Methylated +322-327 CpG site decreases hOGG1 mRNA expression in non-small cell lung cancer

  • Authors:
    • Yuanyuan Zeng
    • Jianjie Zhu
    • Hualong Qin
    • Dan Shen
    • Zhe Lei
    • Wei Li
    • Zongli Ding
    • Jian-An Huang
    • Zeyi Liu
  • View Affiliations

  • Published online on: June 1, 2017     https://doi.org/10.3892/or.2017.5690
  • Pages: 529-537
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

hOGG1 plays a role in several disease pathways, including various cancers. Despite such functional importance, how hOGG1 is regulated at the transcriptional level in human non-small cell lung cancer (NSCLC) remains unknown, particularly via DNA methylation changes. We obtained NSCLC tissues and adjacent non-cancerous tissues and examined hOGG1 mRNA expression levels. NSCLC cells were treated with 5-Aza to test whether DNA methylation can influence the expression of hOGG1. The MassARRAY EpiTYPER and luciferase reporter gene assays were used to define the functional region of the hOGG1 gene (including CpG sites). Finally, ChIP assay was utilized to verify transcription factor binding to the hOGG1 5'-UTR region. Our previous studies supported the idea that the methylation of the hOGG1 gene promoter region occurs frequently in NSCLC. Treatment with 5-Aza, a demethylating agent, led to a significant restoration of hOGG1 expression in NSCLC cell lines. Quantitative PCR and MassARRAY EpiTYPER assays demonstrated that methylation of the +322-327 CpG site in the 5'-UTR region of hOGG1 was higher in NSCLC tissues compared with adjacent non-cancerous tissues. Notably, the methylation level of +322-327 site (T/N) was inversely correlated with that of hOGG1 mRNA level (T/N) in 25 NSCLC tissues. ChIP assay and in silico prediction showed an association between the +322-327 CpG site and Sp1, which has been reported to be an activator of transcription. Importantly, luciferase reporter gene and ChIP assays showed that +322-327 CpG site methylation particularly reduced the recruitment of Sp1 to the 5'-UTR sequence in hOGG1 and reduced transcriptional activity ~50%. In summary, we have demonstrated that hOGG1 mRNA is downregulated in NSCLC tissues. Moreover, we identified that the methylated +322-327 CpG site in the hOGG1 5'-UTR is associated with reduced expression of hOGG1 by decreasing the recruitment of Sp1 to the 5'-UTR of hOGG1.

Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide (1,2). Non-small cell lung cancer (NSCLC) accounts for ~85% of lung cancer. Despite improvement in cancer treatment, the 5-year survival rate remains less than 10%. The 5-year survival rate is estimated to be 55–80% with an earlier lung cancer diagnosis or surgery at an early stage (3). Preferential understanding of how NSCLC develops and progresses plays an important role in early detection and prevention as well as targeted treatment of NSCLC.

Previous studies have shown that DNA repair pathways commonly exhibit functional overlap to ensure genomic stability; therefore, challenging the perception that distinctive lesions are repaired by different mechanisms in the mammalian genome. This finding is especially true for oxidative damaged DNA, such as oxidized bases (4), which might be caused by oxidant exposure or ionizing radiation, but could also result from normal cellular metabolism. Oxidized bases are mutagenic and cytotoxic, and several previous studies have shown that oxidized bases may contribute to neurodegeneration, aging and cancer (5). Base excision repair (BER) is an important DNA repair pathway that is responsible for the repair of DNA base damage and single strand breaks caused by X-rays, oxygen radicals or alkylating agents (6).

OGG1, a DNA repair glycosylase that localizes to both the nucleus and mitochondria, is the main enzyme responsible for the identification and excision of 8-oxoG lesions, which produces G:C to T:A transversions (79). OGG1 is one of the components of the BER pathway. The human OGG1 (hOGG1) gene is found on chromosome 3p26.2, which is one of the most frequent genomic deletion regions that contains some potential tumor suppressor genes in various types of tumors, such as NSCLCs (10). Previous studies suggested that hOGG1 plays a role in several disease pathways, including various cancers (1113). Despite such functional importance, how hOGG1 is regulated at the transcriptional level in NSCLC remains largely unclear, particularly via DNA methylation changes.

It is well known that epigenetic regulation, such as DNA methylation, can alter gene expression (1416). DNA methylation frequently occurs in CpG islands, which are frequently found in the 5′-untranslated regions (5′-UTR) of genes (17). DNA methylation changes at site-specific CpGs may play a crucial role in cancer progression, including hypermethylation of tumor suppressor genes and hypomethylation of oncogenes (14,18). Our previous studies have shown that DNA methylation underlies inactivation of the CpG island methylator phenotype (CIMP) and that TSGs on 3p might be a frequent epigenetic event that confers an increased risk of developing NSCLC (10,19). Moreover, we previously showed that a methylated +58 CpG site in the DCN 5′-UTR was associated with reduced DCN mRNA expression in highly metastatic NSCLC cells (20). Hence, we hypothesize that site-specific CpG methylation affects hOGG1 mRNA expression levels in NSCLC.

Materials and methods

Tissue samples

Seventy-seven paired NSCLC tissues and adjacent non-cancerous tissues were obtained after informed consent from patients in the First Affiliated Hospital of Soochow University between 2009 and 2013. Blood specimens were obtained after informed consent from 30 randomly selected NSCLC and paired non-cancerous lung patients. Blood was isolated by centrifugation at 3,500 rpm for 20 min after blood sampling (10). The Revised International System for Staging Lung Cancer was used to determine histological and pathological diagnostics for NSCLC patients. No chemotherapy or radiotherapy was given to patients with NSCLC before tissue sampling. Tissue samples were stored at −80°C after being snap-frozen. The present study was approved by the Ethics Committee of the First Affiliated Hospital of Soochow University.

Cell culture and drug treatment

Human lung carcinoma cell lines (A549, H1650, H460, SPC-A-1, 95C, 95D, H226 and SK-MES-1) were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China), and human bronchial epithelial (HBE) cells from Shanghai Bogoo Biotechnology, Co., Ltd. (Shanghai, China). Cell lines were seeded and grown in RPMI-1640 medium (HyClone Laboratories, Inc., Logan, UT, USA), with the exception of SK-MES-1, which was seeded in MEM medium (HyClone Laboratories), with 10% heat-inactivated fetal bovine serum (FBS; Gibco, Carlsbad, CA, USA) and L-glutamine and antibiotics (Invitrogen, Carlsbad, CA, USA) in a humidified incubator containing 5% CO2 at 37°C. Treatment with 5-aza-2′-deoxycitidine (5-Aza; Sigma-Aldrich, St. Louis, MO, USA) was used to demethylate cells in culture according to the previously described treatment protocol (10).

Quantitative determination of human 8-oxoguanine DNA glycosydase (hOGG1) concentrations and evaluation of DNA damage in serum

The human 8-oxoguanine DNA glycosydase (hOGG1) ELISA kit (cat. no. CSB-E12686h; Wuhan, Hubei, China) was used to quantitatively determine hOGG1 concentrations in serum. Briefly, standard controls and samples (100 µl) were added to each well and covered with a provided adhesive strip; cells were incubated at 37°C for 2 h. The standard and sample results were recorded in a plate layout that was provided by the manufacturer. Next, each well received 100 µl of biotin-antibody (1x), and the plate was covered with a new adhesive strip. Cells were incubated at 37°C for 1 h. If the biotin-antibody (1x) appeared cloudy, it was warmed to room temperature and mixed gently until the solution appeared uniform. Next, each well received 100 µl of HRP-avidin (1x), and the plate was covered with a new adhesive strip. Cells were incubated at 37°C for 1 h. The aspirate was washed five times, and 90 µl of TMB substrate was added to each well. Cells were incubated at 37°C for 15–30 min. Cell plates were protected from light. A total of 50 µl of stop solution was added to each well, thoroughly mixed and hOGG1 was detected using a microplate reader.

8-OHdG, 8-hydroxyguanine and its 2′-deoxynucleoside equivalent, 8-hydroxy-2′-deoxyguanosine (8-OHdG) are common byproducts of DNA damage. During the repair of damaged DNA in vivo by exonucleases, 8-hydroxy-2′-deoxyguanosine (8-OHdG) is excreted (21,22); therefore, we used an OxiSelect Oxidative DNA Damage ELISA kit (8-OHdG Quantitation; Cell Biolabs, Inc., San Diego, CA, USA) to evaluate the level of DNA damage in serum according to the manufacturers instructions (2325). The extracted DNA was dissolved in water to reach a concentration of 1–5 mg/ml. The DNA was converted to single-stranded DNA by incubating at 95°C for 5 min, then promptly chilling on ice. The denatured DNA samples were digested to nucleosides by incubating with 5–20 units of nuclease P1 at 37°C for 2 h in 20 mM sodium acetate (pH 5.2). DNA samples were treated with 5–10 units of alkaline phosphatase at 37°C for 1 h in 100 mM Tris (pH 7.5), followed by centrifugation for 5 min at 6,000 × g. Subsequently, the supernatant was used for the 8-OHdG enzyme-linked immunosorbent assay (ELISA).

RNA extraction, cDNA synthesis and quantitative real-time PCR (qRT-PCR)

Total RNA of cells and tissues were extracted by adding 1.0 ml RNAiso Plus (Takara Bio, Osaka, Japan) according to the manufacturers protocol. The RNA concentration was measured using a NanoDrop 2000 (Thermo Fisher Scientific, Waltham, MA, USA) and synthesis of cDNA was performed using Reverse Transcriptase M-MLV (Takara Bio) with reverse transcriptase. The sequences of qRT-PCR for hOGG1 and GAPDH were as follows: hOGG1, forward, 5′-ATCGTACTCTAGCCTCCACTCC-3′ and reverse, 5′-GTCAGTGTCCATACTTGATCCGC-3′; GAPDH, forward, 5′-TGCACCACCAACTGCTTAGC-3′ and reverse, 5′-TGCACCACCAACTGCTTAGC-3′. qRT-PCR was performed using SYBR Premix ExTaq™ (Takara Bio) according to the manufacturers instructions on an ABI StepOnePlus Real-Time PCR system (Applied Biosystems, Foster City, CA, USA). The PCR program was 50°C for 2 min, 95°C for 10 min, followed by 45 cycles of 95°C for 15 sec and 60°C for 1 min.

Quantitative methylation analysis of DNA was performed using MassARRAY EpiTYPER assays

Quantitative methylation analysis of DNA was performed using MassARRAY EpiTYPER assays (Sequenom, Inc., San Diego, CA, USA) according to the protocol recommended by the manufacturer (26). This system uses matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry in combination with RNA base-specific cleavage (Mass Cleave). After bisulfite modification, genomic DNA was amplified using MassArray primers. PCR products were introduced to a T7 promoter sequence by the Beijing Bio-Miao Biotechnology, Co., Ltd. (Beijing, China). Next, RNA products were transcribed in vitro using T-base-specific cleavage, in which small RNA fragments were obtained. The molecular weight of each fragment was detected by flight mass spectrometry (MALDI-TOF) and methylation levels were analyzed using EpiTyper software. PCR amplification bias was controlled for by using DNA methylation standards (0, 20, 40, 60, 80 and 100%) and data was normalized by correction algorithms based on an R statistical computing environment.

Construction of luciferase reporter plasmids, transient transfection and luciferase assay

To construct a plasmid containing the hOGG1 promoter, we used pGL3 basic vector (Promega, Madison, WI, USA). Briefly, an 88-bp fragment containing the predicted Sp1 target site (positions +322–327) was chosen for the luciferase assay. The wild-type and mutated fragment was directly synthesized (Genewiz, Suzhou, China) and subcloned into the pGL3 basic vector to generate pGL3-wild-type (WT: tggtccttgtctgggCGgggtctttgggCGtCGaCGaggcctggt tctggg taggCGgggctactaCGgggCGgtgcctgctgtggaa) and pGL3-mutant plasmid (Mut: tggtccttgtctgggCGgggtctttgggCGtCG aCGaggcctggttctgggtaggCGgggctactaCTggAATgtgcctgctgt ggaa). Subsequently, A549 and SPC-A1 cells were plated in a 24-well plate and cotransfected with wild-type plasmid, mutated plasmid, or pRL-TK plasmid using Lipofectamine 2000 (Life Technologies, Carlsbad, CA, USA). After 48 h, cells were collected, and luciferase activities were measured by the Dual-Luciferase reporter assay kit (Promega). Each experiment was performed in triplicate.

Chromatin immunoprecipitation (ChIP) assay

ChIP assay was carried out as previously described (20). Briefly, immunoprecipitation was performed using 5 µg anti-Sp1 antibody (Cell Signaling Technology, Beverly, MA, USA). Purified ChIP DNA was subjected to PCR, using primers specific for the hOGG1 promoter region (positions +247 to +398) encompassing the putative Sp1-binding site. Specific ChIP primers used for PCR were as follows: forward, 5-TAAGGGTCGTG GTCCTTGTC-3 and reverse, 5-TGGAGGCTAGAGTACGA TGC-3.

Results

Serum levels of hOGG1 are decreased and 8-OHdG levels are increased in NSCLC samples

hOGG1 gene encodes a DNA glycosylase that catalyzes the excision and removal of 8-OH-dG adducts (27). A previous report has shown a decrease of hOGG1 in the brain of Alzheimers patients (28), which caused the accumulation of 8-oxoG in the mitochondrial DNA of neurons and calpain-dependent neuronal loss (12). In addition, an association between the hOGG1 gene and lung cancer risk has been reported (27,29). Here, we detected the level of hOGG1 and 8-OH-dG using ELISA assays. As illustrated in Fig. 1A, the level of hOGG1 was lower in NSCLC serum than in paired normal serum. Furthermore, we detected 8-OH-dG and found that the level of 8-OH-dG was higher in NSCLC serum than in paired normal serum (Fig. 1B).

hOGG1 mRNA expression is downregulated in NSCLC tissues

hOGG1 mRNA levels were significantly lower in NSCLC tissues compared with adjacent non-cancerous lung tissues (P=0.034; Fig. 1D). No significant differences were observed in hOGG1 mRNA levels between NSCLC tissues classified by various clinicopathological characteristics (Table I). Moreover, a public data set (GSE19188) containing 91 NSCLC tissues and 65 normal lung tissues showed that hOGG1 mRNA expression was downregulated in human NSCLC tissues (P=0.002; Fig. 1C).

Table I.

Demographic and clinical characteristics of NSCLC patients and the association with hOGG1 mRNA expression in tumor tissue specimens.

Table I.

Demographic and clinical characteristics of NSCLC patients and the association with hOGG1 mRNA expression in tumor tissue specimens.

CharacteristicsNo. of cases (%)hOGG1 expressionP-value
Age (years)
  ≤6536 (46.8)0.0098±0.00110.6832
  >6541 (53.2)0.0091±0.0013
Sex
  Male52 (67.5)0.0092±0.00110.7653
  Female25 (32.5)0.0098±0.0014
Histology
  Adenocarcinomas35 (45.5)0.0119±0.00160.0707
  Squamous cell carcinomas29 (37.7)0.0073±0.0010
  Others13 (16.8)0.0075±0.0011
Smokers
  Yes44 (57.1)0.0098±0.00120.6264
  No33 (42.9)0.0090±0.0016
Clinical stage
  I21 (27.3)0.0077±0.00120.2214
  II19 (24.7)0.0112±0.0017
  III26 (33.8)0.0078±0.0012
  IV11 (14.2)0.0134±0.0036
Lymph node
  Yes36 (46.8)0.0095±0.00120.9264
  No41 (53.2)0.0093±0.0012
Distant metastases
  Yes11 (14.3)0.0134±0.00360.0562
  No66 (85.7)0.0088±0.0008

[i] Data are presented as mean ± SE. Unpaired t-test for two groups. Kruskal-wallis test for three or more groups.

hOGG1 mRNA expression is downregulated in NSCLC cell lines and associated with DNA methylation

As shown in Fig. 2A, hOGG1 mRNA levels were significantly lower in A549, H1650, H460, 95C, 95D, H226 and SK-MES-1 cells compared with control HBE cells, except for SPC-A-1. Our previous study supported the idea that DNA methylation could be epigenetically responsible for inactivation of tumor suppressor genes in NSCLC, and the methylation of the hOGG1 gene promoter region occurs frequently in NSCLC (10,19). Therefore, to determine whether methylation of hOGG1 gene promoter is an alternative mechanism underlying inactivation of hOGG1 mRNA expression, we detected the mRNA expression after using demethylating agent 5-Aza on NSCLC cell lines. As illustrated in Fig. 2B, after 5-Aza treatment, hOGG1 mRNA expression was increased in NSCLC cell lines (A549, H460, SPC-A1, 95D and SK-MES-1); therefore, we suggest that hOGG1 expression is silenced by DNA methylation.

Methylation levels of the +322–327 CpG site is higher in NSCLC than adjacent non-cancerous lung tissues and inversely correlated with hOGG1 mRNA expression

It is known that CpG islands are located −200 to −1,000 bp from the transcription start site of a gene. Based on this knowledge, we used Methyl Primer Express® software to identify potential CpG sites in the hOGG1 promoter and observed a GC-rich region (Fig. 3A). Furthermore, by using the MassARRAY EpiTYPER application, methylation levels of CpG sites in the hOGG1 gene were observed in 10 paired NSCLC tissues and adjacent non-cancerous lung tissues (Tables IIIV). In the present study, we detected three separate regions (position −1000 - −643, −463 - +34 and +35 - +412), including 68 CpG sites, in the hOGG1 promoter. Several CpG sites were detected between positions −1000 - −643 and −463 - +34, and differences in methylation levels were detect (P<0.05). However, we found that these regions have no transcriptional binding sites or have lower frequency of methylation. Consequently, we expanded the sample size to 25-paired tissues to detect potential CpG sites within the third area (position +35 - +412). We observed significantly higher methylation of CpG site-3 in NSCLC patients compared with the control group (Fig. 3B; Table V). Notably, the methylation level of +322 - 327 site (T/N) was inversely correlated with hOGG1 mRNA level (T/N) in 25 paired tissues (P=0.0104; Fig. 4C).

Table II.

CpG methylation of −1000 - −643 of the hOGG1 promoter in NSCLC and paired normal tissues.

Table II.

CpG methylation of −1000 - −643 of the hOGG1 promoter in NSCLC and paired normal tissues.

TumorParied-normal


No.CpG siten∑X/n±sn∑X/n±sP-value
1CpG_1100.417±0.175100.261±0.0610.016
2CpG_3101100.983±0.0180.010
3CpG_4.5100.933±0.018100.929±0.0140.594
4CpG_6100.900±0.024100.889±0.0260.339
5CpG_7.8100.951±0.015100.960±0.0090.129
6CpG_10100.835±0.064100.828±0.0820.835
7CpG_11.12100.938±0.013100.937±0.0120.863
8CpG_13101101

Table IV.

CpG methylation of +35 - +412 of the hOGG1 promoter in NSCLC and paired normal tissues.

Table IV.

CpG methylation of +35 - +412 of the hOGG1 promoter in NSCLC and paired normal tissues.

TumorParied-normal


No.CpG siten∑X/n±sn∑X/n±sP-value
  1CpG_2  90.088±0.11490.101±0.1370.840
  2CpG_3.4.590.031±0.05690.048±0.0680.554
  3CpG_6.790.356±0.20190.291±0.2210.520
  4CpG_890.035±0.04090.021±0.0350.431
  5CpG_9.1090.035±0.04490.023±0.0350.529
  6CpG_1190.195±0.19690.17±0.1660.769
  7CpG_1290.071±0.05590.063±0.0460.750
  8CpG_1490.022±0.01790.021±0.0180.898
  9CpG_1590.012±0.01690.017±0.0190.526
10CpG_16.1790.101±0.07590.073±0.0620.405
11CpG_18.1990.058±0.03290.054±0.0340.780
12CpG_2090.025±0.02690.032±0.0250.599
13CpG_21.2290.133±0.07390.078±0.0650.117
14CpG_2390.051±0.03990.035±0.0310.370
15CpG_24.2590.034±0.03090.025±0.0190.471

Table V.

CpG methylation of +35 - +412 of the hOGG1 promoter in NSCLC and paired normal tissues.

Table V.

CpG methylation of +35 - +412 of the hOGG1 promoter in NSCLC and paired normal tissues.

TumorParied-normal


No.CpG siten∑X/n±sn∑X/n±sP-value
  1CpG_2250.057±0.050250.055±0.0920.945
  2CpG_3.4.5250.010±0.033250.024±0.0450.210
  3CpG_6.7240.158±0.079240.092±0.0660.001
  4CpG_8250.044±0.028250.039±0.0300.535
  5CpG_9.10250.082±0.091250.034±0.0420.226
  6CpG_11250.111±0.145250.105±0.1200.874
  7CpG_12250.082±0.043250.074±0.0350.475
  8CpG_14250.033±0.024250.035±0.0350.815
  9CpG_15240.011±0.015240.006±0.0130.276
  10CpG_16.17240.140±0.058240.135±0.0950.827
  11CpG_18.19240.092±0.032240.092±0.0430.955
12CpG_20250.061±0.052250.054±0.0440.625
13CpG_21.22250.321±0.209250.306±0.1950.802
14CpG_23250.080±0.064250.048±0.0460.073
15CpG_24.25250.026±0.017250.035±0.0320.245
Methylation of the +322–327 CpG site inhibits hOGG1 mRNA expression by inhibiting Sp1 binding to the hOGG1 promoter region

Because methylation of individual CpG dinucleotides may contribute to cancer development (20,30), we postulated that site-specific CpG methylation could alter the expression of hOGG1 in NSCLC. We found one putative functional CpG site at position +322–327 in the proximal promoter region of hOGG1 that was located within a transcription factor Sp1-binding sequence (5-CGGGGCGG-3) using TRANSFAC, TFSEARCH and Methyl Primer Express® software (Fig. 4A). We utilized ChIP analysis to examine whether Sp1 binds to the hOGG1 proximal promoter region at the +322–327 CpG site, and found that Sp1 was recruited to the +322–327 CpG site in A549 cells (Fig. 4B). Collectively, the results demonstrated that Sp1 may be target hOGG1 5-promoter containing the +322–327 CpG site and thereby upregulate hOGG1 expression in NSCLC cells. Subsequently, we synthesized segments of the hOGG1 promoter that contained a wild-type and a mutant Sp1 binding site for use in luciferase reporter constructs; these constructs were transfected into A549 and SPC-A1 cells. The mutant Sp1 binding site construct displayed a significant decrease in luciferase activity compared with the wild-type construct (Fig. 4C). In summary, our results suggest that the methylation of the +322–327 site in the hOGG1 promoter represses Sp1 binding and regulates the expression of hOGG1 in NSCLC cells.

Discussion

Lung cancer is a leading cause of death throughout the world. The morbidity and mortality of lung cancer has significantly increased in the past decade in China (2). Studies have shown that NSCLC may result from the accumulation of multiple genetic and/or epigenetic aberrations. DNA methylation could be responsible for the inactivation of the tumor suppressor genes found in NSCLC.

Endogenous and exogenous sources cause oxidatively induced DNA damage in living organisms by a variety of mechanisms. The resulting DNA lesions are mutagenic and, unless repaired, lead to a variety of mutations and consequently to genetic instability, which is a hallmark of cancer. The BER is known to preserve genome integrity by removing damaged bases. It is the main pathway for the repair of oxidized modifications in both nuclear and mitochondrial DNA. Compelling evidence has shown that hOGG1 plays an important role in tumorigenesis (1113). Lower hOGG1 activity has been reported in patients with NSCLC and downregulation of hOGG1 mRNA and protein levels are compromised in their ability to remove 8-oxoG from their DNA (31,32). Despite such functional importance, it remains largely unknown how hOGG1 is regulated at the transcriptional level in human NSCLC, particularly through epigenetic mechanisms, such as DNA methylation. Genetic polymorphisms in individuals have recently been implicated to account for some of the observed differences in lung cancer susceptibility. The Ser326Cys hOGG1 polymorphism may be the most frequently reported; it is associated with increased risk for lung cancers, but its function is still controversial (33). Our previous studies showed that DNA methylation could underlie epigenetic inactivation of the CpG island methylator phenotype (CIMP) involving TSGs on 3p, suggesting that this is a frequent epigenetic event that may confer an increased risk of NSCLC (19,20). A previous study reported that the CpG methylation of an adjacent cytosine could moderately decrease the oxoGua excision rate, whereas methylation opposite oxoGua could lower the rate of product release (34).

We showed that +322–327 CpG methylation in the hOGG1 5′-UTR decreased hOGG1 mRNA expression in NSCLC tissues and cells using MassARRAY Epi-TYPER applications. Our findings revealed that +322–327 CpG methylation may reduce the recruitment of the transcriptional activator Sp1 to the hOGG1 5′-UTR. Sp1 is a well-characterized sequence-specific transcriptional factor that regulates a large number of housekeeping and tissue-specific genes by binding to GC-rich DNA sequences in the promoter region of many human genes (35,36).

Our findings support the idea that site-specific CpG methylation may play an important role in cancer progression (14,18,20,31). To date, the mechanistic roles of individual CpG site methylation are rarely reported in cancer (20,31). This encourages us to investigate how the +322–327 CpG site epigenetically affects the regulation of hOGG1 expression. In the present study, we identified that the +322–327 CpG in the hOGG1 proximal promoter is within a putative transcription factor Sp1-binding sequence. Furthermore, cell-based and biochemical analyses revealed that +322–327 CpG methylation can inhibit hOGG1 transcriptional expression by interfering with the recruitment of Sp1 to the hOGG1 promoter. However, we cannot exclude the possibility of the roles of other functional CpG sites in the hOGG1 promoter region.

Acknowledgements

We are grateful to all the patients who participated in the present study. This study was supported by grants from the National Natural Science Foundation of China (no. 31270940 to J.-A.H., no. 81201575 to Z.-Y.L.), the Jiangsu Province Colleges and Universities Natural Science Research Foundation (No.1 4KJB0017 to Z.L.), the Science and Technology Plan Projects of Suzhou (no. SYS201612 to Z.-Y.L.) the Foundation of Health Care Rejuvenation by Science and education (KJXW2016003 to Y.-Y.Z.), Huaian City Science and Technology Support Program (no. HAS2015013-4), the Clinical Medicine Center of Suzhou (no. Szzx201502), the Suzhou Key Laboratory for Respiratory Medicine (no. SZS201617), the Societal and Developmental Project of Suzhou (no. SS201630) and the Clinical Key Speciality Project of China.

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 

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

3 

Mulshine JL and Sullivan DC: Clinical practice. Lung cancer screening. N Engl J Med. 352:2714–2720. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Fortini P, Pascucci B, Parlanti E, DErrico M, Simonelli V and Dogliotti E: 8-Oxoguanine DNA damage: At the crossroad of alternative repair pathways. Mutat Res. 531:127–139. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Evans MD, Dizdaroglu M and Cooke MS: Oxidative DNA damage and disease: induction, repair and significance. Mutat Res. 567:1–61. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Fortini P and Dogliotti E: Base damage and single-strand break repair: Mechanisms and functional significance of short- and long-patch repair subpathways. DNA Repair (Amst). 6:398–409. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Klungland A, Rosewell I, Hollenbach S, Larsen E, Daly G, Epe B, Seeberg E, Lindahl T and Barnes DE: Accumulation of premutagenic DNA lesions in mice defective in removal of oxidative base damage. Proc Natl Acad Sci USA. 96:13300–13305. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Radicella JP, Dherin C, Desmaze C, Fox MS and Boiteux S: Cloning and characterization of hOGG1, a human homolog of the OGG1 gene of Saccharomyces cerevisiae. Proc Natl Acad Sci USA. 94:8010–8015. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Rosenquist TA, Zharkov DO and Grollman AP: Cloning and characterization of a mammalian 8-oxoguanine DNA glycosylase. Proc Natl Acad Sci USA. 94:7429–7434. 1997. View Article : Google Scholar : PubMed/NCBI

10 

Liu Z, Li W, Lei Z, Zhao J, Chen XF, Liu R, Peng X, Wu ZH, Chen J, Liu H, et al: CpG island methylator phenotype involving chromosome 3p confers an increased risk of non-small cell lung cancer. J Thorac Oncol. 5:790–797. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Sampath H, Vartanian V, Rollins MR, Sakumi K, Nakabeppu Y and Lloyd RS: 8-Oxoguanine DNA glycosylase (OGG1) deficiency increases susceptibility to obesity and metabolic dysfunction. PLoS One. 7:e516972012. View Article : Google Scholar : PubMed/NCBI

12 

Sheng Z, Oka S, Tsuchimoto D, Abolhassani N, Nomaru H, Sakumi K, Yamada H and Nakabeppu Y: 8-Oxoguanine causes neurodegeneration during MUTYH-mediated DNA base excision repair. J Clin Invest. 122:4344–4361. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Peng Y, Li Z, Zhang S, Xiong Y, Cun Y, Qian C, Li M, Ren T, Xia L, Cheng Y, et al: Association of DNA base excision repair genes (OGG1, APE1 and XRCC1) polymorphisms with outcome to platinum-based chemotherapy in advanced nonsmall-cell lung cancer patients. Int J Cancer. 135:2687–2696. 2014. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

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

16 

Das PM and Singal R: DNA methylation and cancer. J Clin Oncol. 22:4632–4642. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Sekido Y, Fong KM and Minna JD: Progress in understanding the molecular pathogenesis of human lung cancer. Biochim Biophys Acta. 1378:F21–F59. 1998.PubMed/NCBI

18 

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

19 

Liu Z, Zhao J, Chen XF, Li W, Liu R, Lei Z, Liu X, Peng X, Xu K, Chen J, et al: CpG island methylator phenotype involving tumor suppressor genes located on chromosome 3p in non-small cell lung cancer. Lung Cancer. 62:15–22. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Qian Q, Shi X, Lei Z, Zhan L, Liu RY, Zhao J, Yang B, Liu Z and Zhang HT: Methylated +58CpG site decreases DCN mRNA expression and enhances TGF-β/Smad signaling in NSCLC cells with high metastatic potential. Int J Oncol. 44:874–882. 2014.PubMed/NCBI

21 

Loft S, Vistisen K, Ewertz M, Tjønneland A, Overvad K and Poulsen HE: Oxidative DNA damage estimated by 8-hydroxydeoxyguanosine excretion in humans: Influence of smoking, gender and body mass index. Carcinogenesis. 13:2241–2247. 1992. View Article : Google Scholar : PubMed/NCBI

22 

Loft S, Poulsen HE, Vistisen K and Knudsen LE: Increased urinary excretion of 8-oxo-2-deoxyguanosine, a biomarker of oxidative DNA damage, in urban bus drivers. Mutat Res. 441:11–19. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Bhat HK and Singh B: Induction of NAD(P)H-quinone oxidoreductase 1 by antioxidants in female ACI rats is associated with decrease in oxidative DNA damage and inhibition of estrogen-induced breast cancer. Carcinogenesis. 3:156–163. 2012.

24 

Tzortzaki EG, Dimakou K, Neofytou E, Tsikritsaki K, Samara K, Avgousti M, Amargianitakis V, Gousiou A, Menikou S and Siafakas NM: Oxidative DNA damage and somatic mutations: A link to the molecular pathogenesis of chronic inflammatory airway diseases. Chest. 141:1243–1250. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Burnham EL, McCord JM, Bose S, Brown LA, House R, Moss M and Gaydos J: Protandim does not influence alveolar epithelial permeability or intrapulmonary oxidative stress in human subjects with alcohol use disorders. Am J Physiol Lung Cell Mol Physiol. 302:L688–L699. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Ehrich M, Nelson MR, Stanssens P, Zabeau M, Liloglou T, Xinarianos G, Cantor CR, Field JK and van den Boom D: Quantitative high-throughput analysis of DNA methylation patterns by base-specific cleavage and mass spectrometry. Proc Natl Acad Sci USA. 102:15785–15790. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Park J, Chen L, Tockman MS, Elahi A and Lazarus P: The human 8-oxoguanine DNA N-glycosylase 1 (hOGG1) DNA repair enzyme and its association with lung cancer risk. Pharmacogenetics. 14:103–109. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Iida T, Furuta A, Nishioka K, Nakabeppu Y and Iwaki T: Expression of 8-oxoguanine DNA glycosylase is reduced and associated with neurofibrillary tangles in Alzheimers disease brain. Acta Neuropathol. 103:20–25. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Mambo E, Chatterjee A, De Souza-Pinto NC, Mayard S, Hogue BA, Hoque MO, Dizdaroglu M, Bohr VA and Sidransky D: Oxidized guanine lesions and hOgg1 activity in lung cancer. Oncogene. 24:4496–4508. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Chen H, Yang T, Lei Z, Wang L, Yang H, Tong X, Yang WT, Zhao J, Gu Y, Chen Y, et al: RNF111/Arkadia is regulated by DNA methylation and affects TGF-β/Smad signaling associated invasion in NSCLC cells. Lung Cancer. 90:32–40. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Sevilya Z, Leitner-Dagan Y, Pinchev M, Kremer R, Elinger D, Rennert HS, Schechtman E, Freedman LS, Rennert G, Paz-Elizur T, et al: Low integrated DNA repair score and lung cancer risk. Cancer Prev Res (Phila). 7:398–406. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Leitner-Dagan Y, Sevilya Z, Pinchev M, Kramer R, Elinger D, Roisman LC, Rennert HS, Schechtman E, Freedman L, Rennert G, et al: N-methylpurine DNA glycosylase and OGG1 DNA repair activities: Opposite associations with lung cancer risk. J Natl Cancer Inst. 104:1765–1769. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Li H, Hao X, Zhang W, Wei Q and Chen K: The hOGG1 Ser326Cys polymorphism and lung cancer risk: A meta-analysis. Cancer Epidemiol Biomarkers Prev. 17:1739–1745. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Kasymov RD, Grin IR, Endutkin AV, Smirnov SL, Ishchenko AA, Saparbaev MK and Zharkov DO: Excision of 8-oxoguanine from methylated CpG dinucleotides by human 8-oxoguanine DNA glycosylase. FEBS Lett. 587:3129–3134. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Black AR, Black JD and Azizkhan-Clifford J: Sp1 and Krüppel-like factor family of transcription factors in cell growth regulation and cancer. J Cell Physiol. 188:143–160. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Suske G: The Sp-family of transcription factors. Gene. 238:291–300. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 38 Issue 1

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
Zeng Y, Zhu J, Qin H, Shen D, Lei Z, Li W, Ding Z, Huang J and Liu Z: Methylated +322-327 CpG site decreases hOGG1 mRNA expression in non-small cell lung cancer. Oncol Rep 38: 529-537, 2017
APA
Zeng, Y., Zhu, J., Qin, H., Shen, D., Lei, Z., Li, W. ... Liu, Z. (2017). Methylated +322-327 CpG site decreases hOGG1 mRNA expression in non-small cell lung cancer. Oncology Reports, 38, 529-537. https://doi.org/10.3892/or.2017.5690
MLA
Zeng, Y., Zhu, J., Qin, H., Shen, D., Lei, Z., Li, W., Ding, Z., Huang, J., Liu, Z."Methylated +322-327 CpG site decreases hOGG1 mRNA expression in non-small cell lung cancer". Oncology Reports 38.1 (2017): 529-537.
Chicago
Zeng, Y., Zhu, J., Qin, H., Shen, D., Lei, Z., Li, W., Ding, Z., Huang, J., Liu, Z."Methylated +322-327 CpG site decreases hOGG1 mRNA expression in non-small cell lung cancer". Oncology Reports 38, no. 1 (2017): 529-537. https://doi.org/10.3892/or.2017.5690