SHORT COMMUNICATION

Genomic DNA Extracted from Lactiplantibacillus plantarum Attenuates Porphyromonas gingivalis Lipopolysaccharide (LPS)-Induced Inflammatory Responses via Suppression of Toll-Like Receptor (TLR)-Mediated Mitogen-Activated Protein Kinase (MAPK) and Nuclear Factor-κB (NF-κB) Signaling Pathways

Young Hyeon Choi1,https://orcid.org/0000-0002-4954-7766, Bong Sun Kim2,https://orcid.org/0000-0001-7676-6238, Seok-Seong Kang1,*https://orcid.org/0000-0001-7029-9122
Author Information & Copyright
1Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Korea
2Division of Applied Food System, Major in Food Science & Technology, Seoul Women’s University, Seoul 01797, Korea

† These authors contributed equally to this work.

*Corresponding author : Seok-Seong Kang, Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Korea, Tel: +82-31-961-5150, Fax: +82-31-961-5108, E-mail: sskang@dgu.ac.kr

© Korean Society for Food Science of Animal Resources. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Jul 07, 2023 ; Revised: Aug 04, 2023 ; Accepted: Aug 04, 2023

Published Online: Sep 01, 2023

Abstract

In the present study, we aimed to examine the inhibition of genomic DNA from Lactiplantibacillus plantarum (LpDNA) on Porphyromonas gingivalis lipopolysaccharide (PgLPS)-induced inflammatory responses in RAW264.7 cells. Pretreatment with LpDNA for 15 h significantly inhibited PgLPS-induced mRNA expression and protein secretion of interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein-1. LpDNA pretreatment also reduced the mRNA expression of Toll-like receptor (TLR)2 and TLR4. Furthermore, LpDNA inhibited the phosphorylation of mitogen-activated protein kinases (MAPKs) and the activation of nuclear factor-κB (NF-κB) induced by PgLPS. Taken together, these findings demonstrate that LpDNA attenuates PgLPS-induced inflammatory responses by regulating MAPKs and NF-κB signaling pathways through the suppression of TLR2 and TLR4 expression.

Keywords: Lactiplantibacillus plantarum; genomic DNA; Porphyromonas gingivalis; periodontitis; inflammation

Introduction

Probiotics are live microorganisms that are well known to bring beneficial effects to the host and have been extensively studied as a therapeutic intervention by alleviating intestinal inflammatory responses and normalizing gut mucosal dysfunction (Borchers et al., 2009). Previous studies have demonstrated that not only whole live bacteria of probiotics but also cell wall components, bacterial lysates, enzymes, and amino acids from probiotics exert immunomodulatory effects (Izuddin et al., 2020; Kaji et al., 2010; Żółkiewicz et al., 2020). Genomic DNA of probiotics has also been recognized as a component that is capable of modulating immune responses. Indeed, genomic DNA extracted from Bifidobacterium and Lactobacillus has been shown to stimulate the production of interleukin (IL)-1β, IL-6, and IL-10 in human peripheral blood mononuclear cells (Lammers et al., 2003). Additionally, genomic DNA derived from Lactiplantibacillus plantarum is known to inhibit Escherichia coli lipopolysaccharide (LPS)-induced inflammatory responses in THP-1 cells (Kim et al., 2012).

Oral diseases impose a huge economic burden worldwide, with periodontitis representing one of the most common dental infections (Tonetti et al., 2017). Periodontitis is known as a chronic inflammatory disease present in tissues supporting the tooth, which destroys the alveolar bone and may result in tooth loss (Könönen et al., 2019). The pathogenesis of periodontitis is largely attributed to Porphyromonas gingivalis, a Gram-negative anaerobic bacteria that expresses several virulence factors (Mysak et al., 2014). Among these factors, P. gingivalis LPS is considered a key pathogenic component of periodontal diseases (Wang and Ohura, 2002). P. gingivalis LPS, which is recognized by Toll-like receptor (TLR)2 and TLR4, activates inflammatory signaling pathways and thus various pro-inflammatory cytokines and chemokines, including IL-1β, IL-6, and monocyte chemoattractant protein-1 (MCP-1; Darveau et al., 2004; Kato et al., 2014; Zhou et al., 2005).

Many previous studies have shown that probiotics, such as Lactobacillus spp., can reduce inflammatory responses and alleviate oral diseases (Gatej et al., 2018; Zhao et al., 2012). However, side effects have been reported for probiotics, including systemic infections and excessive immune stimulation (Doron and Snydman, 2015). Therefore, bioactive compounds from probiotics, known as postbiotics, have been introduced as novel alternatives (Moradi et al., 2020). In this study, we aimed to assess the postbiotic potential of genomic DNA from the probiotic strain Lactiplantibacillus plantarum against periodontitis by evaluating the anti-inflammatory effects of L. plantarum genomic DNA on P. gingivalis LPS-induced inflammatory responses.

Materials and Methods

Lactiplantibacillus plantarum culture and genomic DNA purification

L. plantarum ATCC 14917 was obtained from the American Type Cell Culture (ATCC; Manassas, VA, USA) and cultured as described previously (Kim et al., 2022). Genomic DNA of L. plantarum (LpDNA) was extracted using a Wizard Genomic DNA Purification Kit (Promega, Madison, WI, USA) following the manufacturer’s instructions with slight modifications. Briefly, bacteria were harvested by centrifuging and extensively washed with phosphate-buffered saline. The pellets were resuspended in 50 mM EDTA and lysozyme (10 mg/mL) followed by incubation at 37°C for 1 h. After centrifugation to obtain lysates, nuclei lysis solution (600 μL) was added to the lysates and incubated at 80°C for 5 min. Subsequently, RNase solution (3 μL) was added to the lysates and further incubated at 37°C for 40 min. Thereafter, the lysates were mixed with protein precipitation solution (200 μL), incubated on ice for 5 min, and centrifuged. After centrifugation, the supernatants were obtained and mixed with 600 μL of isopropanol and centrifuged as described above. The remaining lysates were washed with 70% ethanol, rehydrated by adding DNA rehydration solution, and incubated for 1 h at 65°C. The concentration and purity of LpDNA were measured using a NanoDrop spectrophotometer (ND-1000, NanoDrop Technologies, Hampton, NH, USA).

Cell culture

RAW 264.7, the murine macrophage cell line, was purchased from the ATCC (Manassas, VA, USA) and maintained in Dulbecco’s modified Eagle’s medium (DMEM; Welgene, Gyeongsan, Korea) supplemented with 10% heat-inactivated fetal bovine serum (Gibco, Burlington, ON, Canada), 100 U/mL penicillin, and 100 μg/mL streptomycin (HyClone, Logan, UT, USA) at 37°C in a humidified incubator under 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RAW 264.7 cells were plated in a 12-well culture plate and grown at 37°C for 24 h. The cells were pretreated with LpDNA (1 μg/mL) for 3, 9, or 15 h and subsequently stimulated with P. gingivalis LPS (PgLPS; 1 μg/mL) for a further 3 h. Total RNA was extracted and complementary DNA (cDNA) was synthesized with total RNA, random hexamers, and reverse transcriptase (Promega). cDNA was amplified by qPCR using a StepOnePlusTM Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) with SYBR Green Real-Time PCR Master Mix (Toyobo, Osaka, Japan). The amplification was conducted as follows: denaturation at 95°C for 10 s and amplification for 40 cycles at 95°C for 15 s and 60°C for 31 s. The sequences of specific PCR primers used in the study were as follows: IL-1β, forward, 5′-CTCACAAGCAGAGCACAAGC-3′ and reverse, 5′-TCTTGGCCGAGGACTAAGGA-3′; IL-6, forward, 5′-TCCTACCCCAATTTCCAATGCT-3′ and reverse, 5′-TCTGACCACAGTGAGGAATGTC-3′; MCP-1, forward, 5′-AGCCAACTCTCACTGAAGCC-3′ and reverse, 5′-TCTCC AGCCTACTCATTGGGA-3;′ TLR2, forward, 5’-CTGCGAAGTGGAAACCATCC-3′ and reverse, 5′-GCACTTCTGTTCC TCGCAGA-3′; TLR4, forward 5′-ACTCAGCAAAGTCCCTGATGAC-3′ and reverse, 5′-AGTTTGAGAGGTGGTGTAAGCC-3′; and β-actin, forward, 5′-TACAGCTTCACCACCACAGC-3′ and reverse, 5′-GGAAAAGAGCCTCAGGGCAT-3′. The relative mRNA expression of IL-1β, IL-6, MCP-1, TLR2, and TLR4 genes was normalized to that of β-actin using the 2−ΔΔCt method.

Enzyme-linked immunosorbent assay (ELISA)

RAW 264.7 cells were plated in a 48-well culture plate and incubated at 37°C for 24 h. The cells were pretreated with LpDNA (1 μg/mL) for 15 h and stimulated with PgLPS (1 μg/mL) for a further 24 h. The secretion of IL-1β, IL-6, and MCP-1 in the spent culture supernatants was analyzed using commercial ELISA kits (R&D Systems, Minneapolis, MN, USA).

Western blot analysis

RAW 264.7 cells were incubated at 37°C for 24 h and pretreated with LpDNA (1 μg/mL) for 15 h and stimulated with PgLPS (1 μg/mL) for a further 30 min. Cell lysates were extracted using a lysis buffer containing enzyme inhibitors. Samples were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and electrotransferred onto polyvinylidene difluoride membranes (Millipore, Bedford, MA, USA). After blocking the membranes with 5% skimmed milk in Tris-buffered saline containing 0.1% Tween 20, the membranes were incubated with primary antibodies specific for extracellular signal-regulated kinase (ERK), phosphorylated ERK, p38 kinase, phosphorylated p38 kinase, c-Jun-N-terminal kinase (JNK), phosphorylated JNK, IκBα (Cell Signaling Technology, Danvers, MA, USA), or β-actin (Santa Cruz Biotechnology, Santa Cruz, CA, USA) at 4°C overnight. The membranes were incubated with horseradish peroxidase-conjugated anti-rabbit IgG (Cell Signaling Technology) or anti-mouse IgG (Santa Cruz Biotechnology) for 2 h at room temperature. Immunoreactive bands were detected and analyzed with an enhanced chemiluminescence reagent (Dyne Bio, Seongnam, Korea) and C-DiGit blot scanner (LI-COR Biosciences, Lincoln, NE, USA).

Statistical analysis

The results were collected from at least three independent experiments. The results are expressed as means±SDs. Statistical significance between groups was evaluated by one-way analysis of variance (ANOVA) at p<0.05 using IBM SPSS Statistics 23 software (IBM, Armonk, NY, USA).

Results

Lactiplantibacillus plantarum DNA (LpDNA) inhibits Porphyromonas gingivalis lipopolysaccharide (PgLPS)-induced interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein-1 (MCP-1) mRNA expression after 15 h of pretreatment

To investigate whether LpDNA inhibits PgLPS-induced IL-1β, IL-6, and MCP-1 mRNA expression, RAW 264.7 cells were pretreated with LpDNA for 3, 9, or 15 h and subsequently stimulated with PgLPS for 3 h. As expected, PgLPS alone significantly induced IL-1β, IL-6, and MCP-1 mRNA expression. Although pretreatment with LpDNA for 3 h moderately inhibited PgLPS-induced IL-1β mRNA expression, LpDNA did not significantly inhibit PgLPS-induced IL-6 and MCP1 mRNA expression (Fig. 1A). Similarly, pretreatment with LpLTA for 9 h inhibited IL-1β mRNA expression induced by PgLPS, but not IL-6 and MCP-1 mRNA expression (Fig. 1B). However, pretreatment with LpDNA remarkably inhibited PgLPS-induced mRNA expression of those inflammatory mediators (Fig. 1C). Moreover, when RAW 264.7 cells were cotreated with LpDNA and PgLPS, LpDNA failed to inhibit PgLPS-induced IL-1β, IL-6, and MCP-1 mRNA expression (data not shown), suggesting that pretreatment with LpDNA for 15 h effectively inhibited PgLPS-induced inflammatory responses. Further experiments were conducted to investigate the anti-inflammatory effect of LpDNA after 15 h of pretreatment.

kosfa-43-5-938-g1
Fig. 1. Inhibitory effects of LpDNA pretreatment on Porphyromonas gingivalis LPS-induced IL-1β, IL-6, and MCP-1 in RAW 264.7 cells. Cells were pretreated with LpDNA (1 μg/mL) for 3 h (A), 9 h (B), or 15 h (C) and stimulated with P. gingivalis LPS (1 μg/mL) for an additional 3 h. IL-1β, IL-6, and MCP-1 mRNA expression were measured using qRT-PCR. The results are represented as the means SDs from three independent experiments. Statistical significance between groups was determined by ANOVA. p<0.05 was considered statistically significant. a–c Letters on each graph indicate significant difference (p<0.05). IL, interleukin; PgLPS, Porphyromonas gingivalis lipopolysaccharide; LpDNA, Lactiplantibacillus plantarum DNA; MCP-1, monocyte chemoattractant protein-1; qRT-PCR, quantitative reverse transcription-polymerase chain reaction; ANOVA, analysis of variance.
Download Original Figure
Lactiplantibacillus plantarum DNA (LpDNA) significantly inhibits the Porphyromonas gingivalis lipopolysaccharide (PgLPS)-induced interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein-1 (MCP-1) secretion

To investigate whether LpDNA inhibits the PgLPS-induced IL-1β, IL-6, and MCP-1 protein secretion, RAW 264.7 cells were pretreated with LpDNA for 15 h and subsequently stimulated with PgLPS for 24 h. As shown in Fig. 2, PgLPS significantly increased the protein secretion of IL-1β, IL-6, and MCP-1 in the absence of LpDNA pretreatment. However, IL-1β, IL-6, and MCP-1 protein secretion was dramatically decreased by pretreatment with LpDNA. These results suggest that LpDNA effectively inhibits PgLPS-induced inflammatory responses.

kosfa-43-5-938-g2
Fig. 2. Inhibitory effects of LpDNA pretreatment on Porphyromonas gingivalis LPS-induced IL-1β, IL-6, and MCP-1 secretion in RAW 264.7 cells. Cells were pretreated with LpDNA (1 μg/mL) or for 15 h and stimulated with P. gingivalis LPS (1 μg/mL) for an additional 24 h. IL-6, IL-1β, and MCP-1 protein secretion was assessed by ELISA. The results are represented as the means±SDs from three independent experiments. Statistical significance between groups was determined by ANOVA. p<0.05 was considered statistically significant. a–c Letters on each graph indicate significant difference (p<0.05). IL, interleukin; PgLPS, Porphyromonas gingivalis lipopolysaccharide; LpDNA, Lactiplantibacillus plantarum DNA; MCP-1, monocyte chemoattractant protein-1; ELISA, enzyme-linked immunosorbent assay; ANOVA, analysis of variance.
Download Original Figure
Lactiplantibacillus plantarum DNA (LpDNA) attenuates mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) signaling cascades through suppressing Toll-like receptor (TLR)2 and TLR4 expression

Both TLR2 and TLR4 are known to be simultaneously activated following recognition of PgLPS, resulting in the induction of inflammatory responses through downstream signaling pathways (Kocgozlu et al., 2009). Therefore, we examined whether LpDNA attenuates TLR2 and TLR4 expression. After pretreating RAW 264.7 cells with LpDNA and stimulating with PgLPS for 3 h, both TLR2 and TLR4 were significantly downregulated (Fig. 3A). TLRs recognize their ligands and trigger intracellular signal cascades to mount acute inflammatory responses (Wang and Ohura, 2002). The MAPK signaling pathways, including p38 kinase, ERK, and JNK, and the NF-κB signaling pathway are essential signaling pathways of inflammation that induce the secretion of proinflammatory cytokines (e.g., IL-1β) and subsequent inflammation (Kim et al., 2004). Hence, the activation of MAPKs and NF-κB was evaluated in RAW 264.7 cells pretreated with LpDNA in the presence of PgLPS. As shown in Fig. 3B, PgLPS alone increased the phosphorylation of p38 kinase, ERK, and JNK, while LpDNA pretreatment significantly reduced the phosphorylation of p38 kinases, ERK, and JNK. Moreover, an inhibitor of NF-κB, IκBα, which downregulates NF-κB activation (Wang et al., 2020), was degraded by PgLPS treatment alone, while LpDNA pretreatment restored IκBα in the presence of PgLPS (Fig. 3C). These results suggest that LpDNA-mediated TLR suppression downregulates the signaling cascades associated with PgLPS-induced inflammatory responses.

kosfa-43-5-938-g3
Fig. 3. Inhibitory effect of LpDNA on TLR/MAPK/NF-κB signaling pathways induced by Porphyromonas gingivalis LPS in RAW 264.7 cells. Cells were pretreated with LP gDNA (1 μg/mL) for 15 h and stimulated with P. gingivalis LPS (1 μg/mL) for an additional 3 h. The mRNA expression levels of TLR2 and TLR4 were measured by qRT-PCR (A). Cells were pretreated with LpDNA (1 μg/mL) for 15 h followed by stimulation with P. gingivalis LPS (1 μg/mL) for 30 min. The phosphorylation of p38 kinase, ERK, and JNK (B), and the degradation of IκBα (C) were determined by western blot analysis. The relative expression levels are represented as the means±SDs from three independent experiments. Statistical significance between groups was determined by ANOVA. p<0.05 was considered statistically significant. a–c Letters on each graph indicate significant difference (p<0.05). TLR, Toll-like receptor; PgLPS, Porphyromonas gingivalis lipopolysaccharide; LpDNA, Lactiplantibacillus plantarum DNA; ERK, extracellular signal-regulated kinase; JNK, c-Jun-N-terminal kinase; NT, non-treatment; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-κB; qRT-PCR, quantitative reverse transcription-polymerase chain reaction; ANOVA, analysis of variance.
Download Original Figure

Discussion

Many studies have demonstrated that probiotics, in particular lactobacilli, exert beneficial effects, including antimicrobial, immune modulating, and blood cholesterol reducing activities (Duary et al., 2012; Kõll-Klais et al., 2005; Kwak et al., 2014). Lactobacilli also play an important role in attenuating periodontal diseases. However, the beneficial effects of structural components derived from L. plantarum, such as genomic DNA, have not been widely documented. Although a previous study has reported that genomic DNA of Pediococcus acidilactici suppressed P. gingivalis LPS-induced inflammatory responses (Choi et al., 2023), we investigated the inhibition of genomic DNA derived from L. plantarum on P. gingivalis LPS-induced inflammatory responses.

Pro-inflammatory cytokine production is known to be elevated in gingival crevicular fluid (GCF) where it facilitates bone resorption, resulting in loss of connective tissue attachment (Graves, 2008). The chemokine MCP-1 plays a crucial role in periodontitis, and it can be induced and stimulated by cytokines like IL-1β (Ozaki et al., 1996). Indeed, higher expression of MCP-1 was observed in the GCF and serum of patients with chronic periodontitis compared to healthy subjects (Emingil et al., 2004; Pradeep et al., 2009). We found that LpDNA effectively inhibited these cytokines and chemokine by regulating the MAPK and NF-κB signaling pathways. Additionally, LpDNA efficiently reduced TLR2 and TLR4 expression, implying that modulation of TLR expression may lead to inhibition of the inflammatory responses induced by P. gingivalis LPS through the suppression of MAPKs and NF-κB activation. These findings are in accordance with those of previous studies, which have shown that TLR2-JNK is the key signaling pathway activated during P. gingivalis LPS-induced cytokine production (Zhang et al., 2008). Moreover, it has been confirmed that P. gingivalis LPS-induced IL-1β production was inhibited by anti-TLR4 antibody in human gingival fibroblasts (Wang and Ohura, 2002).

Many studies have emphasized the role of MAPK and NF-κB in the development of periodontitis, particularly the importance of p38 MAPK signaling in periodontitis progression (Li et al., 2012). Treatment strategies to block signal transduction pathways, including MAPK and NF-κB pathways, have been developed to target periodontal diseases (Chopra et al., 2010; McIntyre et al., 2003). However, these inhibitors are associated with side effects due to the lack of specificity in modulating the host response (de Souza et al., 2012). Hence, novel therapeutic approaches that overcome these limitations represent promising alternatives for periodontal treatment. In this study, LpDNA was demonstrated to exert anti-inflammatory activities against P. gingivalis. These results highlight the potential of LpDNA as a novel postbiotic compound against oral pathogens, which may be implemented to effectively enhance oral health.

Conflicts of Interest

The authors declare no potential conflicts of interest.

Acknowledgements

This work was carried out with the support of “Cooperative Research Program for Agriculture Science and Technology Development (Project No. PJ01669005)” Rural Development Administration, Korea.

Author Contributions

Conceptualization: Choi YH, Kim BS, Kang SS. Data curation: Choi YH, Kim BS. Formal analysis: Choi YH, Kim BS. Methodology: Choi YH, Kim BS. Software: Choi YH, Kim BS. Validation: Choi YH, Kim BS. Investigation: Choi YH, Kim BS. Writing - original draft: Choi YH. Writing - review & editing: Choi YH, Kim BS, Kang SS.

Ethics Approval

This article does not require IRB/IACUC approval because there are no human and animal participants.

References

1.

Borchers AT, Selmi C, Meyers FJ, Keen CL, Gershwin ME. 2009; Probiotics and immunity. J Gastroenterol. 44:26-46

2.

Choi YH, Kim BS, Kang SS. 2023; Inhibitory effect of genomic DNA extracted from Pediococcus acidilactici on Porphyromonas gingivalis lipopolysaccharide-induced inflammatory responses. Food Sci Anim Resour. 43:101-112

3.

Chopra P, Kulkarni O, Gupta S, Bajpai M, Kanoje V, Banerjee M, Bansal V, Visaga S, Chatterjee M, Chaira T, Shirumalla RK, Verma AK, Dastidar SG, Sharma G, Ray A. 2010; Pharmacological profile of AW-814141, a novel, potent, selective and orally active inhibitor of p38 MAP kinase. Int Immunopharmacol. 10:467-473

4.

Darveau RP, Pham TTT, Lemley K, Reife RA, Bainbridge BW, Coats SR, Howald WN, Way SS, Hajjar AM. 2004; Porphyromonas gingivalis lipopolysaccharide contains multiple lipid a species that functionally interact with both toll-like receptors 2 and 4. Infect Immun. 72:5041-5051

5.

de Souza JAC, Rossa C, Garlet GP, Nogueira AVB, Cirelli JA. 2012; Modulation of host cell signaling pathways as a therapeutic approach in periodontal disease. J Appl Oral Sci. 20:128-138

6.

Doron S, Snydman DR. 2015; Risk and safety of probiotics. Clin Infect Dis. 60:S129-S134

7.

Duary RK, Bhausaheb MA, Batish VK, Grover S. 2012; Anti-inflammatory and immunomodulatory efficacy of indigenous probiotic Lactobacillus plantarum Lp91 in colitis mouse model. Mol Biol Rep. 39:4765-4775

8.

Emingil G, Atilla G, Hüseyinov A. 2004; Gingival crevicular fluid monocyte chemoattractant protein-1 and RANTES levels in patients with generalized aggressive periodontitis. J Clin Periodontol. 31:829-834

9.

Gatej SM, Marino V, Bright R, Fitzsimmons TR, Gully N, Zilm P, Gibson RJ, Edwards S, Bartold PM. 2018; Probiotic Lactobacillus rhamnosus GG prevents alveolar bone loss in a mouse model of experimental periodontitis. J Clin Periodontol. 45:204-212

10.

Graves D. 2008; Cytokines that promote periodontal tissue destruction. J Periodontol. 79:1585-1591

11.

Izuddin WI, Humam AM, Loh TC, Foo HL, Samsudin AA. 2020; Dietary postbiotic Lactobacillus plantarum improves serum and ruminal antioxidant activity and upregulates hepatic antioxidant enzymes and ruminal barrier function in post-weaning lambs. Antioxidants. 9:250

12.

Kaji R, Kiyoshima-Shibata J, Nagaoka M, Nanno M, Shida K. 2010; Bacterial teichoic acids reverse predominant IL-12 production induced by certain Lactobacillus strains into predominant IL-10 production via TLR2-dependent ERK activation in macrophages. J Immunol. 184:3505-3513

13.

Kato H, Taguchi Y, Tominaga K, Umeda M, Tanaka A. 2014; Porphyromonas gingivalis LPS inhibits osteoblastic differentiation and promotes pro-inflammatory cytokine production in human periodontal ligament stem cells. Arch Oral Biol. 59:167-175

14.

Kim CH, Kim HG, Kim JY, Kim NR, Jung BJ, Jeong JH, Chung DK. 2012; Probiotic genomic DNA reduces the production of pro-inflammatory cytokine tumor necrosis factor-alpha. FEMS Microbiol Lett. 328:13-19

15.

Kim MY, Hyun IK, An S, Kim D, Kim KH, Kang SS. 2022; In vitro anti-inflammatory and antibiofilm activities of bacterial lysates from lactobacilli against oral pathogenic bacteria. Food Funct. 13:12755-12765

16.

Kim SH, Smith CJ, Van Eldik LJ. 2004; Importance of MAPK pathways for microglial pro-inflammatory cytokine IL-1β production. Neurobiol Aging. 25:431-439

17.

Kocgozlu L, Elkaim R, Tenenbaum H, Werner S. 2009; Variable cell responses to P. gingivalis lipopolysaccharide. J Dent Res. 88:741-745

18.

Kõll-Klais P, Mändar R, Leibur E, Marcotte H, Hammarström L, Mikelsaar M. 2005; Oral lactobacilli in chronic periodontitis and periodontal health: Species composition and antimicrobial activity. Oral Microbiol Immunol. 20:354-361

19.

Könönen E, Gursoy M, Gursoy UK. 2019; Periodontitis: A multifaceted disease of tooth-supporting tissues. J Clin Med. 8:1135

20.

Kwak SH, Cho YM, Noh GM, Om AS. 2014; Cancer preventive potential of kimchi lactic acid bacteria (Weissella cibaria, Lactobacillus plantarum). J Cancer Prev. 19:253-258

21.

Lammers KM, Brigidi P, Vitali B, Gionchetti P, Rizzello F, Caramelli E, Matteuzzi D, Campieri M. 2003; Immunomodulatory effects of probiotic bacteria DNA: IL-1 and IL-10 response in human peripheral blood mononuclear cells. FEMS Immunol Med Microbiol. 38:165-172

22.

Li Q, Valerio MS, Kirkwood KL. 2012; MAPK usage in periodontal disease progression. J Signal Transduct. 2012:308943

23.

McIntyre KW, Shuster DJ, Gillooly KM, Dambach DM, Pattoli MA, Lu P, Zhou XD, Qiu Y, Zusi FC, Burke JR. 2003; A highly selective inhibitor of IκB kinase, BMS-345541, blocks both joint inflammation and destruction in collagen-induced arthritis in mice. Arthritis Rheum. 48:2652-2659

24.

Moradi M, Kousheh SA, Almasi H, Alizadeh A, Guimarães JT, Yılmaz N, Lotfi A. 2020; Postbiotics produced by lactic acid bacteria: The next frontier in food safety. Compr Rev Food Sci Food Saf. 19:3390-3415

25.

Mysak J, Podzimek S, Sommerova P, Lyuya-Mi Y, Bartova J, Janatova T, Prochazkova J, Duskova J. 2014; Porphyromonas gingivalis: Major periodontopathic pathogen overview. J Immunol Res. 2014; :476068

26.

Ozaki K, Hanazawa S, Takeshita A, Chen Y, Watanabe A, Nishida K, Miyata Y, Kitano S. 1996; Interleukin-1β and tumor necrosis factor-α stimulate synergistically the expression of monocyte chemoattractant protein-1 in fibroblastic cells derived from human periodontal ligament. Oral Microbiol Immunol. 11:109-114

27.

Pradeep AR, Daisy H, Hadge P. 2009; Serum levels of monocyte chemoattractant protein-1 in periodontal health and disease. Cytokine. 47:77-81

28.

Tonetti MS, Jepsen S, Jin L, Otomo-Corgel J. 2017; Impact of the global burden of periodontal diseases on health, nutrition and wellbeing of mankind: A call for global action. J Clin Periodontol. 44:456-462

29.

Wang PL, Ohura K. 2002; Porphyromonas gingivalis lipopolysaccharide signaling in gingival fibroblasts: CD14 and Toll-like receptors. Crit Rev Oral Biol Med. 13:132-142

30.

Wang X, Peng H, Huang Y, Kong W, Cui Q, Du J, Jin H. 2020; Post-translational modifications of IκBα: The state of the art. Front Cell Dev Biol. 8:574706

31.

Zhang D, Chen L, Li S, Gu Z, Yan J. 2008; Lipopolysaccharide (LPS) of Porphyromonas gingivalis induces IL-1β, TNF-α and IL-6 production by THP-1 cells in a way different from that of Escherichia coli LPS. Innate Immun. 14:99-107

32.

Zhao J, Feng X, Zhang X, Le K. 2012; Effect of Porphyromonas gingivalis and Lactobacillus acidophilus on secretion of IL1B, IL6, and IL8 by gingival epithelial cells. Inflammation. 35:1330-1337

33.

Zhou Q, Desta T, Fenton M, Graves DT, Amar S. 2005; Cytokine profiling of macrophages exposed to Porphyromonas gingivalis, its lipopolysaccharide, or its FimA protein. Infect Immun. 73:935-943

34.

Żółkiewicz J, Marzec A, Ruszczyński M, Feleszko W. 2020; Postbiotics: A step beyond pre- and probiotics. Nutrients. 12:2189