, suggesting that they may have vital roles in H2O2-induced inhibition of the proliferation of A549 cells. However, further experiments are required to validate the specific function of these genes in A549 cells. In addition, the association analysis between gene expression and methylation levels indicated that their association may be far more complicated than previously thought, with the effects of DNA methylation on gene expression appearing to be position-dependent; whether the effects are also sequence-dependent requires further investigation. In conclusion, the present study integrated mRNA expression and DNA methylation profiling, providing novel insights into the molecular mechanisms underlying the pathological processes of lung cancer, and contributed to the identification of biomarkers and novel strategies for drug design for the treatment of lung cancer.

Acknowledgements

Not applicable.

Funding

The present work was supported by grants from the Affiliated Hospital of Youjiang Medical University for Nationalities Outstanding Scholar Funding (grant no. R20196313) and Doctorate Awarding Unit Funding of the Affiliated Hospital of Youjiang Medical University for Nationalities [grant no. (2019)48]. The funders had no role in the design of the study, the collection, analyses or interpretation of data, the writing of the manuscript or the decision to publish the results.

Availability of data and materials

The datasets generated and/or analysed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YL was involved in the design of the study, analysis and interpretation of data, and drafting the manuscript. BY gave final approval of the version of the manuscript to be published and was involved in data analysis. SH revised the manuscript critically for important intellectual content, and was involved in the acquisition and analysis of data. ZW made substantial contributions to conception and design. All authors have 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 

Collins Y, Chouchani ET, James AM, Menger KE, Cochemé HM and Murphy MP: Mitochondrial redox signalling at a glance. J Cell Sci. 125:801–806. 2012.PubMed/NCBI

2 

Forkink M, Basit F, Teixeira J, Swarts HG, Koopman WJH and Willems PHGM: Complex I and complex III inhibition specifically increase cytosolic hydrogen peroxide levels without inducing oxidative stress in HEK293 cells. Redox Biol. 6:607–616. 2015.PubMed/NCBI

3 

Zhang YW, Zheng Y, Wang JZ, Lu XX, Wang Z, Chen LB, Guan XX and Tong JD: Integrated analysis of DNA methylation and mRNA expression profiling reveals candidate genes associated with cisplatin resistance in non-small cell lung cancer. Epigenetics. 9:896–909. 2014.PubMed/NCBI

4 

Wan M, Bennett BD, Pittman GS, Campbell MR, Reynolds LM, Porter DK, Crowl CL, Wang X, Su D, Englert NA, et al: Identification of smoking-associated differentially methylated regions using reduced representation bisulfite sequencing and cell type-specific enhancer activation and gene expression. Environ Health Perspect. 126:0470152018.PubMed/NCBI

5 

Deng F, Yang ZF and Sun CQ: The role of Notch1 genes in lung cancer A594 cells and the impact on chemosensitivity. Eur Rev Med Pharmacol Sci. 21:2659–2664. 2017.PubMed/NCBI

6 

Park W: MAPK inhibitors, particularly the JNK inhibitor, increase cell death effects in H2O2-treated lung cancer cells via increased superoxide anion and glutathione depletion. Oncol Rep. 39:860–870. 2018.PubMed/NCBI

7 

Sun C, Jin W and Shi H: Oligomeric proanthocyanidins protects A549 cells against H2O2-induced oxidative stress via the Nrf2-ARE pathway. Int J Mol Med. 39:1548–1554. 2017.PubMed/NCBI

8 

Bird A: DNA methylation patterns and epigenetic memory. Genes Dev. 16:6–21. 2002.PubMed/NCBI

9 

Rauscher GH, Kresovich JK, Poulin M, Yan L, Macias V, Mahmoud AM, Al-Alem U, Kajdacsy-Balla A, Wiley EL, Tonetti D and Ehrlich M: Exploring DNA methylation changes in promoter, intragenic, and intergenic regions as early and late events in breast cancer formation. BMC Cancer. 15:8162015.PubMed/NCBI

10 

Chen X, Liu L, Mims J, Punska EC, Williams KE, Zhao W, Arcaro KF, Tsang AW, Zhou X and Furdui CM: Analysis of DNA methylation and gene expression in radiation-resistant head and neck tumors. Epigenetics. 10:545–561. 2015.PubMed/NCBI

11 

Cheah SY, Lawford BR, Young RM, Morris CP and Voisey J: mRNA Expression and DNA methylation analysis of serotonin receptor 2A (HTR2A) in the human schizophrenic brain. Genes (Basel). 8:142017.

12 

Diederich M, Hansmann T, Heinzmann J, Barg-Kues B, Herrmann D, Aldag P, Baulain U, Reinhard R, Kues W, Weissgerber C, et al: DNA methylation and mRNA expression profiles in bovine oocytes derived from prepubertal and adult donors. Reproduction. 144:319–330. 2012.PubMed/NCBI

13 

Anglim PP, Galler JS, Koss MN, Hagen JA, Turla S, Campan M, Weisenberger DJ, Laird PW, Siegmund KD and Laird-Offringa IA: Identification of a panel of sensitive and specific DNA methylation markers for squamous cell lung cancer. Mol Cancer. 7:622008.PubMed/NCBI

14 

Del Real A, Pérez-Campo FM, Fernández AF, Sañudo C, Ibarbia CG, Pérez-Núñez MI, Criekinge WV, Braspenning M, Alonso MA, et al: Differential analysis of genome-wide methylation and gene expression in mesenchymal stem cells of patients with fractures and osteoarthritis. Epigenetics. 12:113–122. 2017.PubMed/NCBI

15 

Kernaleguen M, Daviaud C, Shen Y, Bonnet E, Renault V, Deleuze JF, Mauger F and Tost J: Whole-genome bisulfite sequencing for the analysis of genome-wide dnamethylation and hydroxymethylation patterns at single-nucleotide resolution. Methods Mol Biol. 1767:311–349. 2018.PubMed/NCBI

16 

Riesewijk A, Martín J, van Os R, Horcajadas JA, Polman J, Pellicer A, Mosselman S and Simón C: Gene expression profiling of human endometrial receptivity on days LH+2 versus LH+7 by microarray technology. Mol Hum Reprod. 9:253–264. 2003.PubMed/NCBI

17 

Guo K, Elzinga S, Eid S, Figueroa-Romero C, Hinder LM, Pacut C, Feldman EL and Hur J: Genome-wide DNA methylation profiling of human diabetic peripheral neuropathy in subjects with type 2 diabetes mellitus. Epigenetics. 14:766–779. 2019.PubMed/NCBI

18 

Navarro A, Yin P, Monsivais D, Lin SM, Du P, Wei JJ and Bulun SE: Genome-Wide DNA methylation indicates silencing of tumor suppressor genes in uterine leiomyoma. PLoS One. 7:e332842012.PubMed/NCBI

19 

Li P, Shi J, He Q, Hu Q, Wang YY, Zhang LJ, Chan WT and Chen WX: Streptococcus pneumoniae induces autophagy through the Inhibition of the PI3K-I/Akt/mTOR Pathway and ROS Hypergeneration in A549 Cells. PLoS One. 10:e01227532015.PubMed/NCBI

20 

Daugaard I, Dominguez D, Kjeldsen TE, Kristensen LS, Hager H, Wojdacz TK and Hansen LL: Identification and validation of candidate epigenetic biomarkers in lung adenocarcinoma. Sci Rep. 6:358072016.PubMed/NCBI

21 

Ponsuksili S, Trakooljul N, Basavaraj S, Hadlich F, Murani E and Wimmers K: Epigenome-wide skeletal muscle DNA methylation profiles at the background of distinct metabolic types and ryanodine receptor variation in pigs. BMC Genomics. 20:4922019.PubMed/NCBI

22 

Srinivas V, Kitagawa M, Wong J, Liao PJ and Lee SH: The Tumor suppressor Cdkn3 is required for maintaining the proper number of centrosomes by regulating the centrosomal stability of Mps1. Cell Rep. 13:1569–1577. 2015.PubMed/NCBI

23 

Zang X, Chen M, Zhou Y, Xiao G, Xie Y and Wang X: Identifying CDKN3 gene expression as a prognostic biomarker in lung adenocarcinoma via meta-analysis. Cancer Inform. 14 (Suppl 2):S183–S191. 2015.

24 

Fan C, Chen L, Huang Q, Shen T, Welsh EA, Teer JK, Cai J, Cress WD and Wu J: Overexpression of major CDKN3 transcripts is associated with poor survival in lung adenocarcinoma. Br J Cancer. 113:1735–1743. 2015.PubMed/NCBI

25 

Imai K, Hirata S, Irie A, Senju S, Ikuta Y, Yokomine K, Harao M, Inoue M, Tomita Y, Tsunoda T, et al: Identification of HLA-A2-restricted CTL epitopes of a novel Tumour-associated antigen, KIF20A, overexpressed in pancreatic cancer. Br J Cancer. 104:300–307. 2011.PubMed/NCBI

26 

Yan GR, Zou FY, Dang BL, Zhang Y, Yu G, Liu X and He QY: Genistein-induced mitotic arrest of gastric cancer cells by downregulating KIF20A, a proteomics study. Proteomics. 12:2391–2399. 2012.PubMed/NCBI

27 

Saito K, Ohta S, Kawakami Y, Yoshida K and Toda M: Functional analysis of KIF20A, a potential immunotherapeutic target for glioma. J Neurooncol. 132:63–74. 2017.PubMed/NCBI

28 

Ni M, Liu X, Wu J, Zhang D, Tian J, Wang T, Liu S, Meng Z, Wang K, Duan X, et al: Identification of candidate biomarkers correlated with the pathogenesis and prognosis of non-small cell lung cancer via integrated bioinformatics analysis. Front Genet. 9:4692018.PubMed/NCBI

29 

Gasnereau I, Boissan M, Margall-Ducos G, Couchy G, Wendum D, Bourgain-Guglielmetti F, Desdouets C, Lacombe ML, Zucman-Rossi J and Sobczak-Thépot J: KIF20A mRNA and its product MKlp2 Are increased during hepatocyte proliferation and hepatocarcinogenesis. Am J Pathol. 180:131–140. 2012.PubMed/NCBI

30 

Yang H, Chang F, You C, Cui J, Zhu G, Wang L, Zheng Y, Qi J and Ma H: Whole-genome DNA methylation patterns and complex associations with gene structure and expression during flower development in Arabidopsis. Plant J. 81:268–281. 2014.PubMed/NCBI

31 

Xie H, Chen P, Huang HW, Liu LP and Zhao F: Reactive oxygen species downregulate ARID1A expression via its promoter methylation during the pathogenesis of endometriosis. Eur Rev Med Pharmacol Sci. 21:4509–4515. 2017.PubMed/NCBI

32 

Wang YW, Ma X, Zhang YA, Wang MJ, Yatabe Y, Lam S, Girard L, Chen JY and Gazdar AF: ITPKA gene body methylation regulates gene expression and serves as an early diagnostic marker in lung and other cancers. J Thorac Oncol. 11:1469–1481. 2016.PubMed/NCBI

Related Articles

Journal Cover

December-2020
Volume 20 Issue 6

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
Li Y, Wei Z, Huang S and Yang B: mRNA expression and DNA methylation analysis of the inhibitory mechanism of H<sub>2</sub>O<sub>2</sub> on the proliferation of A549&nbsp;cells. Oncol Lett 20: 288, 2020
APA
Li, Y., Wei, Z., Huang, S., & Yang, B. (2020). mRNA expression and DNA methylation analysis of the inhibitory mechanism of H<sub>2</sub>O<sub>2</sub> on the proliferation of A549&nbsp;cells. Oncology Letters, 20, 288. https://doi.org/10.3892/ol.2020.12151
MLA
Li, Y., Wei, Z., Huang, S., Yang, B."mRNA expression and DNA methylation analysis of the inhibitory mechanism of H<sub>2</sub>O<sub>2</sub> on the proliferation of A549&nbsp;cells". Oncology Letters 20.6 (2020): 288.
Chicago
Li, Y., Wei, Z., Huang, S., Yang, B."mRNA expression and DNA methylation analysis of the inhibitory mechanism of H<sub>2</sub>O<sub>2</sub> on the proliferation of A549&nbsp;cells". Oncology Letters 20, no. 6 (2020): 288. https://doi.org/10.3892/ol.2020.12151