Genome‑wide analysis of histone modifications by ChIP‑chip to identify silenced genes in gastric cancer

  • Authors:
    • Xinjiang Zhu
    • Jian Liu
    • Xiaoyang Xu
    • Chundong Zhang
    • Dongqiu Dai
  • View Affiliations

  • Published online on: February 27, 2015     https://doi.org/10.3892/or.2015.3824
  • Pages: 2567-2574
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to identify novel histone modification markers in gastric cancer (GC) by chromatin immunoprecipitation microarray (ChIP‑chip) analysis and to determine whether these markers were able to discriminate between normal and GC cells. We also tested for correlations with DNA methylation. We probed a human CpG island microarray with DNA from a GC cell line (MKN45) by chromatin immunoprecipitation (ChIP). ChIP‑reverse‑transcriptase quantitative polymerase chain reaction PCR (RT‑qPCR) was used to validate the microarray results. Additionally, mRNA expression levels and the DNA methylation of potential target genes were evaluated by RT‑qPCR and methylation‑specific PCR (MSP). The moults showed that 134 genes exhibited the highest signal‑to‑noise ratio of H3‑K9 trimethylation over acetylation and 46 genes exhibited the highest signal‑to‑noise ratio of H3‑K9 trimethylation over H3‑K4 trimethylation in MKN45 cells. The ChIP‑qPCR results agreed with those obtained from the ChIP‑chip analysis. Aberrant DNA methylation status and mRNA expression levels were also identified for selected genes (PSD, SMARCC1 and Vps37A) in the GC cell lines. The results suggest that CpG island microarray coupled with ChIP (ChIP‑chip) can identify novel targets of gene silencing in GC. Additionally, ChIP‑chip is the best approach for assessing the genome‑wide status of epigenetic regulation, which may allow for a broader genomic understanding compared to the knowledge that has been accumulated from single‑gene studies.

Introduction

Gastric cancer (GC) is one of the most common malignancies and is the second and third leading cause of cancer-related mortality in men and women, respectively, worldwide. Compared to other malignant types of cancer, the incidence and mortality of GC ranks first in China; where the cases account for 42% of all GC cases worldwide (1). Most patients are diagnosed at an advanced stage, thus GC continues to be a highly aggressive malignancy that is associated with poor prognosis and a low survival rate. Therefore, additional studies are needed to explore the molecular pathophysiology of GC.

Mounting evidence has indicated that epigenetic alterations play a key role in the occurrence and development of GC (2,3). These epigenetic alterations, such as DNA and histone methylation, provide an alternative pathway for gene silencing that is distinct from gene mutation and deletion. Tumor-suppressor gene (TSG) silencing associated with DNA methylation in cancer is accompanied by abnormal histone modifications (4,5). Histone modifications, which have been recently recognized to generate a ‘histone code’ that affects chromatin structure and gene expression, also play an important role in the establishment of gene silencing during tumorigenesis (6). Histone lysine methylation and acetylation, which are two of the best-studied histone modifications, are involved in many biological processes, including gene activation and silencing, DNA methylation, X-chromosome inactivation, DNA repair and cell cycle control (7). H3-K9 acetylation is known to be associated with active transcription, whereas H3-K9 trimethylation has been connected with repressed transcription (8). Furthermore, H3-K4 trimethylation has been detected at active genes and is thought to promote gene expression. Histone acetylation and methylation are likely to act cooperatively to regulate gene transcription.

In the post-genomic era, high-throughput technologies have made it possible to analyze the chromatin structure at the genomic scale in cancer cells. Chromatin immunoprecipitation coupled with microarray (ChIP-chip) has been used by investigators to conduct genome-wide analyses (9,10). In the recent study, we used the ChIP-chip technique to characterize inactive genes in GC, with an aim to identify the targets of genes controlled by histone modifications. We also examined the relevance of histone modifications and DNA methylation for these genes. The results showed that the histone modifications and DNA methylation study acted cooperatively to affect gene expression in GC.

Materials and methods

Cell culture and treatment with epigenetic agents

The human MKN45, AGS, BGC823 and SGC7901 GC cell lines were obtained from the Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai, China). The immortalized GES1 normal gastric cell line was obtained from the oncology Institute of China Medical University. The cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (Gibco-BRL, Grand Island, NY, USA) and incubated at 37°C in a humidified 5% Co2 atmosphere. The four GC lines were incubated in culture media with 5 μM of the DNA methyltransferase (DNMT) inhibitor 5-aza-2′-deoxycytidine (DAC) for 3 days, and 0.3 μM of the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) (both from Sigma-Aldrich, St. Louis, MO, USA) for 1 day. The time, dose and sequence of DAC and/or TSA were based on previous studies (11,12).

Chromatin immunoprecipitation (ChIP) assay

Five million cells were crosslinked with 1% formaldehyde for 10 min at 37°C, and then 0.125 M of glycine was added to stop the crosslinking. After washing with ice-cold PBS, the cell pellets were resuspended in lysis buffer, and sonicated to generate 200 to 1,000-bp DNA fragments. The lysate was then divided into three fractions. The first lysate was precipitated using antibodies against Lys-9 trimethylated histone H3 (05-1242), Lys-9 acetylated histone H3 (07-352) and Lys-4 trimethylated histone H3 (07-472) (all from Millipore, Billerica, MA, USA) at 4°C overnight. The second lysate was incubated with normal rabbit IgG (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) as a negative control. The third lysate was used as an input control. Protein G-Sepharose beads were added to collect the immunoprecipitated complexes and left them to incubate for 1 h at 4°C. After washing, the beads were treated with RNase (50 mg/ml) for 30 min at 37°C and then proteinase K overnight. The crosslinks were then reversed by heating the sample at 65°C for 6 h. DNA was extracted by the phenol/chloroform method, ethanol precipitated, and resuspended in 20 μl water.

ChIP-chip

Immunoaffinity-enriched DNA fragments (IP) and input samples were amplified using a Whole Genome Amplification kit (Sigma-Aldrich). Input and IP samples were labeled in separate reactions with Cy3 and Cy5, respectively and were co-hybridized to a Human 3 × 720K RefSeq Promoter Array (NimbleGen, Madison, WI, USA), containing 22,542 human promoter regions that cover a range from −3,200 to +800 bp, relative to the transcription start site. The hybridized microarray slides were scanned using a GenePix 4000B scanner (Axon Instruments, Foster City, CA, USA). Data were extracted using NimbleScan software. From the normalized log2-ratio data, a permutation-based peak finding algorithm provided by NimbleScan version 2.3 (NimbleGen) was used to detect peaks that represented significant positive enrichment. To detect the peaks for histone modification, only the false discovery rate (FDR <0.05) was considered for analysis.

ChIP-qPCR

ChIP was conducted in the same manner as for ChIP-chip. Briefly, immunoaffinity-enriched, input, and negative control DNA were used for PCR. Primers were designed based on the promoter structure of the genes selected for evaluating ChIP-chip data (Table I). PCR products were visualized on a 2.5% agarose gel. For quantification, PCR amplification was performed on an ABI 7700 real-time PCR (Applied Biosystems, Foster City, CA, USA). PCR conditions included an initial denaturation step of 4 min at 95°C, followed by 35 cycles of 5 sec at 95°C, 30 sec at 59°(60°)C and 20 sec at 72°C. Quantitative ChIP-PCR values were normalized against values from a standard curve constructed using input DNA that was extracted for the ChIP experiment. ChIP experiments were repeated three times for each target gene.

Table I

ChIP-qPCR primers used for validation of the ChIP-chip data.

Table I

ChIP-qPCR primers used for validation of the ChIP-chip data.

GenePrimer sequence (5→3)Annealing temperature (°C)Size (bp)
PSDF: GACTGGCTTCTGTCGTCCTC
R: GGCAGACAGTAAGAGCCTGG
60190
NLKF: TGTCTATTTGGCCCAGGTTC
R: AAAGGCAGAGTTTGGCTTGA
59201
SMARCC1F: TTCAAGCAATTCTCCTGCCT
R: CGCCTGTAATCCCAACACTT
59183
Vps37AF: CAGTGCGAAGGTGCTGATAA
R: ACGATGAACCTGAGAGGGTG
60280

[i] F, forward primer; R, reverse primer; ChIP-chip, chromatin immunoprecipitation microarray; qPCR, quantitative polymerase chain reaction.

RNA extraction and reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from cells and tissues with TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. The quality and concentration of RNA were measured by ultraviolet absorbance at 260 and 280 nm (A260/A280 ratio) and checked by agarose gel electrophoresis individually. Total RNA was reverse transcribed into cDNA using an Expand Reverse Transcriptase kit (Takara, Dalian, China). Expression of PSD mRNA was detected using qPCR under the following conditions: 95°C for 30 sec, 35 cycles of 95°C for 5 sec and 60°C for 30 sec. The reaction mixture contained 12.5 μl SYBR-Green (Takara), 1 μl of each primer, 2 μl cDNA, and 8.5 μl diethylpyrocarbonate (DEPC)-treated water. The PCR primers used for each gene in this analysis are shown in Table II. Negative control used DEPC-treated water to replace cDNA templates for each PCR. The PSD level was expressed as Ct after normalization to the levels of GAPDH mRNA. The experiment was performed in triplicate.

Table II

Primers used for RT-qPCR analysis.

Table II

Primers used for RT-qPCR analysis.

GenePrimer sequence (5→3)Annealing temperature (°C)Size (bp)
PSDF: CTGGGCAAGAACAATGACTTC
R: GAGGACAGGGCTTCAGGATT
58140
SMARCC1F: TGAACGGGAAGCTCACTGG
R: TCCACCACCCTGTTGCTGTA
59249
Vps37AF: CGCCAGCATCCACCACG
R: TTGAGTTTGTGAATGAC
60283
GAPDHF: CATGAGAAGTATGACAACAGCCT
R: AGTCCTTCCACGATACCAAAGT
60257

[i] F, forward primer; R, reverse primer; RT-PCR, reverse-transcriptase quantitative polymerase chain reaction.

Methylation-specific PCR (MSP)

Genomic DNA was extracted from cells with phenolchloroform-isoamyl alcohol and collected by ethanol precipitation. Genomic DNA (2 μg) was treated with NaOH (2 M) at 42°C for 20 min. After denaturation, the DNA was incubated with hydroquinone and sodium bisulfate at 54°C for 16 h in the dark. DNA was purified using a DNA Cleanup kit (Promega, Madison, WI, USA), followed by incubation with 3 M NaOH at 37°C for 15 min, and precipitation with ammonium acetate and 100% ethanol at −20°C overnight. The following day, DNA was washed with 70% ethanol, and dissolved in 15 μl TE buffer. The primers used for MSP were located in the promoter region of the genes (Table III). Peripheral blood cell DNA from healthy adults treated with SssI methyltransferase (New England Biolabs, Ipswich, MA, USA) and untreated DNA were used as positive and negative controls, respectively. PCR products were separated by electrophoresis on 2% agarose gels, and quantified using the Fluor Chen 2.0 system.

Table III

Primers used for MSP analysis.

Table III

Primers used for MSP analysis.

GenePrimer sequence (5→3)Annealing temperature (°C)Size (bp)
PSD-MF: GTTGTAGGGAAGCGGTTC
R: CGACCACGAAAAAAAACC
55150
PSD-UF: AGGGTTGTAGGGAAGTGGTTT
R: CAACCACAAAAAAAAACCTA
55151
SMARCC1-MF: GGATTACGAGGTTAGGAGATC
R: CGACTCACTACAAACTCCG
56180
SMARCC1-UF: GGATTATGAGGTTAGGAGATT
R: CAACTCACTACAAACTCCA
56181
Vps37A-MF: TAGAGATAGTATTCGGCGGC
R: TCAAAACGTACGAAAAACGA
59130
Vps37A-UF: TTATAGAGATAGTATTTGGTG
R: TCAAAACATACAAAAAACAAC
59131

[i] F, forward primer; R, reverse primer; MSP, methylation-specific PCR.

Statistical analysis

Statistical analyses were performed using SPSS version 17.0 (SPSS, Chicago, IL, USA). Quantitative data are shown as mean values ± SD. Statistical analyses were performed using the independent sample t-test. P<0.05 was considered statistically significant.

Results

Genome-wide analysis of histone modifications by ChIP-chip

TSG inactivation resulting from higher H3-K9 trimethylation, lower H3-K9 acetylation and H3-K4 trimethylation has been reported to be involved in carcinogenesis. We aimed to identify genes that underwent similar histone modifications in GC cells. We used a two-step strategy to analyze the data. First, we selected 134 candidate genes that showed a high ratio of H3-K9 trimethylation over acetylation (H3-K9Me3/H3-K9Ac >2.0). The selected 20 genes with a high ratio of H3-K9 trimethylation over H3-K9 acetylation are presented in Table IV. We then analyzed 46 candidate genes that showed a high ratio of H3-K9 trimethylation over H3-K4 trimethylation (H3-K9Me3/H3-K4Me3 >2.0). The selected 20 genes with a high ratio of H3-K9 trimethylation over H3-K4 trimethylation are presented in Table V.

Table IV

Twenty genes showing a high ratio of H3-K9 trimethylation over acetylation (H3-K9 Me3/H3-K9Ac >2.0), identified by ChIP-CpG microarray.

Table IV

Twenty genes showing a high ratio of H3-K9 trimethylation over acetylation (H3-K9 Me3/H3-K9Ac >2.0), identified by ChIP-CpG microarray.

Accession no.LocationGene symbolDescriptionFold-change
NM_170743Chr 1: 24386073–24386599IL28RAInterleukin 28 receptor, α (interferon, λ receptor)4.14
NM_001145442Chr 14: 19090441–19091120POTEMPOTE ankyrin domain family, member M3.58
NM_021080Chr 1: 58488062–58488534DAB1Disabled homolog 1 (Drosophila)3.19
NM_005186Chr 11: 64705434–64705961CAPN1Calpain 1, (mu/I) large subunit3.08
NM_014588Chr 2: 25010707–25011384VSX1Visual system homeobox 12.99
NM_001145152aChr 8: 17149196–17149585Vps37AVacuolar protein sorting 37 homolog A2.98
NM_001079526Chr 2: 213724263–213724861IKZF2IKAROS family zinc finger 22.97
NM_153456Chr 13: 95541348–95541914HS6ST3Heparan sulfate 6-O-sulfotransferase 32.96
NM_201269Chr 1: 91259477–91260224ZNF644Zinc finger protein 6442.95
NM_032991Chr 4: 185807070–185807632CASP3Caspase-3, apoptosis-related cysteine peptidase2.94
NM_138785Chr 6: 149928999–149929638C6orf72Chromosome 6 open reading frame 722.94
NM_020680Chr 11:65049471–65049926SCYL1SCY1-like 1 (S. cerevisiae)2.92
NM_024907Chr 19: 44158164–44158927FBXO17F-box protein 172.92
NM_014718Chr 12: 7173825–7174446CLSTN3Calsyntenin 32.92
NM_017924Chr 14: 22633202–22634021 C14orf119Chromosome 14 open reading frame 1192.92
NM_022898Chr 14: 98807632–98808446BCL11BB-cell CLL/lymphoma 11B (zinc finger protein)2.90
NM_014229Chr 22: 10832813–10833404SLC6A11Solute carrier family 6 member 112.90
NM_021255Chr 14: 55654533–55655026PELI2Pellino homolog 2 (Drosophila)2.89
NM_016231aChr 17: 23392754–23393241NLKNemo-like kinase2.48
NM_003074aChr 3: 47798031–47798567SMARCC1SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily c, member 12.44

a Selected genes used for validation of the ChIP-chip data. ChIP-chip, chromatin immunoprecipitation microarray; Chr, chromosome.

Table V

Twenty selected genes showing a high ratio of H3-K9 trimethylation over H3-K4 trimethylation (H3-K9 Me3/H3-K4Me3 >2.0), identified by ChIP-CpG microarray.

Table V

Twenty selected genes showing a high ratio of H3-K9 trimethylation over H3-K4 trimethylation (H3-K9 Me3/H3-K4Me3 >2.0), identified by ChIP-CpG microarray.

Accession no.LocationGene symbolDescriptionFold-change
NM_002226Chr14: 104705881–104706590JAG2Jagged 23.02
NM_001127899ChrX: 49573893–49574453CLCN5Chloride channel 52.96
NM_032560Chr14: 91045898–91046864SMEK1SMEK homolog 1, suppressor of mek1 (Dictyostelium)2.95
NM_015500Chr21: 42246382–42246894C2CD2C2 calcium-dependent domain containing 22.92
NM_003224Chr20: 61809198–61810017ARFRP1ADP-ribosylation factor-related protein 12.92
NM_001101314Chr7: 150128124–150128968 TMEM176BTransmembrane protein 176B2.91
NM_080622Chr20: 61963760–61964422 C20orf135Chromosome 20 open reading frame 1352.90
NM_014618Chr9: 121171512–121171970DBC1Deleted in bladder cancer 12.90
NM_001006Chr4: 152239592–152240103RPS3ARibosomal protein S3A2.86
NM_002779a Chr10:104167927–104168755PSDPleckstrin and Sec7 domain containing2.60
NM_001012614Chr4: 1232543–1233011CTBP1C-terminal binding protein 12.57
NM_001409Chr1: 3517795–3518285MEGF6Multiple EGF-like-domains 62.46
NM_016231aChr17: 23392754–23393241NLKNemo-like kinase2.45
NM_138328Chr17: 27616688–27617453RHBDL3Rhomboid, veinlet-like 3 (Drosophila)2.39
NM_005923Chr6: 137155422–137155958MAP3K5Mitogen-activated protein kinase 52.35
NM_003074aChr3: 47798031–47798567SMARCC1SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily c, member 12.30
NM_014806Chr9: 35479779–35480211RUSC2RuN and SH3 domain containing 22.26
NM_015549Chr14: 64241089–64241538PLEKHG3Pleckstrin homology domain containing, family G (with RhoGef domain) member 32.22
NM_001083591Chr10: 11247329–11247847CELF2CuGBP, Elav-like family member 22.17
NM_001031807Chr15: 65903316–65904082SKOR1SKI family transcriptional corepressor 12.10

a Selected genes used for validation of the ChIP-chip data; ChIP-chip, chromatin immunoprecipitation microarray.

Validation for ChIP-chip data

To validate the microarray results, the selected genes, PSD, NLK, SMARCC1 and Vps37A exhibited a high ratio of H3-K9 trimethylation over acetylation or a high ratio of H3-K9 methylation over H3-K4 trimethylation in the MKN45 cell line. These results were verified by ChIP-qPCR. As shown in Table VI and Fig. 1, qPCR results for the selected candidate genes were consistent with the ChIP-chip data.

Table VI

Results of ChIP-qPCR and ChIP-chip.

Table VI

Results of ChIP-qPCR and ChIP-chip.

VariableNLKPSDVps37ASMARCC1
ChIP-qPCR
 H3-K9Me30.9204±0.00690.8760±0.00490.8789±0.0090.8811±0.0018
 H3-K9Ac0.3924±0.00240.3910±0.00150.3540±0.00580.3176±0.0015
 H3-K4Me30.4185±0.00150.3909±0.00170.7920±0.00360.3708±0.0091
 K9Me3/K9Ac2.342.242.482.77
 K9Me3/K4Me32.202.241.112.38
ChIP-chip
 K9Me3/K9Ac2.482.342.982.44
 K9Me3/K4Me32.452.600.972.30

[i] ChIP-chip, chromatin immunoprecipitation microarray; qPCR, quantitative polymerase chain reaction.

Differential mRNA expression in GC cells

Using qRT-PCR to assess the mRNA expression levels of SMARCC1 and Vps37A, we found that SMARCC1 was downregulated in MKN45 (0.5377±0.01126), AGS (0.5697±0.00963), BGC823 (0.3157±0.0099) and SGC7901 (0.527±0.0046) cells compared to the normal mucosa cell line, GES-1 (1-fold, as was the control). Of note, the mRNA expression levels of Vps37A were similar between GC and GES-1 cells (Fig. 2).

Effects of 5-aza-2′-DAC and TSA on gene re-expression

To determine whether epigenetic agents altered PSD, SMARCC1 and Vps37A gene expression levels, we treated MKN45 cells with the DNMT inhibitor DAC, the HDAC inhibitor TSA, or a combined treatment of the two agents. Using RT-qPCR, we found that DAC and TSA had different effects on the expression of these genes in MKN45 cells. DAC alone restored PSD expression, whereas TSA had no effect on PSD expression. The combined treatment restored PSD expression levels similar to those observed after DAC treatment. DAC or TSA alone restored SMARCC1 expression and the combined treatment with the two agents restored SMARCC1 expression to a significantly greater degree than the treatment with either agent alone. Treatment with DAC and TSA, alone or in combination, had no significant effect on Vps37A expression (Fig. 3).

Expression of the PSD, SMARCC1 and Vps37A genes is associated with DNA methylation

To determine whether DNA methylation affected the expression levels of these genes, we assessed their DNA methylation status in MKN45 cell lines using MSP. We observed hypermethylation of the PSD promoter, partial methylation of the SMARCC1 promoter, and no methylation of the Vps37A promoter, suggesting that the methylation status of these gene promoters was partially responsible for the mRNA expression levels. In MKN45 cells, treatment with DAC resulted in demethylation of the PSD and SMARCC1 promoters, and treatment with TSA resulted in demethylation of the SMARCC1 promoter. However, TSA had no effect on the PSD promoter. Treatment with DAC, TSA or both had no effect on the Vps37A promoter (Fig. 4).

Discussion

Epigenetic dysregulation of gene silencing plays a causal role in cancer development and progression. One of the most common epigenetic alterations in cancer results from histone modifications. Modifications of histone tails are thought to specify a code that regulates gene expression (13). The emerging consensus is that high levels of H3-K9 trimethylation, and low levels of H3-K9 acetylation and H3-K4 trimethylation are associated with inactive genes. Previous studies of histone modifications were limited to single-gene disruption (?). However, accumulating evidence shows that multiple genes may simultaneously be involved in gastric tumorigenesis. Advances in high-throughput techniques have enabled the development of ChIP analysis combined with DNA microarray (ChIP-chip), making it possible to profile and quantify the expression of thousands of genes simultaneously. This new high-throughput method has been widely used in epigenetics (1417). Thus, in the present study, we used a ChIP-chip strategy to explore gene silencing events associated with higher levels of H3-K9 trimethylation, and lower levels of H3-K9 acetylation and H3-K4 trimethylation. These genes included TSG, oncogenes, cell adhesion molecules and cycle regulators, and apoptosis-regulating genes. We also used ChIP-qPCR to confirm the microarray data. We selected four genes with a high ratio of H3-K9 trimethylation over acetylation, or a high ratio of H3-K9 methylation over H3-K4 trimethylation. The NLK, PSD, SMARCC1 and Vps37A genes were used to validate the microarray results in the MKN45 cell line, and the result consistency ultimately proved the value of this approach.

Among the candidates identified by ChIP-chip, PSD is a guanine nucleotide exchange factor for ADP-ribosylation factor 6 (ARF6) (18), which regulates the membrane trafficking of small G proteins (19). The PSD allele is located on human chromosome 10q24 and encodes a 71-kDa protein (20). Okada et al (21) reported that PSD was more frequently methylated in ulcerative colitis (UC)-associated colorectal cancer tissues than in non-neoplastic UC epithelia. Additionally, PSD mRNA expression levels were positively correlated with the methylation status of PSD. SMARCC1 is a member of the SWI/SNF complex that has been shown to have tumor-suppressive abilities related to cell cycle control (22,23). However, the expression and function of SMARCC1 in tumorigenesis remains unclear. Loss of SMARCC1 expression contributed to tumor development as a consequence of its location on chromosome band 3p21.31, which includes other suspected TSGs, such as SEM3B and FUS1 (24,25). DelBove et al (26) indicated that lower levels of SMARCC1 protein expression in an ovarian carcinoma cell line, SKoV3, closely correlated with mutations in exon 24. The human homologue of Vps37A (hVps37A) is located on the short arm of chromosome 8. In this region, 8p22, loss of heterozygosity (LOH) occurs at a high frequency in several human cancers (27). Wittinger et al found that hVps37A was significantly downregulated in ovarian cancer and that hVps37A-deficient cells become non-responsive to inhibition by the therapeutic antibody cetuximab (28).

However, the roles of the selected genes in GC have not been investigated. In the present study, as anticipated, there was a low level of SMARCC1 expression in GC cell lines compared to GES-1 cells. Of note, mRNA expression levels of Vps37A in GC cell lines were similar to GES-1 cells. This exception is probably a consequence of other epigenetic modifications associated with transcriptional activation in this gene. DNA methylation is the most widely studied epigenetic event. Recent investigations have indicated that aberrant DNA methylation is associated with GC (29,30). In order to gain a better understanding of these potential exceptions, we characterized DNA methylation. Our data confirmed hypermethylation of the PSD and SMARCC1 promoters, and hypomethylation of the Vps37A promoter. Additionally, epigenetic agents resulted in the demethylation of the PSD and SMARCC1 promoters and reversed PSD and SMARCC1 expression levels. However, there was no obvious change in Vps37A expression levels following treatment with epigenetic modifying agents.

In summary, data from our current study show that microarray profiling coupled with ChIP using anti-H3-K9 trimethylation and acetylation, as well as H3-K4 trimethylation antibodies is a useful approach for identifying target genes silenced by the epigenetic machinery. We also identified epigenetic mechanisms underlying the decreased expression of PSD and SMARCC1: DNA hypermethylation, hypertrimethylation of H3-K9 and hypotrimethylation of H3-K4 in the promoter domain. By contrast, Vps37A expression was mainly affected by DNA methylation, but not by H3-K9 trimethylation, H3-K9 acetylation or H3-K4 trimethylation. These novel candidate genes are potential biomarkers or future therapeutic targets. Further investigations are needed to clarify the roles of these candidate genes in the development of GC.

Acknowledgments

The present study was supported partly by a grant from the National Natural Science Foundation of China (grant no. 30572162), the Foundation of Liaoning Province Science and Technology Plan Project (grant no. 2013225021) and the Higher Specialized Research Fund for Doctoral Program of the Ministry of Education of China (grant no. 20102104110001).

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Park YS, Jin MY, Kim YJ, Yook JH, Kim BS and Jang SJ: The global histone modification pattern correlates with cancer recurrence and overall survival in gastric adenocarcinoma. Ann Surg Oncol. 15:1968–1976. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Meng CF, Zhu XJ, Peng G and Dai DQ: Role of histone modifications and DNA methylation in the regulation of O6-methylguanine-DNA methyltransferase gene expression in human stomach cancer cells. Cancer Invest. 28:331–339. 2010. View Article : Google Scholar

4 

Kim M, Jang HR, Kim JH, et al: Epigenetic inactivation of protein kinase D1 in gastric cancer and its role in gastric cancer cell migration and invasion. Carcinogenesis. 29:629–637. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Jung Y, Park J, Bang YJ and Kim TY: Gene silencing of TSPYL5 mediated by aberrant promoter methylation in gastric cancers. Lab Invest. 88:153–160. 2008. View Article : Google Scholar

6 

Jones PA and Baylin SB: The fundamental role of epigenetic events in cancer. Nat Rev Genet. 3:415–428. 2002.PubMed/NCBI

7 

Ruthenburg AJ, Allis CD and Wysocka J: Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol Cell. 25:15–30. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Fuks F: DNA methylation and histone modifications: teaming up to silence genes. Curr Opin Genet Dev. 15:490–495. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Kondo Y, Shen L, Cheng AS, et al: Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet. 40:741–750. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Zha L, Wang Z, Tang W, Zhang N, Liao G and Huang Z: Genome-wide analysis of HMGA2 transcription factor binding sites by ChIP on chip in gastric carcinoma cells. Mol Cell Biochem. 364:243–251. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Fahrner JA, Eguchi S, Herman JG and Baylin SB: Dependence of histone modifications and gene expression on DNA hypermethylation in cancer. Cancer Res. 62:7213–7218. 2002.PubMed/NCBI

12 

Cameron EE, Bachman KE, Myöhänen S, Herman JG and Baylin SB: Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet. 21:103–107. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Jenuwein T and Allis CD: Translating the histone code. Science. 293:1074–1080. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Zhang L, Zhong K, Dai Y and Zhou H: Genome-wide analysis of histone H3 lysine 27 trimethylation by ChIP-chip in gastric cancer patients. J Gastroenterol. 44:305–312. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Wu J, Wang SH, Potter D, Liu JC, Smith LT, Wu YZ, Huang TH and Plass C: Diverse histone modifications on histone 3 lysine 9 and their relation to DNA methylation in specifying gene silencing. BMC Genomics. 8:1312007. View Article : Google Scholar : PubMed/NCBI

16 

Miao F, Wu X, Zhang L, Yuan YC, Riggs AD and Natarajan R: Genome-wide analysis of histone lysine methylation variations caused by diabetic conditions in human monocytes. J Biol Chem. 282:13854–13863. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Zhang L, Dai Y, Wang L, Peng W, Zhang Y, Ou Y and Lu J: CpG array analysis of histone H3 lysine 4 trimethylation in peripheral blood mononuclear cells of uremia patients. DNA Cell Biol. 30:179–186. 2011. View Article : Google Scholar

18 

Gulbins E, Coggeshall KM, Brenner B, Schlottmann K, Linderkamp O and Lang F: Fas-induced apoptosis is mediated by activation of a Ras and Rac protein-regulated signaling pathway. J Biol Chem. 271:26389–26394. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Esteve P, Embade N, Perona R, Jiménez B, del Peso L, León J, Arends M, Miki T and Lacal JC: Rho-regulated signals induce apoptosis in vitro and in vivo by a p53-independent, but Bcl2 dependent pathway. Oncogene. 17:1855–1869. 1998. View Article : Google Scholar : PubMed/NCBI

20 

Perletti L, Talarico D, Trecca D, Ronchetti D, Fracchiolla NS, Maiolo AT and Neri A: Identification of a novel gene, PSD, adjacent to NFKB2/lyt-10, which contains Sec7 and pleckstrin-homology domains. Genomics. 46:251–259. 1997. View Article : Google Scholar

21 

Okada S, Suzuki K, Takaharu K, Noda H, Kamiyama H, Maeda T, Saito M, Koizumi K, Miyaki Y and Konishi F: Aberrant methylation of the pleckstrin and Sec7 domain-containing gene is implicated in ulcerative colitis-associated carcinogenesis through its inhibitory effect on apoptosis. Int J Oncol. 40:686–694. 2012.

22 

Kang H, Cui K and Zhao K: BRG1 controls the activity of the retinoblastoma protein via regulation of p21CIP1/WAF1/SDI. Mol Cell Biol. 24:1188–1199. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Zhang HS, Gavin M, Dahiya A, Postigo AA, Ma D, Luo RX, Harbour JW and Dean DC: Exit from G1 and S phase of the cell cycle is regulated by repressor complexes containing HDAC-Rb-hSWI/SNF and Rb-hSWI/SNF. Cell. 101:79–89. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Maitra A, Wistuba II, Washington C, Virmani AK, Ashfaq R, Milchgrub S, Gazdar AF and Minna JD: High-resolution chromosome 3p allelotyping of breast carcinomas and precursor lesions demonstrates frequent loss of heterozygosity and a discontinuous pattern of allele loss. Am J Pathol. 159:119–130. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Wistuba II, Gazdar AF and Minna JD: Molecular genetics of small cell lung carcinoma. Semin Oncol. 28(Suppl 4): 3–13. 2001. View Article : Google Scholar : PubMed/NCBI

26 

DelBove J, Rosson G, Strobeck M, et al: Identification of a core member of the SWI/SNF complex, BAF155/SMARCC1, as a human tumor suppressor gene. Epigenetics. 6:1444–1453. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Xu Z, Liang L, Wang H, Li T and Zhao M: HCRP1, a novel gene that is downregulated in hepatocellular carcinoma, encodes a growth-inhibitory protein. Biochem Biophys Res Commun. 311:1057–1066. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Wittinger M, Vanhara P, El-Gazzar A, et al: hVps37A status affects prognosis and cetuximab sensitivity in ovarian cancer. Clin Cancer Res. 17:7816–7827. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Chang X, Li Z, Ma J, Deng P, Zhang S, Zhi Y, Chen J and Dai D: DNA methylation of NDRG2 in gastric cancer and its clinical significance. Dig Dis Sci. 58:715–723. 2013. View Article : Google Scholar

30 

Zhi Y, Chen J, Zhang S, Chang X, Ma J and Dai D: Down-regulation of CXCL12 by DNA hypermethylation and its involvement in gastric cancer metastatic progression. Dig Dis Sci. 57:650–659. 2012. View Article : Google Scholar

Related Articles

Journal Cover

May-2015
Volume 33 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu X, Liu J, Xu X, Zhang C and Dai D: Genome‑wide analysis of histone modifications by ChIP‑chip to identify silenced genes in gastric cancer. Oncol Rep 33: 2567-2574, 2015
APA
Zhu, X., Liu, J., Xu, X., Zhang, C., & Dai, D. (2015). Genome‑wide analysis of histone modifications by ChIP‑chip to identify silenced genes in gastric cancer. Oncology Reports, 33, 2567-2574. https://doi.org/10.3892/or.2015.3824
MLA
Zhu, X., Liu, J., Xu, X., Zhang, C., Dai, D."Genome‑wide analysis of histone modifications by ChIP‑chip to identify silenced genes in gastric cancer". Oncology Reports 33.5 (2015): 2567-2574.
Chicago
Zhu, X., Liu, J., Xu, X., Zhang, C., Dai, D."Genome‑wide analysis of histone modifications by ChIP‑chip to identify silenced genes in gastric cancer". Oncology Reports 33, no. 5 (2015): 2567-2574. https://doi.org/10.3892/or.2015.3824