DNA methyltransferase DNMT1 inhibits lipopolysaccharide‑induced inflammatory response in human dental pulp cells involving the methylation changes of IL‑6 and TRAF6

  • Authors:
    • Luhui Cai
    • Minkang Zhan
    • Qimeng Li
    • Di Li
    • Qiong Xu
  • View Affiliations

  • Published online on: December 4, 2019     https://doi.org/10.3892/mmr.2019.10860
  • Pages: 959-968
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Dental pulp inflammation is a pathological process characterized by local lesions in dental pulp and the accumulation of inflammatory mediators. DNA methylation of cytosine residues is a key epigenetic modification that is essential for gene transcription, and plays pivotal roles in inflammatory reactions and immune responses. However, the function of cytosine DNA methylation in the innate immune defense against the inflammation of dental pulp is poorly understood. To investigate the effect of DNA methylation in inflamed dental pulp upon innate immune responses, expression levels of the DNA methyltransferases (DNMT1, DNMT3a and DNMT3b) in human dental pulp cells (hDPCs) after lipopolysaccharide (LPS) stimulation were evaluated by western blotting and reverse transcription‑quantitative (RT‑q) PCR. Only DNMT1 expression was decreased, while the transcription of inflammatory cytokines was increased. In the immune responses of LPS‑induced hDPCs, the results of RT‑qPCR and ELISA showed that DNMT1 knockdown promoted the production of the pro‑inflammatory cytokines, interleukin (IL)‑6 and IL‑8. Western blotting demonstrated that DNMT1 knockdown increased the phosphorylation levels of IKKα/β and p38 in the NF‑κB and MAPK signaling pathways, respectively. Furthermore, MeDIP and RT‑qPCR analysis demonstrated that the 5‑methylcytosine levels of the IL‑6 and TNF receptor‑associated factor 6 (TRAF6) promoters were significantly decreased in DNMT1‑deficient hDPCs. Taken together, these results indicated that the expression of DNMT1 was decreased after LPS stimulation in hDPCs. DNMT1 depletion increased LPS‑induced cytokine secretion, and activated NF‑κB and MAPK signaling; these mechanisms may involve the decreased methylation levels of the IL‑6 and TRAF6 gene promoters. This study emphasized the role of DNMT1‑dependent DNA methylation on the inflammation of LPS‑infected dental pulp and provides a new rationale for the investigation of the molecular mechanisms of inflamed dental pulps.

Introduction

Dental pulp inflammation is a pathological process characterized by various bacterial virulence factors that often elicits a dental emergency, and may develop into periapical disease or pulp necrosis (1). Lipopolysaccharide (LPS) is commonly released from gram-negative bacteria. When LPS enters the dental pulp, it can evoke an inflammatory response; LPS is also closely associated with pulpitis and periapical periodontitis (2). Previous studies have found that LPS can stimulate Toll-like receptor (TLR)4 in the cell membranes of human dental pulp cells (hDPCs), and activate the NF-κB, ERK1/2 and p38 pathways, thereby producing inflammation-related cytokines, including interleukin (IL)-6 and IL-8 (3,4). Although there are a number of mechanisms associated with the development of dental pulp infection, the specific molecular mechanism is still unclear (5,6). Recent studies have suggested that epigenetic alterations are crucial regulators in the occurrence and development of dental pulp infection (79).

DNA methylation that occurs at cytosine-phosphate-guanine (CpG) dinucleotide sites is the most common epigenetic modification event in the genome (10). The DNA methylation process involves placing a methyl group onto the 5-position of cytosines situated in CpG dinucleotides and turning the cytosine into 5-methylcytosine (5mC), which is catalyzed by members of the DNA methyltransferase (DNMT) family (11). DNMT1 can methylate hemimethylated CpGs and is a well-known maintenance methyltransferase that can preserve methylation patterns during DNA replication (12). DNMT3a and DNMT3b are de novo methyltransferases that can methylate unmethylated and hemimethylated DNA, and establish DNA methylation patterns in embryo development (13). The roles and functions of DNA methylation patterns have attracted extensive attention, but there has been particular emphasis on their roles in the pathological processes of cancer (14). Only recently have studies begun to shed light on the contribution of DNMTs to the initiation and progression of inflammatory diseases (15). A study on inflamed peripheral blood mononuclear cells (PBMCs) showed that DNMT1 expression decreased after treatment with LPS; DNMT1 modulated the methylation level of gene promoters, thus mediating the transcription of pro-inflammatory cytokines, including IL-6, IL-8 and tumor necrosis factor-α (TNF-α) (16). In macrophages, DNMT1 contributes to the hypermethylation of suppressor of cytokine signaling 1, a negative regulator of cytokine signal transduction, thereby enhancing the secretion of pro-inflammatory cytokines induced by LPS indirectly (17). DNA methylation could also affect inflammatory reactions by modulating the activation levels of crucial proteins of the NF-κB and/or MAPK pathways (18,19). In addition, DNA methylation epigenetically regulates the transcription of TLRs and signal transduction molecules, including TNF receptor-associated factor 6 (TRAF6) and myeloid differentiation primary response 88 (MyD88). This suggests that DNA methylation is engaged in signaling pathways related to inflammation (20,21). These studies provide evidence indicating that DNA methylation can epigenetically regulate inflammatory reactions via several different mechanisms. However, whether DNA methylation is involved in the modification of dental pulp immunity remains unclear.

Preliminary experiments by our lab showed that in LPS-treated hDPCs, 5-aza-2′-deoxycytidine (5-Aza-CdR), a DNA methyltransferase inhibitor, increased the production of several inflammation-related cytokines (unpublished data). The present study aimed to investigate the effect of DNMT1 on the LPS-induced inflammatory response in hDPCs, thereby exploring the role of DNA methylation in dental pulp inflammation. The results demonstrated that DNMT1 knockdown promoted the expression of pro-inflammatory cytokines and the phosphorylation of IKKα/β and p38 in LPS-treated hDPCs. Moreover, DNMT1 depletion decreased the 5mC level in the IL-6 and TRAF6 promoters. These data suggested that DNMT1 may be involved in inhibiting the LPS-induced inflammatory response in hDPCs.

Materials and methods

Isolation and culture of hDPCs

Healthy permanent premolars and third molars were collected from donors aged 18 to 25 for orthodontic reasons from the Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Sun Yat-sen University for approximately one year between March 2018 and April 2019. Only healthy teeth without carious disease or hyperemic pulp tissue were selected. A total of 128 teeth from 58 donors (29 males and 29 females) were obtained for dental pulp tissue isolation and cell culture. hDPCs were isolated and cultivated using an enzymatic method as described by Gronthos et al (22). After extraction, the teeth were washed with 70% ethanol and PBS (pH 7.4) and then split open to expose the pulp chamber. The dental pulp tissue was gently isolated with forceps and minced into small pieces, which were then digested in 3 mg/ml collagenase type I (Gibco; Thermo Fisher Scientific, Inc.) for 20 min at 37°C. Subsequently, the minced pulp tissue was cultured in DMEM containing 20% FBS, 100 mg/ml streptomycin and 100 U/ml penicillin (all purchased from Gibco; Thermo Fisher Scientific, Inc.) at 37°C with 5% CO2. The medium was changed every 3 days. When the cells reached 80% confluence, they were detached using trypsin/EDTA (Gibco; Thermo Fisher Scientific, Inc.) and subcultured at a ratio of 1:2. Generally, 2–3 teeth from one donor were used for each primary culture. For each primary culture, ~106 cells at the zero passage were obtained. All experiments were performed with cells from passages two or three. To avoid inter-individual variation, the experiments were performed at least three times for each sample and each experiment, and average data were generated. For each parameter, experiments were replicated three times each using donor cells from three samples, and average data for the three different cell types were obtained.

Treatment with LPS

hDPCs were stimulated for the indicated times (0, 3, 6, 12 and 24 h) with 1 µg/ml purified Escherichia coli (E. coli) LPS (Sigma-Aldrich; Merck KGaA) at 37°C with 5% CO2 (4,23). The blank controls were cells without LPS stimulation.

DNMT1 small interfering RNA (siRNA) transfection

siRNA was used in hDPCs to knockdown DNMT1. A total of 3 siRNA sequences (Invitrogen; Thermo Fisher Scientific, Inc.) were designed to target the human DNMT1 gene. Before transfection, hDPCs were seeded in 6-well plates in 2 ml of α-MEM at 4×105 cells/well containing 10% FBS for 24 h. After attachment overnight, hDPCs were then transfected with siRNA (50 nM) against DNMT1 or a nontargeting siRNA control using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. After incubation for 24 h, the media was changed, and DMEM supplemented with 10% FBS was added with or without 1 µg/ml E. coli LPS. All siRNA sequences are listed in Table I. siRNA #1 with the best interference effect was selected as the DNMT1 target sequence for the subsequent experiments.

Table I.

Sequences used for DNMT1 siRNA.

Table I.

Sequences used for DNMT1 siRNA.

DNMT1 siRNASequence (5′-3′)
#1 siRNASense: GGGACUGUGUCUCUGUUAUTT dTdT
Antisense: dTdT AUAACAGAGACACAGUCCCTT
#2 siRNASense: GCACCUCAUUUGCCGAAUATT dTdT
Antisense: dTdT UAUUCGGCAAAUGAGGUGCTT
#3 siRNASense: GAGGCCUAUAAUGCAAAGATT dTdT
Antisense: dTdT UCUUUGCAUUAUAGGCCUCTT

[i] DNMT1, DNA methyltransferases; siRNA, small interfering RNA.

Reverse transcription quantitative (RT-q)PCR

Cells were lysed using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's protocols, and RNA was extracted and reverse transcribed into cDNA with a RevertAid First Strand cDNA Synthesis kit (Fermentas; Thermo Fisher Scientific, Inc.). PCR was performed using the complementary DNA as a template. SYBR-Green I (Roche Diagnostics) RT-qPCR results were detected by a LightCycler® 480 thermal cycler. Thermal cycling conditions consisted of initial denaturation at 95°C for 5 min, followed by 45 cycles of 95°C for 10 sec, 65°C for 20 sec and 72°C for 30 sec. The relative results were normalized to the GAPDH mRNA levels (24). The primer sequences were designed using Primer Express Software v3.0.1 (Thermo Fisher Scientific, Inc.) and are listed in Table II.

Table II.

Primers used for the analysis of mRNA levels by reverse transcription-quantitative PCR.

Table II.

Primers used for the analysis of mRNA levels by reverse transcription-quantitative PCR.

GenePrimer sequences (5′-3′)
DNMT1F: GGCTGAGATGAGGCAAAAAG
R: ACCAACTCGGTACAGGATGC
DNMT3 AF: AGGGAAGACTCGATCCTCGTC
R: GTGTGTAGCTTAGCAGACTGG
DNMT3 BF: GCCTCAATGTTACCCTGGAA
R: CAGCAGATGGTGCAGTAGGA
IL-6F: TGCAATAACCACCCCTGACC
R: AGCTGCGCAGAATGAGATGA
IL-8F: GGTGCAGTTTTGCCAAGGAG
R: TTCCTTGGGGTCCAGACAGA
GAPDHF: TCTCCTCTGACTTCAACAGCGACA
R: CCCTGTTGCTGTAGCCAAATTCGT

[i] IL, interleukin; F, forward; R, reverse.

Western blot analysis

Protein was extracted from hDPCs using RIPA buffer (Beyotime Institute of Biotechnology), and the concentrations were detected using a BCA Protein Assay kit (Beyotime Institute of Biotechnology). Proteins (30 µg) were separated using electrophoresis on 8% SDS-polyacrylamide gels and transferred to PVDF membranes (EMD Millipore). Next, the membranes were blocked with TBS-Tween 20 (20 mmol Tris-HCl, 150 mmol NaCl, 0.05% Tween-20) containing 5% BSA (Biofroxx; neoFroxx GmbH) for 1 h at room temperature. Then, the membranes were incubated with primary antibodies against DNMT1 (1:2,000; Abcam), IκB kinase αβ (IKKαβ), phosphorylated (p)-IKKαβ, p65, p-p65, IκBα, p-IκBα (1:1,000; NF-κB Pathway Sampler kit, 9936, Cell Signaling Technology, Inc.), p38, ERK, JNK (1:1,000; MAPK Family Antibody Sampler kit, 9926, Cell Signaling Technology, Inc.), p-p38, p-ERK, p-JNK (1:1,000; phospho-MAPK Family Antibody Sampler kit, 9910, Cell Signaling Technology, Inc.) and GAPDH (1:1,000; Cell Signaling Technology, Inc.) overnight at 4°C. After rinsing, the membranes were incubated with horseradish peroxidase-conjugated secondary antibodies (1:2,000; AQ160P and AP307P, EMD Millipore) at room temperature for 1 h. An enhanced chemiluminescence system (EMD Millipore) was used to visualize the antibody binding. The relative protein expression levels were normalized to that of the GAPDH gene, and the protein band densities were determined by ImageJ v1.47 software (National Institutes of Health). ReBlot Plus (EMD Millipore) was used to strip and re-probe with the p-antibodies for IκBα, p38, ERK and JNK to distinguish different target proteins when they share similar molecular weights with the total IκBα, p38, ERK and JNK, respectively, on the same membrane.

ELISA

Human IL-6 ELISA kits (D6050, R&D Systems, Inc.) and Human IL-8 ELISA kits (D8000C, R&D Systems, Inc.) were used to analyze the culture supernatant protein concentrations of IL-6 and IL-8 collected after LPS stimulation for 6 h according to the manufacturer's protocols. A microplate reader (Tecan Safire microplate reader; Tecan Group, Ltd.) was used to evaluate the optical density (OD) values. Based on the standard solution concentration and corresponding OD value, sample concentrations were calculated.

Methylated DNA immunoprecipitation (MeDIP) and RT-qPCR

DNA was extracted from hDPCs and fragmented to 200–500-bp fragments with a Bioruptor Waterbath Sonicator (8 cycles, 15 sec on/15 sec off, at the highest output level while cooling the tube to 1°C in a waterbath). Then, the DNA fragments were diluted to 700 µl with TE buffer (Invitrogen; Thermo Fisher Scientific, Inc.) with 60 µl Protein G Magnetic Beads (S1430S; New England BioLabs, Inc.) and denatured for 10 min at 94°C. Following denaturation, DNA was immunoprecipitated at 4°C overnight with an anti-5mC antibody (1:40, C02010031; Diagenode SA). Then it was incubated for 2 h with anti-IgG Magnetic Beads (S1430S; New England BioLabs, Inc.) at 4°C with agitation. The beads were trapped on a magnetic rack, the supernatant discarded, and washed three times with 1 ml 1XIP buffer [2 mM EDTA, 20 mM Tris (pH=8.0), 1% Triton X-100, 0.1% SDS, 150 mM NaCl] for 10 min at 4°C with agitation. Beads were then resuspended in 400 µl of Elution Buffer (50 mM Tris-HCl, pH=8.0; 10 mM EDTA, Ph=8.0; 1% SDS) with 10 µl of Proteinase K (Qiagen GmbH). IP with non-specific human IgG was measured as a negative control. After IP, the DNA samples were eluted using phenol-chloroform and precipitated using ethanol. After resuspending the precipitated samples in 10 µl Tris buffer, RT-qPCR was performed using 1 µl harvested DNA fragments. The primers designed for MeDIP-PCR are shown in Table III.

Table III.

Primers used for methylated DNA immunoprecipitation PCR.

Table III.

Primers used for methylated DNA immunoprecipitation PCR.

GenePrimer sequences (5′-3′)
IL6F: TGGCAGCACAAGGCAAACC
R: GCTTCAGCCCACTTAGAGGAGG
IL8F: TAGGAAGTGTGATGACTCAGGTT
R: GTCAGAGGAAATTCCACGATT
TRAF6F: GCTTACTGTAGCCTTGACTGCC
R: GTGGTGCATATCTGTAGTCTCGG
MYD88F: TTCGCTCACCGACACAGATG
R: GGTCACTGCGGCTGCTCTT

[i] IL, interleukin; TRAF6, TNF receptor-associated factor 6; MyD88, myeloid differentiation primary response 88; F, forward; R, reverse.

Statistical analysis

All experiments were carried out at least three times. The data were analyzed by the SPSS 20.0 software package (IBM Corp.) and are shown as the mean ± SD. Student's t-test was used to measure the differences between two groups. To evaluate the differences in multiple sets of data, one-way ANOVA or repeated-measures ANOVA with a post hoc Dunnett's test was performed. P<0.05 was considered statistically significant.

Results

DNMT1 expression in LPS-inflamed hDPCs

To detect the effect of LPS on the inflammatory reaction in hDPCs, hDPCs were stimulated with LPS at a concentration of 1 µg/ml for the indicated times. As illustrated in Fig. 1A and B, compared with the control group, IL-6 and IL-8 mRNA and protein expression was significantly increased by LPS. IL-6 and IL-8 expression was upregulated and peaked after 3 h, which was followed by a gradual decrease. The levels of DNMT1 mRNA were significantly reduced within 24 h after treatment with LPS. DNMT1 protein expression also decreased, with the most significant change at 3 h (Fig. 1C and D). Moreover, the mRNA expression of DNMT3a and DNMT3b did not change significantly before or after LPS treatment (Fig. 1).

Effects of DNMT1 on inflammatory cytokine expression in LPS-induced hDPCs

Our preliminary study found that 5-Aza-CdR, a DNMT inhibitor, can increase the secretion of inflammatory cytokines in LPS-stimulated hDPCs, and among the upregulated cytokines, IL-6 and IL-8 experienced the greatest increase (unpublished data). To investigate the effect of DNMT1-dependent methylation on inflammatory cytokine production in hDPCs stimulated with LPS, the IL-6 and IL-8 expression levels after DNMT1 knockdown in hDPCs transfected with siRNAs were measured. As shown in Fig. 2A and B, after DNMT1 siRNA (#1, #2 and #3) interference, DNMT1 mRNA expression levels were significantly reduced when compared to the negative control group. These data were further confirmed by western blotting, which showed a reduction in the protein expression. Among the DNMT1 siRNAs, the siRNA #1 group showed the best interference effect at ~72% (Fig. 2B). Therefore, siRNA #1 was selected as the DNMT1 target sequence for the subsequent experiments.

IL-6 and IL-8 gene expression levels were then measured in cells stimulated by LPS after DNMT1 depletion (Fig. 2C). The results showed that IL-6 and IL-8 mRNA expression within 24 h after LPS stimulation was notably higher in the DNMT1 knockdown group compared with the control group. In LPS-inflamed hDPCs, the protein levels of IL-6 and IL-8 were also significantly increased after DNMT1 knockdown (Fig. 2D).

Effects of DNMT1 on the NF-κB signaling pathway in LPS-induced hDPCs

One of the most important signaling pathways that influences inflammatory cytokine production in inflammation induced by LPS is the NF-κB signaling pathway (25). By means of western blotting, the phosphorylation levels of three crucial proteins of the NF-κB signaling pathway were examined (IKKα/β, p65 and IκBα) to determine whether DNMT1 is engaged in NF-κB pathway activation. As illustrated in Fig. 3A and B, DNMT1 knockdown significantly increased the phosphorylation of IKKα/β at 15 and 30 min after LPS treatment in hDPCs. The p65 and IκBα phosphorylation levels also increased at several time points, but there was no significant difference.

Effects of DNMT1 on the MAPK signaling pathway in LPS-induced hDPCs

Another vital signaling transduction pathway involved in inflammation in the LPS-related inflammatory response is the MAPK signaling pathway (26). The phosphorylation levels of three key proteins in the MAPK signaling pathway were assessed (p38, ERK1/2 and JNK) to determine whether DNMT1 plays an important role in MAPK signaling pathway activation. As illustrated in Fig. 4A and B, after DNMT1 knockdown in LPS-inflamed hDPCs, the p38 phosphorylation level was increased, while both p-ERK and p-JNK levels were not significantly altered.

Effects of DNMT1 on the dynamic methylation levels of the IL-6, IL-8, TRAF6 and MyD88 gene promoters in LPS-induced hDPC inflammation

DNA methylation can regulate the occurrence and progression of inflammatory responses by modulating the methylation levels of inflammation-related cytokines and signaling molecule promoters (27). TRAF6 and MyD88 are key intracellular signal transducers of LPS-induced signaling pathways (28). To identify whether the methylation of IL-6, IL-8, TRAF6 and MyD88 was regulated through DNMT1, the levels of 5mC present at their gene promoters were examined by means of MeDIP-PCR. The results illustrated that the levels of 5mC at the IL-6 and TRAF6 promoters decreased notably in LPS-stimulated hDPCs after DNMT1 knockdown. However, no significant change was observed in the 5mC levels of the IL-8 and MyD88 promoters (Fig. 5). These experimental results indicated that DNMT1 can modulate the methylation of IL-6 and TRAF6 in hDPCs stimulated by LPS.

Discussion

As a major component of the outer membrane of gram-negative bacteria, LPS serves as the primary pathogenic factor leading to dental pulp inflammation (29). When healthy dental pulp cells are exposed to LPS, pro-inflammatory chemokines and cytokines, including IL-6 and IL-8, are released, thus triggering subsequent inflammatory events (2). DNA methylation is a major epigenetic regulator that can influence the transcriptional expression of pro-inflammatory cytokines in the initiation and development of the inflammatory response (1517). However, very little research has sought to define the function of DNA demethylation in the development of the LPS-inflamed dental pulp.

DNA methylation plays a pivotal role in a wide range of inflammatory diseases, and aberrant DNA methylation is often observed in some inflammation-related conditions (30). DNMT1 expression is increased in the rectal epithelium during ulcerative colitis progression in patients and may be a relatively early event in ulcerative colitis-associated tumorigenesis; consequently, this factor may be useful for predicting the risk of colorectal neoplasia in ulcerative colitis (31). In periodontitis, treating human oral keratinocytes with LPS downregulated DNMT1 expression (32). In Sjögren's syndrome, the global DNA methylation level in patient salivary gland epithelial cells was reduced, with a 7-fold decrease in DNMT1 but no significant difference in DNMT3a/b expression (33). To determine the relationship between DNMTs and LPS-inflamed dental pulp, the expression of three DNMTs in LPS-treated hDPCs were examined. After LPS stimulation, both mRNA and protein expression levels of DNMT1 decreased and reached their lowest level 3 h after stimulation. In addition, the mRNA expression levels of DNMT3a and DNMT3b fluctuated but did not differ significantly. These results suggested that DNMT1-dependent methylation may be involved in the inflammatory progression of dental pulp.

Researchers previously demonstrated that DNA methylation can function as a key epigenetic regulator in the pathogenesis of inflammation-related diseases (34,35). LPS stimulation can induce pro-inflammatory cytokine expression, and the methylation status of their gene promoters is involved in regulating the inflammatory response. In bovine endometrial cells, treatment with LPS can increase IL-6 and IL-8 mRNA expression and decrease the methylation levels of specific CpG sites at the IL-6 promoter (at −366 and −660) and the IL-8 promoter (at −120 and −48) (35). Treating PBMCs with LPS induces the expression of pro-inflammatory cytokines, including IL-6, TNF-α and IL-1β, while also demethylating the IL-6 gene at the −302 and −264 CpG sites, as well as the TNF-α gene at the −371 CpG site (36). However, in human intestinal epithelial cells, the 5 CpG sites located near the IL-8 transcription start site (−83, −7, +73, +119 and +191) were unmethylated on the lower and upper strands in both LPS treated and untreated groups (37). In our previous research, SEQUENOM MassARRAY was used to measure the methylation levels of the IL-6 and IL-8 promoters in hDPCs after LPS stimulation. The results showed that the methylation level at the −276 CpG site in the IL-6 promoter decreased after LPS stimulation. However, there was no difference in the methylation level of the IL-8 promoter (unpublished data). In the present study, to investigate the function of DNMT1 in inflammatory cytokine production by hDPCs after LPS stimulation, DNMT1 knockdown in hDPCs was established through siRNA transfection. The expression of DNMT1 was significantly decreased following depletion of DNMT1, which is consistent with our previous research (20,38). Next, the LPS-stimulated cytokine expression after knocking down DNMT1 was examined. DNMT1 silencing prominently enhanced the production of the cytokines IL-6 and IL-8, thereby indicating that DNMT1 may be a regulator that negatively targets cytokine accumulation in hDPCs inflamed by LPS.

It is commonly known that the MAPK and NF-κB signaling pathways play critical roles in mediating inflammatory reactions and are likely regulated by DNA methylation (19,39). In aged mouse macrophages, phosphorylation of IκBα in the NF-κB signaling pathway was increased after treatment with the demethylation agent 5-Aza-CdR (40). 5-Aza-CdR also increased IκBα and IKKα/β phosphorylation levels to promote the activation of NF-κB signaling in gastric cancer cells (41). A study on lung tissue inflammation revealed that 5-Aza-CdR can markedly decrease p38, JNK and ERK phosphorylation levels, thereby inhibiting MAPK signaling pathway activation under LPS stimulation (19). The levels of DNA methylation were affected in 27 gene promoters of the MAPK pathway in PBMCs and plasma samples from children who were constantly exposed to air pollutants (42). To explore whether DNA methylation influences the signaling pathways in LPS-treated hDPCs, the phosphorylation levels of several important signaling molecules in the MAPK and NF-κB signaling pathways were examined. The data from the present study showed that compared to LPS exposure alone, DNMT1 depletion upregulated the phosphorylation levels of IKKα/β in the NF-κB signaling pathway and the phosphorylation level of p38 in the MAPK signaling pathway. Therefore, DNMT1 suppressed both the MAPK and NF-κB signaling pathways in LPS-stimulated hDPCs, further confirming that DNMT1 acts as a negative regulator in inflamed hDPCs.

Previous studies have proposed that DNA methylation not only affects the methylation level of inflammatory cytokine promoters, but also changes the methylation status of intracellular signal transducers of signaling pathways (43). TRAF6 and MyD88, key intracellular signal transducers of the MAPK and NF-κB signaling pathways, can be regulated by DNA methylation (20,21). TRAF6 hypermethylation has been linked to low TRAF6 gene expression levels in PBMCs during inflammatory bowel diseases (21). In addition, MyD88 was shown to have consistently higher methylation levels in its promoter region in moderate localized aggressive periodontitis (LAP) than in severe LAP (44). In patients with LAP, the methylation level of the MyD88 promoter is negatively associated with several cyto/chemokines, such as IL-8 and IL-6 (44). In the present study, to explore whether these signal transduction factors are regulated by DNA methylation in LPS-treated hDPCs, MeDIP and RT-qPCR were used to analyze the dynamic 5mC levels of the IL-6, IL-8, TRAF6 and MyD88 gene promoters in DNMT1-deficient cells. Notably, the 5mC levels of the IL-6 and TRAF6 gene promoters decreased, suggesting that DNMT1 knockdown downregulated 5mC at the IL-6 and TRAF6 gene promoters. Although a modest decrease in the IL-8 and MyD88 gene promoter 5mC levels was observed, there were no significant differences. These observations indicated that DMNT1 can mediate the 5mC level of IL-6 and TRAF6 in LPS-inflamed hDPCs.

In summary, the present study showed that stimulating hDPCs with LPS decreased the expression of the DNA methyltransferase DNMT1. DNMT1 depletion increased LPS-induced cytokine secretion in hDPCs, and activated NF-κB and MAPK signaling. Furthermore, silencing DNMT1 was involved in downregulating methylation levels at the promoters of IL-6 and TRAF6. This study indicated that DNMT1-dependent DNA methylation plays a role in the inflammatory response of hDPCs stimulated by LPS, and provides a novel rationale for researchers to further reveal the molecular mechanisms of inflamed dental pulp.

Acknowledgements

Not applicable.

Funding

The present study was financially supported by the National Natural Science Foundation of China (grant no. 81771058).

Availability of data and materials

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

Authors' contributions

QX designed the study and provided scientific leadership to junior colleagues. LC and MZ performed the experiments and statistically analyzed the results. LC wrote the manuscript. QL and DL analyzed data, providing constructive comments. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was authorized by the institutional Ethical Review Boards of the Guanghua School of Stomatology of Sun Yat-sen University, and written informed consent for this investigation was provided from all patients who participated in the experiment in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bindal P, Ramasamy TS, Kasim NHA, Gnanasegaran N and Chai WL: Immune responses of human dental pulp stem cells in lipopolysaccharide-induced microenvironment. Cell Biol Int. 42:832–840. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Renard E, Gaudin A, Bienvenu G, Amiaud J, Farges JC, Cuturi MC, Moreau A and Alliot-Licht B: Immune cells and molecular networks in experimentally induced pulpitis. J Dent Res. 95:196–205. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Li JG, Lin JJ, Wang ZL, Cai WK, Wang PN, Jia Q, Zhang AS, Wu GY, Zhu GX and Ni LX: Melatonin attenuates inflammation of acute pulpitis subjected to dental pulp injury. Am J Transl Res. 7:66–78. 2015.PubMed/NCBI

4 

Feng Z, Li Q, Meng R, Yi B and Xu Q: METTL3 regulates alternative splicing of MyD88 upon the lipopolysaccharide-induced inflammatory response in human dental pulp cells. J Cell Mol Med. 22:2558–2568. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Song F, Sun H, Wang Y, Yang H, Huang L, Fu D, Gan J and Huang C: Pannexin3 inhibits TNF-α-induced inflammatory response by suppressing NF-κB signaling pathway in human dental pulp cells. J Cell Mol Med. 21:444–455. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Hui T, A P, Zhao Y, Yang J, Ye L and Wang C: EZH2 regulates dental pulp inflammation by direct effect on inflammatory factors. Arch Oral Biol. 85:16–22. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Hui T, Wang C, Chen D, Zheng L, Huang D and Ye L: Epigenetic regulation in dental pulp inflammation. Oral Dis. 23:22–28. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Bei Y, Tianqian H, Fanyuan Y, Haiyun L, Xueyang L, Jing Y, Chenglin W and Ling Y: ASH1L suppresses matrix metalloproteinase through mitogen-activated protein kinase signaling pathway in pulpitis. J Endod. 43:306.e2–314.e2. 2017. View Article : Google Scholar

9 

Cardoso FP, Viana MB, Sobrinho AP, Diniz MG, Brito JA, Gomes CC, Moreira PR and Gomez RS: Methylation pattern of the IFN-gamma gene in human dental pulp. J Endod. 36:642–646. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Smith ZD and Meissner A: DNA methylation: Roles in mammalian development. Nat Rev Genet. 14:204–220. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Moore LD, Le T and Fan G: DNA methylation and its basic function. Neuropsychopharmacology. 38:23–38. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Loo SK, Ab Hamid SS, Musa M and Wong KK: DNMT1 is associated with cell cycle and DNA replication gene sets in diffuse large B-cell lymphoma. Pathol Res Pract. 214:134–143. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Auclair G, Guibert S, Bender A and Weber M: Ontogeny of CpG island methylation and specificity of DNMT3 methyltransferases during embryonic development in the mouse. Genome Biol. 15:5452014. View Article : Google Scholar : PubMed/NCBI

14 

Kettunen E, Hernandez-Vargas H, Cros MP, Durand G, Le Calvez-Kelm F, Stuopelyte K, Jarmalaite S, Salmenkivi K, Anttila S, Wolff H, et al: Asbestos-associated genome-wide DNA methylation changes in lung cancer. Int J Cancer. 141:2014–2029. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Qiu J, Zhang YN, Zheng X, Zhang P, Ma G and Tan H: Notch promotes DNMT-mediated hypermethylation of Klotho leads to COPD-related inflammation. Exp Lung Res. 44:368–377. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Shen J, Wu S, Guo W, Liang S, Li X and Yang X: Epigenetic regulation of pro-inflammatory cytokine genes in lipopolysaccharide-stimulated peripheral blood mononuclear cells from broilers. Immunobiology. 222:308–315. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Cheng C, Huang C, Ma TT, Bian EB, He Y, Zhang L and Li J: SOCS1 hypermethylation mediated by DNMT1 is associated with lipopolysaccharide-induced inflammatory cytokines in macrophages. Toxicol Lett. 225:488–497. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Ma SC, Hao YJ, Jiao Y, Wang YH, Xu LB, Mao CY, Yang XL, Yang AN, Tian J, Zhang MH, et al: Homocysteine-induced oxidative stress through TLR4/NF-κB/DNMT1-mediated LOX-1 DNA methylation in endothelial cells. Mol Med Rep. 16:9181–9188. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Huang X, Kong G, Li Y, Zhu W, Xu H, Zhang X, Li J, Wang L, Zhang Z, Wu Y, et al: Decitabine and 5-azacitidine both alleviate LPS induced ARDS through anti-inflammatory/antioxidant activity and protection of glycocalyx and inhibition of MAPK pathways in mice. Biomed Pharmacother. 84:447–453. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Meng R, Li D, Feng Z and Xu Q: MyD88 hypermethylation mediated by DNMT1 is associated with LTA-induced inflammatory response in human odontoblast-like cells. Cell Tissue Res. 376:413–423. 2019. View Article : Google Scholar : PubMed/NCBI

21 

McDermott E, Ryan EJ, Tosetto M, Gibson D, Burrage J, Keegan D, Byrne K, Crowe E, Sexton G, Malone K, et al: DNA methylation profiling in inflammatory bowel disease provides new insights into disease pathogenesis. J Crohns Colitis. 10:77–86. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Gronthos S, Mankani M, Brahim J, Robey PG and Shi S: Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA. 97:13625–13630. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Jung JY, Woo SM, Kim WJ, Lee BN, Nör JE, Min KS, Choi CH, Koh JT, Lee KJ and Hwang YC: Simvastatin inhibits the expression of inflammatory cytokines and cell adhesion molecules induced by LPS in human dental pulp cells. Int Endod J. 50:377–386. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Liu J, Guo S, Jiang K, Zhang T, Zhiming W, Yaping Y, Jing Y, Shaukat A and Deng G: miR-488 mediates negative regulation of the AKT/NF-κB pathway by targeting Rac1 in LPS-induced inflammation. J Cell Physiol. 2019. View Article : Google Scholar

26 

Shang L, Wang T, Tong D, Kang W, Liang Q and Ge S: Prolyl hydroxylases positively regulated LPS-induced inflammation in human gingival fibroblasts via TLR4/MyD88-mediated AKT/NF-κB and MAPK pathways. Cell Prolif. 51:e125162018. View Article : Google Scholar : PubMed/NCBI

27 

Matt SM, Lawson MA and Johnson RW: Aging and peripheral lipopolysaccharide can modulate epigenetic regulators and decrease IL-1β promoter DNA methylation in microglia. Neurobiol Aging. 47:1–9. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Wang Q, Zhou X, Zhao Y, Xiao J, Lu Y, Shi Q, Wang Y, Wang H and Liang Q: Polyphyllin I ameliorates collagen-induced arthritis by suppressing the inflammation response in macrophages through the NF-κB pathway. Front Immunol. 9:20912018. View Article : Google Scholar : PubMed/NCBI

29 

Love RM and Jenkinson HF: Invasion of dentinal tubules by oral bacteria. Crit Rev Oral Biol Med. 13:171–183. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Barnicle A, Seoighe C, Greally JM, Golden A and Egan LJ: Inflammation-associated DNA methylation patterns in epithelium of ulcerative colitis. Epigenetics. 12:591–606. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Fujii S, Katake Y and Tanaka H: Increased expression of DNA methyltransferase-1 in non-neoplastic epithelium helps predict colorectal neoplasia risk in ulcerative colitis. Digestion. 82:179–186. 2010. View Article : Google Scholar : PubMed/NCBI

32 

de Camargo Pereira G, Guimarães GN, Planello AC, Santamaria MP, de Souza AP, Line SR and Marques MR: Porphyromonas gingivalis LPS stimulation downregulates DNMT1, DNMT3a, and JMJD3 gene expression levels in human HaCaT keratinocytes. Clin Oral Investig. 17:1279–1285. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Thabet Y, Le Dantec C, Ghedira I, Devauchelle V, Cornec D, Pers JO and Renaudineau Y: Epigenetic dysregulation in salivary glands from patients with primary Sjögren's syndrome may be ascribed to infiltrating B cells. J Autoimmun. 41:175–181. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Hedrich CM and Tsokos GC: Epigenetic mechanisms in systemic lupus erythematosus and other autoimmune diseases. Trends Mol Med. 17:714–724. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Wang J, Yan X, Nesengani LT, Ding H, Yang L and Lu W: LPS-induces IL-6 and IL-8 gene expression in bovine endometrial cells ‘through DNA methylation’. Gene. 677:266–272. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Angrisano T, Pero R, Peluso S, Keller S, Sacchetti S, Bruni CB, Chiariotti L and Lembo F: LPS-induced IL-8 activation in human intestinal epithelial cells is accompanied by specific histone H3 acetylation and methylation changes. BMC Microbiol. 10:1722010. View Article : Google Scholar : PubMed/NCBI

37 

Shen J, Liu Y, Ren X, Gao K, Li Y, Li S, Yao J and Yang X: Changes in DNA methylation and chromatin structure of pro-inflammatory cytokines stimulated by LPS in broiler peripheral blood mononuclear cells. Poult Sci. 95:1636–1645. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Mo Z, Li Q, Cai L, Zhan M and Xu Q: The effect of DNA methylation on the miRNA expression pattern in lipopolysaccharide-induced inflammatory responses in human dental pulp cells. Mol Immunol. 111:11–18. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Jangiam W, Tungjai M and Rithidech KN: Induction of chronic oxidative stress, chronic inflammation and aberrant patterns of DNA methylation in the liver of titanium-exposed CBA/CaJ mice. Int J Radiat Biol. 91:389–398. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Jiang M, Xiang Y, Wang D, Gao J, Liu D, Liu Y, Liu S and Zheng D: Dysregulated expression of miR-146a contributes to age-related dysfunction of macrophages. Aging Cell. 11:29–40. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Kim TW, Lee SJ, Oh BM, Lee H, Uhm TG, Min JK, Park YJ, Yoon SR, Kim BY, Kim JW, et al: Epigenetic modification of TLR4 promotes activation of NF-κB by regulating methyl-CpG-binding domain protein 2 and Sp1 in gastric cancer. Oncotarget. 7:4195–4209. 2016.PubMed/NCBI

42 

Carmona JJ, Sofer T, Hutchinson J, Cantone L, Coull B, Maity A, Vokonas P, Lin X, Schwartz J and Baccarelli AA: Short-term airborne particulate matter exposure alters the epigenetic landscape of human genes associated with the mitogen-activated protein kinase network: A cross-sectional study. Environ Health. 13:942014. View Article : Google Scholar : PubMed/NCBI

43 

Wang X, Feng Z, Li Q, Yi B and Xu Q: DNA methylcytosine dioxygenase ten-eleven translocation 2 enhances lipopolysaccharide-induced cytokine expression in human dental pulp cells by regulating MyD88 hydroxymethylation. Cell Tissue Res. 373:477–485. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Shaddox LM, Mullersman AF, Huang H, Wallet SM, Langaee T and Aukhil I: Epigenetic regulation of inflammation in localized aggressive periodontitis. Clin Epigenetics. 9:942017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 21 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cai L, Zhan M, Li Q, Li D and Xu Q: DNA methyltransferase DNMT1 inhibits lipopolysaccharide‑induced inflammatory response in human dental pulp cells involving the methylation changes of IL‑6 and TRAF6. Mol Med Rep 21: 959-968, 2020
APA
Cai, L., Zhan, M., Li, Q., Li, D., & Xu, Q. (2020). DNA methyltransferase DNMT1 inhibits lipopolysaccharide‑induced inflammatory response in human dental pulp cells involving the methylation changes of IL‑6 and TRAF6. Molecular Medicine Reports, 21, 959-968. https://doi.org/10.3892/mmr.2019.10860
MLA
Cai, L., Zhan, M., Li, Q., Li, D., Xu, Q."DNA methyltransferase DNMT1 inhibits lipopolysaccharide‑induced inflammatory response in human dental pulp cells involving the methylation changes of IL‑6 and TRAF6". Molecular Medicine Reports 21.2 (2020): 959-968.
Chicago
Cai, L., Zhan, M., Li, Q., Li, D., Xu, Q."DNA methyltransferase DNMT1 inhibits lipopolysaccharide‑induced inflammatory response in human dental pulp cells involving the methylation changes of IL‑6 and TRAF6". Molecular Medicine Reports 21, no. 2 (2020): 959-968. https://doi.org/10.3892/mmr.2019.10860