Epigenetic regulation of inflammation in localized aggressive periodontitis

L M Shaddox, A F Mullersman, H Huang, S M Wallet, T Langaee, I Aukhil, L M Shaddox, A F Mullersman, H Huang, S M Wallet, T Langaee, I Aukhil

Abstract

Background: We have previously demonstrated a Toll-like receptor (TLR)-mediated hyper-responsive phenotype in our cohort of localized aggressive periodontitis (LAP) individuals. However, mechanisms related to this phenotype are still not clear in the literature. The objective of this cross-sectional study is to examine the role of epigenetic regulation, specifically DNA methylation status of genes in the TLR pathway in this cohort. Peripheral blood was collected from 20 LAP patients and 20 healthy unrelated controls. Whole blood was stimulated with 1 μl (100 ng/μl) of purified Escherichia coli lipopolysaccharide (LPS) for 24 h and cyto/chemokines in the supernatants analyzed by Luminex multiplex assays. Genomic DNA extracted from buffy coats prepared from a second tube of whole blood was used for DNA methylation analysis by pyrosequencing of seven TLR signaling genes (FADD, MAP3K7, MYD88, IL6R, PPARA, IRAK1BP1, RIPK2).

Results: Significant differences in the methylation status were observed at specific CpG positions in LAP patients compared to healthy controls and interestingly also between severe and moderate LAP. Specifically, subjects with moderate LAP presented hypermethylation of both the upregulating (MAP3K7, MYD88, IL6R, and RIPK2) and downregulating (FADD, IRAK, and PPARA) genes, while severe LAP presented hypomethylation of these genes. Further analysis on CpG sites with significant differences in methylation status correlates with an increased pro-inflammatory cytokine profile for LAP patients.

Conclusions: Our findings suggest that epigenetic modifications of genes in the TLR pathway may orchestrate the thresholds for balancing induction and prevention of tissue destruction during the course of disease, and thus differ significantly at different stages of the disease, where moderate LAP shows hypermethylation and severe LAP shows hypomethylation of several genes.

Trial registration: https://ichgcp.net/clinical-trials-registry/NCT01330719" title="See in ClinicalTrials.gov">NCT01330719.

Keywords: Aggressive periodontitis; Epigenetics; Inflammation; Leukotoxins; Toll-like receptors.

Conflict of interest statement

Ethics approval and consent to participate

As stated in the “Methods” section, informed consent was obtained and samples were collected under a protocol approved by the University of Florida Institutional Review Board (IRB 201400349).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
DNA methylation levels of pro-inflammatory genes in periodontitis vs control subjects. ANOVA with Tukey’s comparisons or Kruskal-Wallis with Dunn’s comparisons were carried out among healthy controls (HC), localized aggressive moderate (Mod), and severe (Sev) periodontitis. ***p < 0.001, **p < 0.01, *p < 0.05. Note that severe aggressive periodontitis displays lower methylation levels in several genes when compared to moderate periodontitis, while moderate disease shows higher methylation levels in general. TLR-2 signaling genes that are pro-inflammatory: RIPK2 receptor-interacting serine/threonine-protein kinase 2(c), MP3K7 mitogen-activated protein kinase kinase kinase 7(b), MYD88 myeloid differentiation primary response gene 88(a), IL6R interleukin 6 receptor(d)
Fig. 2
Fig. 2
DNA methylation levels of anti-inflammatory genes in periodontitis vs control subjects. ANOVA with Tukey’s comparisons or Kruskal-Wallis with Dunn’s comparisons were carried out among healthy controls (HC), localized aggressive moderate (Mod), and severe (Sev) periodontitis. ***p < 0.001, **p < 0.01, *p < 0.05. Note that severe aggressive periodontitis displays lower methylation levels in several genes when compared to moderate periodontitis, while moderate disease shows higher methylation levels in general. TLR-2 signaling genes that are anti-inflammatory: FADD Fas-associated protein with death domain(c), IRAKBP1 interleukin 1 receptor-associated kinase 1 binding protein 1(a), PPARA peroxisome proliferator-activated receptor alpha(b)

References

    1. Eke PI, Dye BA, Wei L, Thornton-Evans GO, Genco RJ, GDRP Cdc Periodontal Disease Surveillance workgroup: James Beck. Prevalence of periodontitis in adults in the United States: 2009 and 2010. J Dent Res. 2012;91:914–20.
    1. Trindade F, Oppenheim FG, Helmerhorst EJ, Amado F, Gomes PS, Vitorino R. Uncovering the molecular networks in periodontitis. Proteomics Clin Appl. 2014;8:748–61. doi: 10.1002/prca.201400028.
    1. Shaddox L, Wiedey J, Bimstein E, Magnuson I, Clare-Salzler M, Aukhil I, et al. Hyper-responsive phenotype in localized aggressive periodontitis. J Dent Res. 2010;89:143–8. doi: 10.1177/0022034509353397.
    1. van der Velden U, Abbas F, Armand S, de Graaff J, Timmerman MF, van der Weijden GA, et al. The effect of sibling relationship on the periodontal condition. J Clin Periodontol. 1993;20:683–90. doi: 10.1111/j.1600-051X.1993.tb00716.x.
    1. Diehl SR, Wu T, Michalowicz BS, Brooks CN, Califano JV, Burmeister JA, et al. Quantitative measures of aggressive periodontitis show substantial heritability and consistency with traditional diagnoses. J Periodontol. 2005;76:279–88. doi: 10.1902/jop.2005.76.2.279.
    1. Li Y, Xu L, Hasturk H, Kantarci A, DePalma SR, Van Dyke TE. Localized aggressive periodontitis is linked to human chromosome 1q25. Hum Genet. 2004;114:291–7. doi: 10.1007/s00439-003-1065-7.
    1. Marazita ML, Burmeister JA, Gunsolley JC, Koertge TE, Lake K, Schenkein HA. Evidence for autosomal dominant inheritance and race-specific heterogeneity in early-onset periodontitis. J Periodontol. 1994;65:623–30. doi: 10.1902/jop.1994.65.6.623.
    1. Vieira AR, Albandar JM. Role of genetic factors in the pathogenesis of aggressive periodontitis. Periodontol. 2000;2014(65):92–106.
    1. Lindroth AM, Park YJ. Epigenetic biomarkers: a step forward for understanding periodontitis. J Periodontal Implant Sci. 2013;43:111–20. doi: 10.5051/jpis.2013.43.3.111.
    1. Zhang S, Barros SP, Moretti AJ, Yu N, Zhou J, Preisser JS, et al. Epigenetic regulation of TNFA expression in periodontal disease. J Periodontol. 2013;84:1606–16.
    1. Barros SP, Offenbacher S. Modifiable risk factors in periodontal disease: epigenetic regulation of gene expression in the inflammatory response. Periodontol. 2000;2014(64):95–110.
    1. Stenvinkel P, Karimi M, Johansson S, Axelsson J, Suliman M, Lindholm B, et al. Impact of inflammation on epigenetic DNA methylation—a novel risk factor for cardiovascular disease? J Intern Med. 2007;261:488–99. doi: 10.1111/j.1365-2796.2007.01777.x.
    1. Zhang S, Crivello A, Offenbacher S, Moretti A, Paquette DW, Barros SP. Interferon-gamma promoter hypomethylation and increased expression in chronic periodontitis. J Clin Periodontol. 2010;37:953–61. doi: 10.1111/j.1600-051X.2010.01616.x.
    1. Baptista NB, Portinho D, Casarin RC, Vale HF, Casati MZ, De Souza AP, et al. DNA methylation levels of SOCS1 and LINE-1 in oral epithelial cells from aggressive periodontitis patients. Arch Oral Biol. 2014;59:670–8. doi: 10.1016/j.archoralbio.2014.03.015.
    1. Larsson L, Castilho RM, Giannobile WV. Epigenetics and its role in periodontal diseases: a state-of-the-art review. J Periodontol. 2015;86:556–68. doi: 10.1902/jop.2014.140559.
    1. Ajibade AA, Wang HY, Wang RF. Cell type-specific function of TAK1 in innate immune signaling. Trends Immunol. 2013;34:307–16. doi: 10.1016/j.it.2013.03.007.
    1. Magalhaes JG, Lee J, Geddes K, Rubino S, Philpott DJ, Girardin SE. Essential role of Rip2 in the modulation of innate and adaptive immunity triggered by Nod1 and Nod2 ligands. Eur J Immunol. 2011;41:1445–55. doi: 10.1002/eji.201040827.
    1. Tigno-Aranjuez JT, Asara JM, Abbott DW. Inhibition of RIP2’s tyrosine kinase activity limits NOD2-driven cytokine responses. Genes Dev. 2010;24:2666–77. doi: 10.1101/gad.1964410.
    1. Deguine J, Barton GM. MyD88: a central player in innate immune signaling. F1000Prime Rep. 2014;6:97.
    1. Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16:448–57. doi: 10.1038/ni.3153.
    1. Blander JM. A long-awaited merger of the pathways mediating host defence and programmed cell death. Nat Rev Immunol. 2014;14:601–18. doi: 10.1038/nri3720.
    1. Kono K, Kamijo Y, Hora K, Takahashi K, Higuchi M, Kiyosawa K, et al. PPAR{alpha} attenuates the proinflammatory response in activated mesangial cells. Am J Physiol Renal Physiol. 2009;296:F328–36. doi: 10.1152/ajprenal.00484.2007.
    1. Conner JR, Smirnova II, Moseman AP, Poltorak A. IRAK1BP1 inhibits inflammation by promoting nuclear translocation of NF-kappaB p50. Proc Natl Acad Sci U S A. 2010;107:11477–82. doi: 10.1073/pnas.1006894107.
    1. Zhang X, Fu R, Yu J, Wu X. DNA demethylation: where genetics meets epigenetics. Curr Pharm Des. 2014;20:1625–31. doi: 10.2174/13816128113199990546.
    1. Schenkein HA, Barbour SE, Tew JG. Cytokines and inflammatory factors regulating immunoglobulin production in aggressive periodontitis. Periodontol. 2000;2007(45):113–27.
    1. Lu H, Wang M, Gunsolley JC, Schenkein HA, Tew JG. Serum immunoglobulin G subclass concentrations in periodontally healthy and diseased individuals. Infect Immun. 1994;62:1677–82.
    1. Kawano Y, Noma T, Yata J. Regulation of human IgG subclass production by cytokines. IFN-gamma and IL-6 act antagonistically in the induction of human IgG1 but additively in the induction of IgG2. J Immunol. 1994;153:4948–58.
    1. Ishihara Y, Zhang JB, Fakher M, Best AM, Schenkein HA, Barbour SE, et al. Non-redundant roles for interleukin-1 alpha and interleukin-1 beta in regulating human IgG2. J Periodontol. 2001;72:1332–9. doi: 10.1902/jop.2001.72.10.1332.
    1. Saraiva L, Rebeis ES, Martins Ede S, Sekiguchi RT, Ando-Suguimoto ES, Mafra CE, et al. IgG sera levels against a subset of periodontopathogens and severity of disease in aggressive periodontitis patients: a cross-sectional study of selected pocket sites. J Clin Periodontol. 2014;41:943–51. doi: 10.1111/jcpe.12296.
    1. Hendler A. TK Mulli, FJ Hughes, D Perrett, M Bombardieri, Y Houri-Haddad, et al. Involvement of autoimmunity in the pathogenesis of aggressive periodontitis. J Dent Res. 2010;89:1389–94. doi: 10.1177/0022034510381903.
    1. de Faria Amormino SA, Arao TC, Saraiva AM, Gomez RS, Dutra WO, da Costa JE, et al. Hypermethylation and low transcription of TLR2 gene in chronic periodontitis. Hum Immunol. 2013;74:1231–6. doi: 10.1016/j.humimm.2013.04.037.
    1. Schulz S, Immel UD, Just L, Schaller HG, Glaser C, Reichert S. Epigenetic characteristics in inflammatory candidate genes in aggressive periodontitis. Hum Immunol. 2016;77:71–5. doi: 10.1016/j.humimm.2015.10.007.
    1. Stefani FA, Viana MB, Dupim AC, Brito JA, Gomez RS, da Costa JE, et al. Expression, polymorphism and methylation pattern of interleukin-6 in periodontal tissues. Immunobiology. 2013;218:1012–7. doi: 10.1016/j.imbio.2012.12.001.
    1. Niu Y, DesMarais TL, Tong Z, Yao Y, Costa M. Oxidative stress alters global histone modification and DNA methylation. Free Radic Biol Med. 2015;82:22–8. doi: 10.1016/j.freeradbiomed.2015.01.028.
    1. Soma T, Kaganoi J, Kawabe A, Kondo K, Imamura M, Shimada Y. Nicotine induces the fragile histidine triad methylation in human esophageal squamous epithelial cells. Int J Cancer. 2006;119:1023–7. doi: 10.1002/ijc.21948.
    1. Bobetsis YA, Barros SP, Lin DM, Weidman JR, Dolinoy DC, Jirtle RL, et al. Bacterial infection promotes DNA hypermethylation. J Dent Res. 2007;86:169–74. doi: 10.1177/154405910708600212.
    1. Ross SA, Poirier L. Proceedings of the Trans-HHS workshop: diet, DNA methylation processes and health. J Nutr. 2002;132:2329S–32S.
    1. Strickland FM, Hewagama A, Wu A, Sawalha AH, Delaney C, Hoeltzel MF, et al. Diet influences expression of autoimmune-associated genes and disease severity by epigenetic mechanisms in a transgenic mouse model of lupus. Arthritis Rheum. 2013;65:1872–81. doi: 10.1002/art.37967.
    1. Pan W, Zhu S, Yuan M, Cui H, Wang L, Luo X, et al. MicroRNA-21 and microRNA-148a contribute to DNA hypomethylation in lupus CD4+ T cells by directly and indirectly targeting DNA methyltransferase 1. J Immunol. 2010;184:6773–81. doi: 10.4049/jimmunol.0904060.
    1. Houseman EA, Kim S, Kelsey KT, Wiencke JK. DNA methylation in whole blood: uses and challenges. Curr Environ Health Rep. 2015;2:145–54. doi: 10.1007/s40572-015-0050-3.
    1. Honda T. G Tamura, T Waki, S Kawata, M Terashima, S Nishizuka, et al. Demethylation of MAGE promoters during gastric cancer progression. Br J Cancer. 2004;90:838–43. doi: 10.1038/sj.bjc.6601600.
    1. Armitage GC. Development of a classification system for periodontal diseases and conditions. Ann Periodontol. 1999;4:1–6. doi: 10.1902/annals.1999.4.1.1.

Source: PubMed

3
購読する