Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Periodontitis: a polymicrobial disruption of host homeostasis

Key Points

  • Periodontitis is intimately associated with a characterized polymicrobial dental-plaque community. However, the species and mechanisms that result in disease remain unclear.

  • The main protective mechanism of the periodontium is the orchestrated expression of select innate host defence mediators. These mediators reduce the microbial load on the epithelial-cell surface by both neutralization and bacterial killing.

  • The main mechanism of bone loss in periodontitis seems to be mediated by the host response to a unique microbial consortium. However, most of the bacteria found in the dental-plaque biofilm are capable of initiating inflammatory cytokine responses, rendering identification of key indicators of disease difficult.

  • Examination of gingival crevicular fluid from clinically healthy sites revealed that this fluid contains cytokines that may induce inflammation. However, the levels of these cytokines in healthy sites are lower than levels in diseased sites, indicating that a disruption of host homeostasis contributes to disease.

  • Extensive analysis of dental-plaque bacteria associated with disease has revealed three bacterial species that display strong associations both with each other and diseased sites. Evidence indicates that these bacteria, designated the 'red-complex' species, may interfere with the protective barrier of the host innate defence response.

  • Red-complex bacteria maybe key species in the pathogenic dental-plaque biofilm. By modulating the innate host defence barrier, they facilitate the growth of other members of the dental-plaque biofilm. The increase in the number and types of bacterial species that occupy the gingival crevice present multiple opportunities to disrupt host homeostasis programmes.

  • Physical removal of the dental-plaque biofilm remains the most effective treatment. However, vaccine strategies targeting red-complex bacteria have also shown efficacy in animal models of disease. The ability to attenuate bone loss by the inhibition of a single bacterial species is consistent with the key species concept.

  • A novel pro-resolving mediator shows promise as an effective periodontitis treatment in pre-clinical studies. A compound that restores host homeostasis is consistent with the theory that an important component of the disease is disruption of host homeostasis programmes.

Abstract

Periodontitis, or gum disease, affects millions of people each year. Although it is associated with a defined microbial composition found on the surface of the tooth and tooth root, the contribution of bacteria to disease progression is poorly understood. Commensal bacteria probably induce a protective response that prevents the host from developing disease. However, several bacterial species found in plaque (the 'red-complex' bacteria: Porphyromonas gingivalis, Tannerella forsythia and Treponema denticola) use various mechanisms to interfere with host defence mechanisms. Furthermore, disease may result from 'community-based' attack on the host. Here, I describe the interaction of the host immune system with the oral bacteria in healthy states and in diseased states.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The effects of periodontitis.
Figure 2: Transit of neutrophils through periodontal tissue.
Figure 3: Microbial alteration of bone homeostasis leads to localized bone loss.
Figure 4: Porphyromonas gingivalis, a member of the red-complex bacteria, inhibits innate host defence functions in gingival epithelium.

Similar content being viewed by others

References

  1. Socransky, S. S. & Haffajee, A. D. Evidence of bacterial etiology: a historical perspective. Periodontol. 2000 5, 7–25 (1994).

    Article  CAS  PubMed  Google Scholar 

  2. Socransky, S. S., Haffajee, A. D., Cugini, M. A., Smith, C. & Kent, R. L. Jr. Microbial complexes in subgingival plaque. J. Clin. Periodontol. 25, 134–144 (1998). This landmark work describes the different bacterial complexes associated with periodontal disease. It greatly influenced the field as other investigators sought bacterial virulence properties that could be associated with red-complex bacteria.

    Article  CAS  PubMed  Google Scholar 

  3. Ximenez-Fyvie, L. A., Haffajee, A. D. & Socransky, S. S. Comparison of the microbiota of supra- and subgingival plaque in health and periodontitis. J. Clin. Periodontol. 27, 648–657 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Darveau, R. P., Tanner, A. & Page, R. C. The microbial challenge in periodontitis. Periodontol. 2000 14, 12–32 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Tanner, A., Kent, R., Maiden, M. F. & Taubman, M. A. Clinical, microbiological and immunological profile of healthy, gingivitis and putative active periodontal subjects. J. Periodont. Res. 31, 195–204 (1996).

    Article  CAS  Google Scholar 

  6. Huyghe, A. et al. Novel microarray design strategy to study complex bacterial communities. Appl. Environ. Microbiol. 74, 1876–1885 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kroes, I., Lepp, P. W. & Relman, D. A. Bacterial diversity within the human subgingival crevice. Proc. Natl Acad. Sci. USA 96, 14547–14552 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Berglundh, T., Liljenberg, B. & Lindhe, J. Some effects of periodontal therapy on local and systemic immunological parameters. J. Clin. Periodontol. 26, 91–98 (1999).

    Article  CAS  PubMed  Google Scholar 

  9. Yoshinari, N. et al. Effects of scaling and root planing on the amounts of interleukin-1 and interleukin-1 receptor antagonist and the mRNA expression of interleukin-1β in gingival crevicular fluid and gingival tissues. J. Periodont. Res. 39, 158–167 (2004).

    Article  CAS  Google Scholar 

  10. Roberts, F. A., Hockett, R. D. Jr, Bucy, R. P. & Michalek, S. M. Quantitative assessment of inflammatory cytokine gene expression in chronic adult periodontitis. Oral Microbiol. Immunol. 12, 336–344 (1997).

    Article  CAS  PubMed  Google Scholar 

  11. Champagne, C. M. et al. Potential for gingival crevice fluid measures as predictors of risk for periodontal diseases. Periodontol. 2000 31, 167–180 (2003).

    Article  PubMed  Google Scholar 

  12. Giannobile, W. V. Crevicular fluid biomarkers of oral bone loss. Curr. Opin. Periodontol. 4, 11–20 (1997).

    CAS  PubMed  Google Scholar 

  13. Kamma, J., Mombelli, A., Tsinidou, K., Vasdekis, V. & Giannopoulou, C. Cytokines in gingival crevicular fluid of adolescents and young adults. Oral Microbiol. Immunol. 24, 7–10 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Thunell, D. H. et al. A multiplex immunoassay demonstrates reductions in gingival crevicular fluid cytokines following initial periodontal therapy. J. Periodont. Res. 45, 148–152 (2009).

    Article  Google Scholar 

  15. Page, R. C. & Kornman, K. S. The pathogenesis of human periodontitis: an introduction. Periodontol. 2000 14, 9–11 (1997).

    Article  CAS  PubMed  Google Scholar 

  16. Van Dyke, T. E. The management of inflammation in periodontal disease. J. Periodontol. 79, 1601–1608 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. O'Neill, L. A. The interleukin-1 receptor/Toll-like receptor superfamily: 10 years of progress. Immunol. Rev. 226, 10–18 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Beutler, B., Hoebe, K., Du, X. & Ulevitch, R. J. How we detect microbes and respond to them: the Toll-like receptors and their transducers. J. Leukoc. Biol. 74, 479–485 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Kolenbrander, P. E. et al. Bacterial interactions and successions during plaque development. Periodontol. 2000 42, 47–79 (2006). This manuscript describes the ecological succession that occurs in dental-plaque formation and details the known adhesive interactions that occur among different oral species and that facilitate their highly ordered structure.

    Article  PubMed  Google Scholar 

  20. Frank, D. N. et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl Acad. Sci. USA 104, 13780–13785 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Oakley, B. B., Fiedler, T. L., Marrazzo, J. M. & Fredricks, D. N. Diversity of human vaginal bacterial communities and associations with clinically defined bacterial vaginosis. Appl. Environ. Microbiol. 74, 4898–4909 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, L. et al. Inflammation and intestinal metaplasia of the distal esophagus are associated with alterations in the microbiome. Gastroenterology 137, 588–597 (2009).

    Article  PubMed  Google Scholar 

  23. Marsh, P. D. Microbial ecology of dental plaque and its significance in health and disease. Adv. Dent. Res. 8, 263–271 (1994). This article describes the microbial shift that occurs in periodontitis.

    Article  CAS  PubMed  Google Scholar 

  24. Paster, B. J. et al. Bacterial diversity in human subgingival plaque. J. Bacteriol. 183, 3770–3783 (2001). One of the most extensive analyses of dental plaque, carried out by 16s RNA identification. This work notably increased the number of identified species in dental plaque by identifying non-cultivatable species.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kumar, P. S., Griffen, A. L., Moeschberger, M. L. & Leys, E. J. Identification of candidate periodontal pathogens and beneficial species by quantitative 16S clonal analysis. J. Clin. Microbiol. 43, 3944–3955 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Brinig, M. M., Lepp, P. W., Ouverney, C. C., Armitage, G. C. & Relman, D. A. Prevalence of bacteria of division TM7 in human subgingival plaque and their association with disease. Appl. Environ. Microbiol. 69, 1687–1694 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lepp, P. W. et al. Methanogenic Archaea and human periodontal disease. Proc. Natl Acad. Sci. USA 101, 6176–6181 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Vianna, M. E., Holtgraewe, S., Seyfarth, I., Conrads, G. & Horz, H. P. Quantitative analysis of three hydrogenotrophic microbial groups, methanogenic archaea, sulfate-reducing bacteria, and acetogenic bacteria, within plaque biofilms associated with human periodontal disease. J. Bacteriol. 190, 3779–3785 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kumar, P. S. et al. Changes in periodontal health status are associated with bacterial community shifts as assessed by quantitative 16S cloning and sequencing. J. Clin. Microbiol. 44, 3665–3673 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bosshardt, D. D. & Lang, N. P. The junctional epithelium: from health to disease. J. Dent. Res. 84, 9–20 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Duerkop, B. A., Vaishnava, S. & Hooper, L. V. Immune responses to the microbiota at the intestinal mucosal surface. Immunity 31, 368–376 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Moughal, N. A., Adonogianaki, E., Thornhill, M. H. & Kinane, D. F. Endothelial cell leukocyte adhesion molecule-1 (ELAM-1) and intercellular adhesion molecule-1 (ICAM-1) expression in gingival tissue during health and experimentally-induced gingivitis. J. Periodont. Res. 27, 623–630 (1992).

    Article  CAS  Google Scholar 

  33. Nylander, K., Danielsen, B., Fejerskov, O. & Dabelsteen, E. Expression of the endothelial leukocyte adhesion molecule-1 (ELAM-1) on endothelial cells in experimental gingivitis in humans. J. Periodontol. 64, 355–357 (1993).

    Article  CAS  PubMed  Google Scholar 

  34. Gemmell, E., Walsh, L. J., Savage, N. W. & Seymore, G. J. Adhesion molecule expression in chronic inflammatory periodontal disease tissue. J. Periodont. Res. 29, 46–53 (1994).

    Article  CAS  Google Scholar 

  35. Tonetti, M. S. Molecular factors associated with compartmentalization of gingival immune responses and transepithelial neutrophil migration. J. Periodont. Res. 32, 104–109 (1997).

    Article  CAS  Google Scholar 

  36. Tonetti, M. S. et al. Localized expression of mRNA for phagocyte-specific hemotactic cytokines in human periodontal infections. Infect. Immun. 62, 4005–4014 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Tonetti, M. S., Imboden, M. A. & Lang, N. P. Neutrophil migration into the gingival sulcus is associated with transepithelial gradients of interleukin-8 and ICAM-1. J. Periodontol. 69, 1139–1147 (1998). This study provides a mechanism for the transmigration of neutrophils from the vasculature to the gingival crevice. This work initiated further studies aiming to better understand the orchestrated expression of select innate defence mediators in clinically healthy periodontal tissue.

    Article  CAS  PubMed  Google Scholar 

  38. Schiott, C. R. & Loe, H. The origin and variation in number of leukocytes in the human saliva. J. Periodont. Res. 5, 36–41 (1970).

    Article  CAS  Google Scholar 

  39. Hart, T. C., Shapira, L. & Van Dyke, T. E. Neutrophil defects as risk factors for periodontal diseases. J. Periodontol. 65, 521–529 (1994).

    Article  CAS  PubMed  Google Scholar 

  40. Carrassi, A., Abati, S., Santarelli, G. & Vogel, G. Periodontitis in a patient with chronic neutropenia. J. Periodontol. 60, 352–357 (1989).

    Article  CAS  PubMed  Google Scholar 

  41. Page, R. C., Beatty, P. & Waldrop, T. C. Molecular basis for the functional abnormality in neutrophils from patients with generalized prepubertal periodontitis. J. Periodont. Res. 22, 182–183 (1987).

    Article  CAS  Google Scholar 

  42. Waldrop, T. C., Anderson, D. C., Hallmon, W. W., Schmalstieg, F. C. & Jacobs, R. L. Periodontal manifestations of the heritable Mac-1, LFA-1, deficiency syndrome. Clinical, histopathologic and molecular characteristics. J. Periodontol. 58, 400–416 (1987).

    Article  CAS  PubMed  Google Scholar 

  43. Attström, R. & Schroeder, H. E. Effect of experimental neutropenia on initial gingivitis in dogs. Scand. J. Dent. Res. 87, 7–23 (1979).

    PubMed  Google Scholar 

  44. Sallay, K., Listgarten, M., Sanavi, F., Ring, I. & Nowotny, A. Bacterial invasion of oral tissues of immunosuppressed rats. Infect. Immun. 43, 1091–1093 (1984).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Hemmerle, J. & Frank, R. M. Bacterial invasion of periodontal tissues after experimental immunosuppression in rats. J. Biol. Buccale 19, 271–282 (1991).

    CAS  PubMed  Google Scholar 

  46. Yoshinari, N., Kameyama, Y., Aoyama, Y., Nishiyama, H. & Noguchi, T. Effect of long-term methotrexate-induced neutropenia on experimental periodontal lesion in rats. J. Periodont. Res. 29, 393–400 (1994).

    Article  CAS  Google Scholar 

  47. Lu, Q., Jin, L., Darveau, R. P. & Samaranayake, L. P. Expression of human β-defensins-1 and -2 peptides in unresolved chronic periodontitis. J. Periodont. Res. 39, 221–227 (2004).

    Article  CAS  Google Scholar 

  48. Lu, Q., Samaranayake, L. P., Darveau, R. P. & Jin, L. Expression of human β-defensin-3 in gingival epithelia. J. Periodont. Res. 40, 474–481 (2005).

    Article  CAS  Google Scholar 

  49. Jin, L. & Darveau, R. P. Soluble CD14 levels in gingival crevicular fluid of subjects with untreated adult periodontitis. J. Periodontol. 72, 634–640 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Jin, L., Ren, L., Leung, W. K. & Darveau, R. P. The in vivo expression of membrane-bound CD14 in periodontal health and disease. J. Periodontol. 75, 578–585 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Ren, L., Jin, L. & Leung, W. K. Local expression of lipopolysaccharide-binding protein in human gingival tissues. J. Periodont. Res. 39, 242–248 (2004).

    Article  CAS  Google Scholar 

  52. Mahanonda, R. & Pichyangkul, S. Toll-like receptors and their role in periodontal health and disease. Periodontol. 2000 43, 41–55 (2007).

    Article  PubMed  Google Scholar 

  53. Ren, L., Leung, W. K., Darveau, R. P. & Jin, L. The expression profile of lipopolysaccharide-binding protein, membrane-bound CD14, and Toll-like receptors 2 and 4 in chronic periodontitis. J. Periodontol. 76, 1950–1959 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Sugawara, Y. et al. Toll-like receptors, NOD1, and NOD2 in oral epithelial cells. J. Dent. Res. 85, 524–529 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Otte, J. M., Cario, E. & Podolsky, D. K. Mechanisms of cross hyporesponsiveness to Toll-like receptor bacterial ligands in intestinal epithelial cells. Gastroenterology 126, 1054–1070 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Eskan, M. A., Hajishengallis, G. & Kinane, D. F. Differential activation of human gingival epithelial cells and monocytes by Porphyromonas gingivalis fimbriae. Infect. Immun. 75, 892–898 (2007).

    Article  CAS  PubMed  Google Scholar 

  57. Dixon, D. R., Bainbridge, B. W. & Darveau, R. P. Modulation of the innate immune response within the periodontium. Periodontol. 2000 35, 53–74 (2004).

    Article  PubMed  Google Scholar 

  58. Darveau, R. P., Belton, C. M., Reife, R. A. & Lamont, R. J. Local chemokine paralysis, a novel pathogenic mechanism for Porphyromonas gingivalis. Infect. Immun. 66, 1660–1665 (1998). The first observation that P. gingivalis inhibits IL-8 secretion from gingival epithelial cells, and the proposal that P. gingivalis could induce a local chemokine paralysis as a mechanism of virulence.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Hasegawa, Y. et al. Gingival epithelial cell transcriptional responses to commensal and opportunistic oral microbial species. Infect. Immun. 75, 2540–2547 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Huang, G. T., Zhang, H. B., Dang, H. N. & Haake, S. K. Differential regulation of cytokine genes in gingival epithelial cells challenged by Fusobacterium nucleatum and Porphyromonas gingivalis. Microbiol. Pathog. 37, 303–312 (2004).

    Article  CAS  Google Scholar 

  61. Chung, W. O., Dommisch, H., Yin, L. & Dale, B. A. Expression of defensins in gingiva and their role in periodontal health and disease. Curr. Pharm. Des. 13, 3073–3083 (2007).

    Article  CAS  PubMed  Google Scholar 

  62. Krisanaprakornkit, S. et al. Inducible expression of human β-defensin 2 by Fusobacterium nucleatum in oral epithelial cells: multiple signaling pathways and role of commensal bacteria in innate immunity and the epithelial barrier. Infect. Immun. 68, 2907–2915 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Crawford, J. M., Taubman, M. A. & Smith, D. J. The natural history of periodontal bone loss in germfree and gnotobiotic rats infected with periodontopathic microorganisms. J. Periodont. Res. 13, 316–325 (1978).

    Article  CAS  Google Scholar 

  64. Dixon, D. R., Reife, R. A., Cebra, J. J. & Darveau, R. P. Commensal bacteria influence innate status within gingival tissues: a pilot study. J. Periodontol. 75, 1486–1492 (2004).

    Article  PubMed  Google Scholar 

  65. Stappenbeck, T. S., Hooper, L. V. & Gordon, J. I. Developmental regulation of intestinal angiogenesis by indigenous microbes via Paneth cells. Proc. Natl Acad. Sci. USA 99, 15451–15455 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Xu, J. & Gordon, J. I. Inaugural Article: Honor thy symbionts. Proc. Natl Acad. Sci. USA 100, 10452–10459 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Umesaki, Y. & Setoyama, H. Structure of the intestinal flora responsible for development of the gut immune system in a rodent model. Microbes Infect. 2, 1343–1351 (2000).

    Article  CAS  PubMed  Google Scholar 

  68. Gordon, H. A. & Pesti, L. The gnotobiotic animal as a tool in the study of host microbial relationships. Bacteriol. Rev. 35, 390–429 (1971).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Duncan, H. E. & Edberg, S. C. Host-microbe interaction in the gastrointestinal tract. Crit. Rev. Microbiol. 21, 85–100 (1995).

    Article  CAS  PubMed  Google Scholar 

  70. Falk, P. G., Hooper, L. V., Midtvedt, T. & Gordon, J. I. Creating and maintaining the gastrointestinal ecosystem: what we know and need to know from gnotobiology. Microbiol. Mol. Biol. Rev. 62, 1157–1170 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Macpherson, A. J. & Harris, N. L. Interactions between commensal intestinal bacteria and the immune system. Nature Rev. Immunol. 4, 478–485 (2004).

    Article  CAS  Google Scholar 

  72. Cebra, J. J. Influences of microbiota on intestinal immune system development. Am. J. Clin. Nutr. 69, S1046–S1051 (1999).

    Article  Google Scholar 

  73. Chadwick, V. S. & Anderson, R. P. in Inflammatory Bowel Disease (eds MacDermott, R. P. & Stenson, W. F.) 241–258 (Elsevier Science, Amsterdam, 1992).

    Google Scholar 

  74. Hooper, L. V. et al. Molecular analysis of commensal host-microbial relationships in the intestine. Science 291, 881–884 (2001).

    Article  CAS  PubMed  Google Scholar 

  75. Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., Edberg, S. & Medzhitov, R. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell 118, 229–241 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Bartold, P. M., Walsh, L. J. & Narayanan, A. S. Molecular and cell biology of the gingiva. Periodontol. 2000 24, 28–55 (2000).

    Article  CAS  PubMed  Google Scholar 

  77. Mussig, E., Tomakidi, P. & Steinberg, T. Molecules contributing to the maintenance of periodontal tissues. Their possible association with orthodontic tooth movement. J. Orofac. Orthop. 66, 422–433 (2005).

    Article  PubMed  Google Scholar 

  78. Gorska, R. et al. Relationship between clinical parameters and cytokine profiles in inflamed gingival tissue and serum samples from patients with chronic periodontitis. J. Clin. Periodontol. 30, 1046–1052 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Page, R. C., Offenbacher, S., Schroeder, H. E., Seymour, G. J. & Kornman, K. S. Advances in the pathogenesis of periodontitis: summary of developments, clinical implications and future directions. Periodontol. 2000 14, 216–248 (1997). A summary of many of the current theories about periodontitis and the balance between periodontal health and disease, and an excellent review of how clinical markers have influenced our thinking about disease aetiology.

    Article  CAS  PubMed  Google Scholar 

  80. Cochran, D. L. Inflammation and bone loss in periodontal disease. J. Periodontol. 79, 1569–1576 (2008).

    Article  CAS  PubMed  Google Scholar 

  81. Nagasawa, T. et al. Roles of receptor activator of nuclear factor-κB ligand (RANKL) and osteoprotegerin in periodontal health and disease. Periodontol. 2000 43, 65–84 (2007).

    Article  PubMed  Google Scholar 

  82. Boyle, W. J., Simonet, W. S. & Lacey, D. L. Osteoclast differentiation and activation. Nature 423, 337–342 (2003).

    Article  CAS  PubMed  Google Scholar 

  83. Assuma, R., Oates, T., Cochran, D., Amar, S. & Graves, D. T. IL-1 and TNF antagonists inhibit the inflammatory response and bone loss in experimental periodontitis. J. Immunol. 160, 403–409 (1998).

    CAS  PubMed  Google Scholar 

  84. Baker, P. J. et al. CD4+ T cells and the proinflammatory cytokines gamma interferon and interleukin-6 contribute to alveolar bone loss in mice. Infect. Immun. 67, 2804–2809 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Delima, A. J. et al. Soluble antagonists to interleukin-1 (IL-1) and tumor necrosis factor (TNF) inhibits loss of tissue attachment in experimental periodontitis. J. Clin. Periodontol. 28, 233–240 (2001).

    Article  CAS  PubMed  Google Scholar 

  86. Graves, D. T. et al. Interleukin-1 and tumor necrosis factor antagonists inhibit the progression of inflammatory cell infiltration toward alveolar bone in experimental periodontitis. J. Periodontol. 69, 1419–1425 (1998).

    Article  CAS  PubMed  Google Scholar 

  87. Garlet, G. P. et al. The dual role of p55 tumour necrosis factor-α receptor in Actinobacillus actinomycetemcomitans-induced experimental periodontitis: host protection and tissue destruction. Clin. Exp. Immunol. 147, 128–138 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Niederman, R. et al. Infection-mediated early-onset periodontal disease in P/E-selectin-deficient mice. J. Clin. Periodontol. 28, 569–575 (2001).

    Article  CAS  PubMed  Google Scholar 

  89. Al-Rasheed, A., Scheerens, H., Rennick, D. M., Fletcher, H. M. & Tatakis, D. N. Accelerated alveolar bone loss in mice lacking interleukin-10. J. Dent. Res. 82, 632–635 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Dayan, S., Stashenko, P., Niederman, R. & Kupper, T. S. Oral epithelial overexpression of IL-1α causes periodontal disease. J. Dent. Res. 83, 786–790 (2004).

    Article  CAS  PubMed  Google Scholar 

  91. Janeway, C. J. The immune system evolved to discriminate infectious nonself from noninfectious self. Immunol. Today 13, 11–16 (1992).

    Article  CAS  PubMed  Google Scholar 

  92. Kawai, T. & Akira, S. Pathogen recognition with Toll-like receptors. Curr. Opin. Immunol. 17, 338–344 (2005).

    Article  CAS  PubMed  Google Scholar 

  93. Yoshioka, H., Yoshimura, A., Kaneko, T., Golenbock, D. T. & Hara, Y. Analysis of the activity to induce toll-like receptor (TLR)2- and TLR4-mediated stimulation of supragingival plaque. J. Periodontol. 79, 920–928 (2008).

    Article  CAS  PubMed  Google Scholar 

  94. Burns, E., Bachrach, G., Shapira, L. & Nussbaum, G. Cutting Edge: TLR2 is required for the innate response to Porphyromonas gingivalis: activation leads to bacterial persistence and TLR2 deficiency attenuates induced alveolar bone resorption. J. Immunol. 177, 8296–8300 (2006).

    Article  CAS  PubMed  Google Scholar 

  95. Gibson, F. C. 3rd, Ukai, T., Genco, C. Engagement of specific innate immune signaling pathways during Porphyromonas givgivalis induced chronic inflammation and atherosclerosis. Front. Biosci. 13, 2041–2059 (2008).

    Article  CAS  PubMed  Google Scholar 

  96. Ji, S., Kim, Y., Min., B. M., Han, S. H. & Choi, Y. Innate immune responses of gingival epithelial cells to nonperiodontopathic and periodontopathic bacteria. J. Periodont. Res. 42, 503–510 (2007).

    Article  CAS  Google Scholar 

  97. Huang, G. T., Kim, D., Lee, J. K., Kuramitsu, H. K. & Haake, S. K. Interleukin-8 and intercellular adhesion molecule 1 regulation in oral epithelial cells by selected periodontal bacteria: multiple effects of Porphyromonas gingivalis via antagonistic mechanisms. Infect. Immun. 69, 1364–1372 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Vankeerberghen, A. et al. Differential induction of human beta-defensin expression by periodontal commensals and pathogens in periodontal pocket epithelial cells. J. Periodontol. 76, 1293–1303 (2005).

    Article  CAS  PubMed  Google Scholar 

  99. Hasegawa, Y. et al. Role of Porphyromonas gingivalis SerB in gingival epithelial cell cytoskeletal remodeling and cytokine production. Infect. Immun. 76, 2420–2427 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Tribble, G. D., Mao, S., James, C. E. & Lamont, R. J. A Porphyromonas gingivalis haloacid dehalogenase family phosphatase interacts with human phosphoproteins and is important for invasion. Proc. Natl Acad. Sci. USA 103, 11027–11032 (2006). This article describes a novel protein that is secreted by P. gingivalis and that disrupts gingival epithelial cell function. The authors postulate that P. gingivalis has adapted a former metabolic enzyme to facilitate entry into host cells by modulating host cytoskeletal architecture.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Brissette, C. A., Pham, T. T., Coats, S. R., Darveau, R. P. & Lukehart, S. A. Treponema denticola does not induce production of common innate immune mediators from primary gingival epithelial cells. Oral Microbiol. Immunol. 23, 474–481 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hajishengallis, G., Wang, M., Liang, S., Triantafilou, M. & Triantafilou, K. Pathogen induction of CXCR4/TLR2 cross-talk impairs host defense function. Proc. Natl Acad. Sci. USA 105, 13532–13537 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Coats, S. R., Pham, T. T., Bainbridge, B. W., Reife, R. A. & Darveau, R. P. MD-2 mediates the ability of tetra-acylated and penta-acylated lipopolysaccharides to antagonize Escherichia coli lipopolysaccharide at the TLR4 signaling complex. J. Immunol. 175, 4490–4498 (2005).

    Article  CAS  PubMed  Google Scholar 

  104. Coats, S. R., Do, C. T., Karimi-Naser, L. M., Braham, P. H. & Darveau, R. P. Antagonistic lipopolysaccharides block E. coli lipopolysaccharide function at human TLR4 via interaction with the human MD-2 lipopolysaccharide binding site. Cell. Microbiol. 9, 1191–1202 (2007).

    Article  CAS  PubMed  Google Scholar 

  105. Silipo, A., Lanzetta, R., Amoresano, A., Parrilli, M. & Molinaro, A. Ammonium hydroxide hydrolysis: a valuable support in the MALDI-TOF mass spectrometry analysis of lipid A fatty acid distribution. J. Lipid Res. 43, 2188–2195 (2002).

    Article  CAS  PubMed  Google Scholar 

  106. Rund, S., Lindner, B., Brade, H. & Holst, O. Structural analysis of the lipopolysaccharide from Chlamydia trachomatis serotype L2. J. Biol. Chem. 274, 16819–16824 (1999).

    Article  CAS  PubMed  Google Scholar 

  107. Que, N. L., Lin, S., Cotter, R. J. & Raetz, C. R. Purification and mass spectrometry of six lipid A species from the bacterial endosymbiont Rhizobium etli. Demonstration of a conserved distal unit and a variable proximal portion. J. Biol. Chem. 275, 28006–28016 (2000).

    CAS  PubMed  Google Scholar 

  108. Aussel, L., Brisson, J. R., Perry, M. B. & Caroff, M. Structure of the lipid A of Bordetella hinzii ATCC 51730. Rapid Commun. Mass Spectrom. 14, 595–599 (2000).

    Article  CAS  PubMed  Google Scholar 

  109. Therisod, H., Monteiro, M. A., Perry, M. B. & Caroff, M. Helicobacter mustelae lipid A structure differs from that of Helicobacter pylori. FEBS Lett. 499, 1–5 (2001).

    Article  CAS  PubMed  Google Scholar 

  110. Bainbridge, B. W., Coats, S. R. & Darveau, R. P. Porphyromonas gingivalis lipopolysaccharide displays functionally diverse interactions with the innate host defense system. Ann. Periodontol. 7, 1–9 (2002).

    Article  Google Scholar 

  111. Kumada, H., Haishima, Y., Umemoto, T. & Tanamoto, K.-I. Structural study on the free lipid A isolated from lipopolysaccharide of Porphyromonas gingivalis. J. Bacteriol. 177, 2098–2106 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Yi, E. C. & Hackett, M. Rapid isolation method for lipopolysaccharide and lipid A from Gram-negative bacteria. Analyst 125, 651–656 (2000).

    Article  CAS  PubMed  Google Scholar 

  113. Reife, R. A. et al. Porphyromonas gingivalis lipopolysaccharide lipid A heterogeneity: differential activities of tetra- and penta-acylated lipid A structures on E-selectin expression and TLR4 recognition. Cell. Microbiol. 8, 857–868 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Fujiwara, T., Ogawa, T., Sobue, S. & Hamada, S. Chemical, immunobiological and antigenic characterizations of lipopolysaccharides from Bacteroides gingivalis strains. J. Gen. Microbiol. 136, 319–326 (1990).

    Article  CAS  PubMed  Google Scholar 

  115. Zhang, Y., Gaekwad, J., Wolfert, M. A. & Boons, G. J. Synthetic tetra-acylated derivatives of lipid A from Porphyromonas gingivalis are antagonists of human TLR4. Org. Biomol. Chem. 6, 3371–3381 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Kumada, H. et al. Biological properties of the native and synthetic lipid A of Porphyromonas gingivalis lipopolysaccharide. Oral Microbiol. Immunol. 23, 60–69 (2008).

    Article  CAS  PubMed  Google Scholar 

  117. Sawada, N., Ogawa, T., Asai, Y., Makimura, Y. & Sugiyama, A. Toll-like receptor 4-dependent recognition of structurally different forms of chemically synthesized lipid As of Porphyromonas gingivalis. Clin. Exp. Immunol. 148, 529–536 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Al-Qutub, M. N. et al. Hemin-dependent modulation of the lipid A structure of Porphyromonas gingivalis lipopolysaccharide. Infect. Immun. 74, 4474–4485 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Coats, S. R. et al. Human Toll-like receptor 4 responses to P. gingivalis are regulated by lipid A 1- and 4'-phosphatase activities. Cell. Microbiol 11, 1587–1599 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Lu, Q., Darveau, R., Samaranayke, L., Wang, C., Lijian, J. Differential modulation of human β-defensins expression in human gingival epithelia by Porphyromonas gingivalis lipopolysaccharide with tetra- and penta-acylated lipid A structures. Innate Immun. 15, 325–335 (2009).

    Article  CAS  PubMed  Google Scholar 

  121. Quinchia-Rios, B. H. et al. Down-regulation of epidermal growth factor receptor-dependent signaling by Porphyromonas gingivalis lipopolysaccharide in life-expanded human gingival fibroblasts. J. Periodont. Res. 43, 290–304 (2008).

    Article  CAS  Google Scholar 

  122. Cardelli, P. et al. The modification of the extracellular matrix synthesized in vitro by human gingival fibroblasts in relation to aging. G. Chir. 13, 83–86 (1992) (in Italian).

    CAS  PubMed  Google Scholar 

  123. Marsh, P. D. Dental plaque as a biofilm and a microbial community - implications for health and disease. BMC Oral Health 6 (Suppl. 1), S14 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Kuramitsu, H. K., He, X., Lux, R., Anderson, M. H. & Shi, W. Interspecies interactions within oral microbial communities. Microbiol. Mol. Biol. Rev. 71, 653–670 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Costerton, W. et al. The application of biofilm science to the study and control of chronic bacterial infections. J. Clin. Invest. 112, 1466–1477 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Darveau, R. P. et al. Ability of bacteria associated with chronic inflammatory disease to stimulate E-selectin expression and promote neutrophil adhesion. Infect. Immun. 63, 1311–1317 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Grenier, D. & Mayrand, D. Functional characterization of extracellular vesicles produced by Bacteroides gingivalis. Infect. Immun. 55, 111–117 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Schwartz, J., Stinson, F. L. & Parker, R. B. The passage of tritiated bacterial endotoxin across intact gingival crevicular epithelium. J. Periodontol. 43, 270–276 (1972).

    Article  CAS  PubMed  Google Scholar 

  129. Potempa, J., Banbula, A. & Travis, J. Role of bacterial proteinases in matrix destruction and modulation of host responses. Periodontol. 2000 24, 153–192 (2000).

    Article  CAS  PubMed  Google Scholar 

  130. Tanner, A. C. & Izard, J. Tannerella forsythia, a periodontal pathogen entering the genomic era. Periodontol. 2000 42, 88–113 (2006).

    Article  PubMed  Google Scholar 

  131. Finlay, B. B. & Falkow, S. Common themes in microbial pathogenicity. Microbiol. Rev. 53, 210–230 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. McNab, R. et al. LuxS-based signaling in Streptococcus gordonii: autoinducer 2 controls carbohydrate metabolism and biofilm formation with Porphyromonas gingivalis. J. Bacteriol. 185, 274–284 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Yuan, L., Hillman, J. D. & Progulske-Fox, A. Microarray analysis of quorum-sensing-regulated genes in Porphyromonas gingivalis. Infect. Immun. 73, 4146–4154 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Yuan, L., Rodrigues, P. H., Belanger, M., Dunn, W. A. Jr & Progulske-Fox, A. Porphyromonas gingivalis htrA is involved in cellular invasion and in vivo survival. Microbiology 154, 1161–1169 (2008).

    Article  CAS  PubMed  Google Scholar 

  135. Xie, H., Lin, X., Wang, B. Y., Wu, J. & Lamont, R. J. Identification of a signalling molecule involved in bacterial intergeneric communication. Microbiology 153, 3228–3234 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Chalmers, N. I., Palmer, R. J. Jr., Cisar, J. O. & Kolenbrander, P. E. Characterization of a Streptococcus sp-Veillonella sp. community micromanipulated from dental plaque. J. Bacteriol. 190, 8145–8154 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Ramsey, M. M. & Whiteley, M. Polymicrobial interactions stimulate resistance to host innate immunity through metabolite perception. Proc. Natl Acad. Sci. USA 106, 1578–1583 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Haffajee, A. D. & Socransky, S. S. Microbiology of periodontal diseases: introduction. Periodontol. 2000 38, 9–12 (2005).

    Article  PubMed  Google Scholar 

  139. Bonito, A. J., Lux, L. & Lohr, K. N. Impact of local adjuncts to scaling and root planing in periodontal disease therapy: a systematic review. J. Periodontol. 76, 1227–1236 (2005).

    Article  PubMed  Google Scholar 

  140. Rifkin, B. R., Vernillo, A. T. & Golub, L. M. Blocking periodontal disease progression by inhibiting tissue-destructive enzymes: a potential therapeutic role for tetracyclines and their chemically-modified analogs. J. Periodontol. 64, 819–827 (1993).

    Article  CAS  PubMed  Google Scholar 

  141. Kirkwood, K. L., Cirelli, J. A., Rogers, J. E. & Giannobile, W. V. Novel host response therapeutic approaches to treat periodontal diseases. Periodontol. 2000 43, 294–315 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  142. Booth, V., Ashley, F. P. & Lehner, T. Passive immunization with monoclonal antibodies against Porphyromonas gingivalis in patients with periodontitis. Infect. Immun. 64, 422–427 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Page, R. C. et al. Immunization of Macaca fascicularis against experimental periodontitis using a vaccine containing cysteine proteases purified from Porphyromonas gingivalis. Oral Microbiol. Immunol. 22, 162–168 (2007).

    Article  CAS  PubMed  Google Scholar 

  144. O' Brien-Simpson, N. M. et al. An immune response directed to proteinase and adhesin functional epitopes protects against Porphyromonas gingivalis-induced periodontal bone loss. J. Immunol. 175, 3980–3989 (2005).

    Article  Google Scholar 

  145. Katz, J., Black, K. P. & Michalek, S. M. Host responses to recombinant hemagglutinin B of Porphyromonas gingivalis in an experimental rat model. Infect. Immun. 67, 4352–4359 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Momoi, F. et al. Nasal vaccination with the 40-kilodalton outer membrane protein of Porphyromonas gingivalis and a nontoxic chimeric enterotoxin adjuvant induces long-term protective immunity with reduced levels of immunoglobulin E antibodies. Infect. Immun. 76, 2777–2784 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Potempa, J., Sroka, A., Imamura, T. & Travis, J. Gingipains, the major cysteine proteinases and virulence factors of Porphyromonas gingivalis: structure, function and assembly of multidomain protein complexes. Curr. Protein Pept. Sci. 4, 397–407 (2003).

    Article  CAS  PubMed  Google Scholar 

  148. Miyachi, K., Ishihara, K., Kimizuka, R. & Okuda, K. Arg-gingipain A DNA vaccine prevents alveolar bone loss in mice. J. Dent. Res. 86, 446–450 (2007).

    Article  CAS  PubMed  Google Scholar 

  149. Van Dyke, T. E. Control of inflammation and periodontitis. Periodontol. 2000 45, 158–166 (2007). This article describes a new class of pro-resolving agents and points out that active resolution of an inflammatory response can restore host homeostasis in chronic inflammatory diseases; periodontitis is used as an example of this fact.

    Article  PubMed  Google Scholar 

  150. Serhan, C. N., Chiang, N. & Van Dyke, T. E. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nature Rev. Immunol. 8, 349–361 (2008).

    Article  CAS  Google Scholar 

  151. Graves, D. et al. The use of rodent models to investigate host-bacteria interactions related to periodontal diseases. J. Clin. Periodontol. 35, 89–105 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The author thanks M. Curtis for critical review of the manuscript, M. Thomashow and F. Roberts for fruitful discussions and C. Zenobia for help with the figures. He also thanks the past and present members of his lab and department for continued inspiring conversations. The editorial assistance of N. Balch is greatly appreciated. Work in the author's laboratory is supported by the National Institute of Dental and Craniofacial Research.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

Related links

DATABASES

Entrez Genome

Treponema denticola

Entrez Genome Project

Aggregatibacter actinomycetemcomitans

Macaca fascicularis

Porphyromonas gingivalis

Tannerella forsythia

FURTHER INFORMATION

Richard Darveau's homepage

Glossary

Periodontium

The epithelial, connective and bone tissues that both surround and support the teeth. The word comes from the Greek terms 'peri', meaning 'around', and -'odons', meaning 'tooth'.

Dental plaque

A polymicrobial biofilm community that builds up on the surface of the tooth and tooth root. Plaque can also become mineralized and form a calculus.

Microbial-shift disease

A disease caused by a decrease in the number of beneficial symbionts and/or an increase in the number of pathogens. This concept is also known as dysbiosis.

Junctional epithelium

A specialized epithelium located at the interface between the gingival sulcus, which is populated with bacteria, and the periodontal soft and mineralized connective tissues. It connects the tooth surface to the host tissue.

Germ-free mice

Mice that are completely devoid of bacteria. They are generated by sterile Caesarean section, raised aseptically in an isolator with sterile filtered air and housed using sterile food, water and bedding. Germ-free mice are distinct from specific-pathogen-free (SPF) mice, which are devoid of only known mouse pathogens and still contain intestinal bacteria.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Darveau, R. Periodontitis: a polymicrobial disruption of host homeostasis. Nat Rev Microbiol 8, 481–490 (2010). https://doi.org/10.1038/nrmicro2337

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro2337

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing