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:

The oral microbiota: dynamic communities and host interactions

Abstract

The dynamic and polymicrobial oral microbiome is a direct precursor of diseases such as dental caries and periodontitis, two of the most prevalent microbially induced disorders worldwide. Distinct microenvironments at oral barriers harbour unique microbial communities, which are regulated through sophisticated signalling systems and by host and environmental factors. The collective function of microbial communities is a major driver of homeostasis or dysbiosis and ultimately health or disease. Despite different aetiologies, periodontitis and caries are each driven by a feedforward loop between the microbiota and host factors (inflammation and dietary sugars, respectively) that favours the emergence and persistence of dysbiosis. In this Review, we discuss current knowledge and emerging mechanisms governing oral polymicrobial synergy and dysbiosis that have both enhanced our understanding of pathogenic mechanisms and aided the design of innovative therapeutic approaches for oral diseases.

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

Fig. 1: Biogeography of oral microbiota colonization in the diverse habitats of the oral cavity.
Fig. 2: Interactions among bacterial species that affect nososymbiocity.

Part a adapted with permission from ref.56, Wiley-VCH.

Fig. 3: Diet–microbiota interactions trigger the assembly of cariogenic biofilm microenvironment.

Adapted with permission from ref.7, Cell Press.

Fig. 4: Reciprocally reinforced interactions between dysbiosis and inflammation drive chronic periodontitis.
Fig. 5: P. gingivalis induces dysbiosis by impairing innate host defences while promoting inflammatory responses in phagocytic cells.
Fig. 6: Localized chemokine paralysis.

Adapted with permission from ref.10, Cell Press.

Similar content being viewed by others

References

  1. Abusleme, L. et al. The subgingival microbiome in health and periodontitis and its relationship with community biomass and inflammation. ISME J. 7, 1016–1025 (2013). This study documents alterations in subgingival microbial communities that underpin the development of periodontitis and describes the relationship between clinical inflammation and the disease-associated microbiome.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Griffen, A. L. et al. Distinct and complex bacterial profiles in human periodontitis and health revealed by 16S pyrosequencing. ISME J. 6, 1176–1185 (2012). This landmark study establishes the complexity of the periodontal microbial community and the demarcation between health and disease.

    CAS  PubMed  Google Scholar 

  3. Rosan, B. & Lamont, R. J. Dental plaque formation. Microbes Infect. 2, 1599–1607 (2000).

    CAS  PubMed  Google Scholar 

  4. Aas, J. A., Paster, B. J., Stokes, L. N., Olsen, I. & Dewhirst, F. E. Defining the normal bacterial flora of the oral cavity. J. Clin. Microbiol. 43, 5721–5732 (2005).

    PubMed  PubMed Central  Google Scholar 

  5. Dewhirst, F. E. et al. The human oral microbiome. J. Bacteriol. 192, 5002–5017 (2010). References 4 and 5 are the basis of our current understanding of the diversity of the oral microbiome.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Mark Welch, J. L., Rossetti, B. J., Rieken, C. W., Dewhirst, F. E. & Borisy, G. G. Biogeography of a human oral microbiome at the micron scale. Proc. Natl Acad. Sci. USA 113, E791–E800 (2016). This imaging study provides the foundation for concepts of oral microbial biogeography.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Bowen, W. H., Burne, R. A., Wu, H. & Koo, H. Oral biofilms: pathogens, matrix, and polymicrobial interactions in microenvironments. Trends Microbiol. 26, 229–242 (2018).

    CAS  PubMed  Google Scholar 

  8. Kamada, N., Chen, G. Y., Inohara, N. & Nunez, G. Control of pathogens and pathobionts by the gut microbiota. Nat. Immunol. 14, 685–690 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kamada, N., Seo, S. U., Chen, G. Y. & Nunez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 13, 321–335 (2013).

    CAS  PubMed  Google Scholar 

  10. Lamont, R. J. & Hajishengallis, G. Polymicrobial synergy and dysbiosis in inflammatory disease. Trends Mol. Med. 21, 172–183 (2015).

    CAS  PubMed  Google Scholar 

  11. Hajishengallis, G. & Lamont, R. J. Breaking bad: manipulation of the host response by Porphyromonas gingivalis. Eur. J. Immunol. 44, 328–338 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Baker, J. L., Bor, B., Agnello, M., Shi, W. & He, X. Ecology of the oral microbiome: beyond bacteria. Trends Microbiol. 25, 362–374 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Dabdoub, S. M., Ganesan, S. M. & Kumar, P. S. Comparative metagenomics reveals taxonomically idiosyncratic yet functionally congruent communities in periodontitis. Sci. Rep. 6, 38993 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Suwannakul, S., Stafford, G. P., Whawell, S. A. & Douglas, C. W. Identification of bistable populations of Porphyromonas gingivalis that differ in epithelial cell invasion. Microbiology 156, 3052–3064 (2010).

    CAS  PubMed  Google Scholar 

  15. Valm, A. M. et al. Systems-level analysis of microbial community organization through combinatorial labeling and spectral imaging. Proc. Natl Acad. Sci. USA 108, 4152–4157 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Marsh, P. D. & Zaura, E. Dental biofilm: ecological interactions in health and disease. J. Clin. Periodontol. 44 (Suppl. 18), 12–22 (2017).

    Google Scholar 

  17. Takahashi, N. & Nyvad, B. The role of bacteria in the caries process: ecological perspectives. J. Dent. Res. 90, 294–303 (2011).

    CAS  PubMed  Google Scholar 

  18. Xiao, J. et al. The exopolysaccharide matrix modulates the interaction between 3D architecture and virulence of a mixed-species oral biofilm. PLOS Pathog. 8, e1002623 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Guo, L., McLean, J. S., Lux, R., He, X. & Shi, W. The well-coordinated linkage between acidogenicity and aciduricity via insoluble glucans on the surface of Streptococcus mutans. Sci. Rep. 5, 18015 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hajishengallis, E., Parsaei, Y., Klein, M. I. & Koo, H. Advances in the microbial etiology and pathogenesis of early childhood caries. Mol. Oral Microbiol. 32, 24–34 (2017).

    CAS  PubMed  Google Scholar 

  21. Mira, A., Simon-Soro, A. & Curtis, M. A. Role of microbial communities in the pathogenesis of periodontal diseases and caries. J. Clin. Periodontol. 44 (Suppl. 18), S23–S38 (2017).

    PubMed  Google Scholar 

  22. Tanner, A. C. R., Kressirer, C. A., Rothmiller, S., Johansson, I. & Chalmers, N. I. The caries microbiome: implications for reversing dysbiosis. Adv. Dent. Res. 29, 78–85 (2018).

    CAS  PubMed  Google Scholar 

  23. Eriksson, L., Lif Holgerson, P., Esberg, A. & Johansson, I. Microbial complexes and caries in 17-year-olds with and without Streptococcus mutans. J. Dent. Res. 97, 275–282 (2018).

    CAS  PubMed  Google Scholar 

  24. Hajishengallis, G. & Lamont, R. J. Beyond the red complex and into more complexity: the polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology. Mol. Oral Microbiol. 27, 409–419 (2012). This paper questions the primary importance of individual pathogens, such as the red complex bacteria, and proposes that periodontitis is initiated by a synergistic polymicrobial community within which different species, or specific gene combinations thereof, mediate distinct roles that converge to shape and stabilize a dysbiotic and disease-provoking microbiota.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Holt, S. C. & Ebersole, J. L. Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia: the “red complex”, a prototype polybacterial pathogenic consortium in periodontitis. Periodontol. 2000 38, 72–122 (2005).

    PubMed  Google Scholar 

  26. Socransky, S. S. & Haffajee, A. D. Periodontal microbial ecology. Periodontol. 2000 38, 135–187 (2005).

    PubMed  Google Scholar 

  27. Dewhirst, F. E. The oral microbiome: critical for understanding oral health and disease. J. Calif. Dent. Assoc. 44, 409–410 (2016).

    PubMed  PubMed Central  Google Scholar 

  28. Diaz, P. I., Hoare, A. & Hong, B. Y. Subgingival microbiome shifts and community dynamics in periodontal diseases. J. Calif. Dent. Assoc. 44, 421–435 (2016).

    PubMed  Google Scholar 

  29. Moore, W. E. et al. The microflora of periodontal sites showing active destructive progression. J. Clin. Periodontol. 18, 729–739 (1991).

    CAS  PubMed  Google Scholar 

  30. Diaz, P. I. Microbial diversity and interactions in subgingival biofilm communities. Front. Oral Biol. 15, 17–40 (2012).

    PubMed  Google Scholar 

  31. Simon-Soro, A. et al. Microbial geography of the oral cavity. J. Dent. Res. 92, 616–621 (2013).

    CAS  PubMed  Google Scholar 

  32. Nowicki, E. M. et al. Microbiota and metatranscriptome changes accompanying the onset of gingivitis. mBio 9, e00575–18 (2018).

    PubMed  PubMed Central  Google Scholar 

  33. Whitmore, S. E. & Lamont, R. J. Oral bacteria and cancer. PLOS Pathog. 10, e1003933 (2014).

    PubMed  PubMed Central  Google Scholar 

  34. Atanasova, K. R. & Yilmaz, O. Looking in the Porphyromonas gingivalis cabinet of curiosities: the microbium, the host and cancer association. Mol. Oral Microbiol. 29, 55–66 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Sahingur, S. E. & Yeudall, W. A. Chemokine function in periodontal disease and oral cavity cancer. Front. Immunol. 6, 214 (2015).

    PubMed  PubMed Central  Google Scholar 

  36. Cugini, C., Klepac-Ceraj, V., Rackaityte, E., Riggs, J. E. & Davey, M. E. Porphyromonas gingivalis: keeping the pathos out of the biont. J. Oral Microbiol. 5, 19804 (2013).

    Google Scholar 

  37. Takahashi, N. Oral microbiome metabolism: from “who are they?” to “what are they doing?”. J. Dent. Res. 94, 1628–1637 (2015).

    CAS  PubMed  Google Scholar 

  38. Han, Y. W. & Wang, X. Mobile microbiome: oral bacteria in extra-oral infections and inflammation. J. Dent. Res. 92, 485–491 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hajishengallis, G. Periodontitis: from microbial immune subversion to systemic inflammation. Nat. Rev. Immunol. 15, 30–44 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Kumar, P. S. Oral microbiota and systemic disease. Anaerobe 24, 90–93 (2013).

    PubMed  Google Scholar 

  41. Maddi, A. & Scannapieco, F. A. Oral biofilms, oral and periodontal infections, and systemic disease. Am. J. Dent. 26, 249–254 (2013).

    PubMed  Google Scholar 

  42. Potempa, J., Mydel, P. & Koziel, J. The case for periodontitis in the pathogenesis of rheumatoid arthritis. Nat. Rev. Rheumatol. 13, 606–620 (2017).

    CAS  PubMed  Google Scholar 

  43. Ebersole, J. L. et al. The periodontal war: microbes and immunity. Periodontol. 2000 75, 52–115 (2017).

    PubMed  Google Scholar 

  44. Chukkapalli, S. S. et al. Global TLR2 and 4 deficiency in mice impacts bone resorption, inflammatory markers and atherosclerosis to polymicrobial infection. Mol. Oral Microbiol. 32, 211–225 (2017).

    CAS  PubMed  Google Scholar 

  45. Hajishengallis, G. & Lamont, R. J. Dancing with the stars: how choreographed bacterial interactions dictate nososymbiocity and give rise to keystone pathogens, accessory pathogens, and pathobionts. Trends Microbiol. 24, 477–489 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Murray, J. L., Connell, J. L., Stacy, A., Turner, K. H. & Whiteley, M. Mechanisms of synergy in polymicrobial infections. J. Microbiol. 52, 188–199 (2014).

    PubMed  PubMed Central  Google Scholar 

  47. Michie, K. L., Cornforth, D. M. & Whiteley, M. Bacterial tweets and podcasts #signaling#eavesdropping#microbialfightclub. Mol. Biochem. Parasitol. 208, 41–48 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Short, F. L., Murdoch, S. L. & Ryan, R. P. Polybacterial human disease: the ills of social networking. Trends Microbiol. 22, 508–516 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Stacy, A., McNally, L., Darch, S. E., Brown, S. P. & Whiteley, M. The biogeography of polymicrobial infection. Nat. Rev. Microbiol. 14, 93–105 (2016).

    CAS  PubMed  Google Scholar 

  50. Hwang, G. et al. Candida albicans mannans mediate Streptococcus mutans exoenzyme GtfB binding to modulate cross-kingdom biofilm development in vivo. PLOS Pathog. 13, e1006407 (2017).

    PubMed  PubMed Central  Google Scholar 

  51. Kim, D. et al. Candida albicans stimulates Streptococcus mutans microcolony development via cross-kingdom biofilm-derived metabolites. Sci. Rep. 7, 41332 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen, Y. E., Fischbach, M. A. & Belkaid, Y. Skin microbiota-host interactions. Nature 553, 427–436 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Casadevall, A. The pathogenic potential of a microbe. mSphere 2, e00015–17 (2017).

    PubMed  PubMed Central  Google Scholar 

  54. Lin, D. & Koskella, B. Friend and foe: factors influencing the movement of the bacterium Helicobacter pylori along the parasitism-mutualism continuum. Evol. Appl. 8, 9–22 (2015).

    PubMed  Google Scholar 

  55. Nelson, P. G. & May, G. Coevolution between mutualists and parasites in symbiotic communities may lead to the evolution of lower virulence. Am. Nat. 190, 803–817 (2017).

    PubMed  Google Scholar 

  56. Whitmore, S. E. & Lamont, R. J. The pathogenic persona of community-associated oral streptococci. Mol. Microbiol. 81, 305–314 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Nobbs, A. H., Lamont, R. J. & Jenkinson, H. F. Streptococcus adherence and colonization. Microbiol. Mol. Biol. Rev. 73, 407–450 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Lee, S. F. Oral colonization and immune responses to Streptococcus gordonii: potential use as a vector to induce antibodies against respiratory pathogens. Curr. Opin. Infect. Dis. 16, 231–235 (2003).

    CAS  PubMed  Google Scholar 

  59. Xie, E. et al. Oral delivery of a novel recombinant Streptococcus mitis vector elicits robust vaccine antigen-specific oral mucosal and systemic antibody responses and T cell tolerance. PLOS ONE 10, e0143422 (2015).

    PubMed  PubMed Central  Google Scholar 

  60. Daep, C. A., Novak, E. A., Lamont, R. J. & Demuth, D. R. Structural dissection and in vivo effectiveness of a peptide inhibitor of Porphyromonas gingivalis adherence to Streptococcus gordonii. Infect. Immun. 79, 67–74 (2011).

    CAS  PubMed  Google Scholar 

  61. Kuboniwa, M. et al. Metabolic crosstalk regulates Porphyromonas gingivalis colonization and virulence during oral polymicrobial infection. Nat. Microbiol. 2, 1493–1499 (2017). This study establishes multidimensional communication between two organisms in the oral community that separately either enhance or suppress nososymbiocity.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Wright, C. J. et al. Characterization of a bacterial tyrosine kinase in Porphyromonas gingivalis involved in polymicrobial synergy. Microbiologyopen 3, 383–394 (2014).

    PubMed  PubMed Central  Google Scholar 

  63. Liu, C., Miller, D. P., Wang, Y., Merchant, M. & Lamont, R. J. Structure-function aspects of the Porphyromonas gingivalis tyrosine kinase Ptk1. Mol. Oral Microbiol. 32, 314–323 (2017).

    CAS  PubMed  Google Scholar 

  64. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Stacy, A. et al. Bacterial fight-and-flight responses enhance virulence in a polymicrobial infection. Proc. Natl Acad. Sci. USA 111, 7819–7824 (2014). This study reveals that the spatial proximity of organisms can depend on the collective outcome of synergistic and antagonistic interactions.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Duan, D., Scoffield, J. A., Zhou, X. & Wu, H. Fine-tuned production of hydrogen peroxide promotes biofilm formation of Streptococcus parasanguinis by a pathogenic cohabitant Aggregatibacter actinomycetemcomitans. Environ. Microbiol. 18, 4023–4036 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Bertolini, M. M. et al. Candida-streptococcal mucosal biofilms display distinct structural and virulence characteristics depending on growth conditions and hyphal morphotypes. Mol. Oral Microbiol. 30, 307–322 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Xu, H., Jenkinson, H. F. & Dongari-Bagtzoglou, A. Innocent until proven guilty: mechanisms and roles of Streptococcus-Candida interactions in oral health and disease. Mol. Oral Microbiol. 29, 99–116 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Cheng, X. et al. Plasticity of the pyruvate node modulates hydrogen peroxide production and acid tolerance in multiple oral Streptococci. Appl. Environ. Microbiol. 84, e01697–17 (2018).

    PubMed  PubMed Central  Google Scholar 

  70. Thurnheer, T. & Belibasakis, G. N. Streptococcus oralis maintains homeostasis in oral biofilms by antagonizing the cariogenic pathogen Streptococcus mutans. Mol. Oral Microbiol. 33, 234–239 (2018).

    CAS  PubMed  Google Scholar 

  71. Redanz, S. et al. Live and let die: hydrogen peroxide production by the commensal flora and its role in maintaining a symbiotic microbiome. Mol. Oral Microbiol. https://doi.org/10.1111/omi.12231 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Ho, M. H., Lamont, R. J. & Xie, H. Identification of Streptococcus cristatus peptides that repress expression of virulence genes in Porphyromonas gingivalis. Sci. Rep. 7, 1413 (2017).

    PubMed  PubMed Central  Google Scholar 

  73. Ho, M. H., Lamont, R. J. & Xie, H. A novel peptidic inhibitor derived from Streptococcus cristatus ArcA attenuates virulence potential of Porphyromonas gingivalis. Sci. Rep. 7, 16217 (2017).

    PubMed  PubMed Central  Google Scholar 

  74. Xie, H., Hong, J., Sharma, A. & Wang, B. Y. Streptococcus cristatus ArcA interferes with Porphyromonas gingivalis pathogenicity in mice. J. Periodontal. Res. 47, 578–583 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang, B. Y., Wu, J., Lamont, R. J., Lin, X. & Xie, H. Negative correlation of distributions of Streptococcus cristatus and Porphyromonas gingivalis in subgingival plaque. J. Clin. Microbiol. 47, 3902–3906 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Mans, J. J., Hendrickson, E. L., Hackett, M. & Lamont, R. J. Cellular and bacterial profiles associated with oral epithelium-microbiota interactions. Periodontol. 2000 52, 207–217 (2010).

    PubMed  PubMed Central  Google Scholar 

  77. Colombo, A. V., Silva, C. M., Haffajee, A. & Colombo, A. P. Identification of oral bacteria associated with crevicular epithelial cells from chronic periodontitis lesions. J. Med. Microbiol. 55, 609–615 (2006).

    PubMed  Google Scholar 

  78. Kreth, J., Giacaman, R. A., Raghavan, R. & Merritt, J. The road less traveled - defining molecular commensalism with Streptococcus sanguinis. Mol. Oral Microbiol. 32, 181–196 (2017).

    CAS  PubMed  Google Scholar 

  79. Shah, S. A. et al. The making of a miscreant: tobacco smoke and the creation of pathogen-rich biofilms. NPJ Biofilms Microbiomes 3, 26 (2017).

    PubMed  PubMed Central  Google Scholar 

  80. Ghosh, S. K. et al. Conceptual perspectives: bacterial antimicrobial peptide induction as a novel strategy for symbiosis with the human host. Front. Microbiol. 9, 302 (2018).

    PubMed  PubMed Central  Google Scholar 

  81. Al-Attar, A. et al. Activation of Notch-1 in oral epithelial cells by P. gingivalis triggers the expression of the antimicrobial protein PLA2-IIA. Mucosal Immunol. 11, 1047–1059 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Pitts, N. B. et al. Dental caries. Nat. Rev. Dis. Primers 3, 17030 (2017).

    PubMed  Google Scholar 

  83. Liu, Y. L., Nascimento, M. & Burne, R. A. Progress toward understanding the contribution of alkali generation in dental biofilms to inhibition of dental caries. Int. J. Oral Sci. 4, 135–140 (2012).

    PubMed  PubMed Central  Google Scholar 

  84. Merritt, J. & Qi, F. The mutacins of Streptococcus mutans: regulation and ecology. Mol. Oral Microbiol. 27, 57–69 (2012).

    CAS  PubMed  Google Scholar 

  85. Qi, F. & Kreth, J. Methods to study antagonistic activities among oral bacteria. Methods Mol. Biol. 1537, 203–218 (2017).

    CAS  PubMed  Google Scholar 

  86. Gross, E. L. et al. Beyond Streptococcus mutans: dental caries onset linked to multiple species by 16S rRNA community analysis. PLOS ONE 7, e47722 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Johansson, I., Witkowska, E., Kaveh, B., Lif Holgerson, P. & Tanner, A. C. The microbiome in populations with a low and high prevalence of caries. J. Dent. Res. 95, 80–86 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Teng, F. et al. Prediction of early childhood caries via spatial-temporal variations of oral microbiota. Cell Host Microbe 18, 296–306 (2015).

    CAS  PubMed  Google Scholar 

  89. Takahashi, N. & Nyvad, B. Ecological hypothesis of dentin and root caries. Caries Res. 50, 422–431 (2016).

    CAS  PubMed  Google Scholar 

  90. Richards, V. P. et al. Microbiomes of site-specific dental plaques from children with different caries status. Infect. Immun. 85, e00106–17 (2017).

    PubMed  PubMed Central  Google Scholar 

  91. Knapp, S. et al. Natural competence is common among clinical isolates of Veillonella parvula and is useful for genetic manipulation of this key member of the oral microbiome. Front. Cell. Infect. Microbiol. 7, 139 (2017).

    PubMed  PubMed Central  Google Scholar 

  92. Mashima, I. & Nakazawa, F. The influence of oral Veillonella species on biofilms formed by Streptococcus species. Anaerobe 28, 54–61 (2014).

    PubMed  Google Scholar 

  93. Xiao, J. et al. Candida albicans and early childhood caries: a systematic review and meta-analysis. Caries Res. 52, 102–112 (2018).

    PubMed  Google Scholar 

  94. Falsetta, M. L. et al. Symbiotic relationship between Streptococcus mutans and Candida albicans synergizes virulence of plaque biofilms in vivo. Infect. Immun. 82, 1968–1981 (2014).

    PubMed  PubMed Central  Google Scholar 

  95. Sztajer, H. et al. Cross-feeding and interkingdom communication in dual-species biofilms of Streptococcus mutans and Candida albicans. ISME J. 8, 2256–2271 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Flemming, H. C. et al. Biofilms: an emergent form of bacterial life. Nat. Rev. Microbiol. 14, 563–575 (2016).

    CAS  PubMed  Google Scholar 

  97. Hwang, G. et al. Simultaneous spatiotemporal mapping of in situ pH and bacterial activity within an intact 3D microcolony structure. Sci. Rep. 6, 32841 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Jiang, W. et al. Pyrosequencing analysis of oral microbiota shifting in various caries states in childhood. Microb. Ecol. 67, 962–969 (2014).

    PubMed  Google Scholar 

  99. Marsh, P. D. Are dental diseases examples of ecological catastrophes? Microbiology 149, 279–294 (2003). This paper proposes that environmental factors drive the selection and enrichment of specific oral pathogenic bacteria, with implications for both dental caries and periodontitis.

    CAS  PubMed  Google Scholar 

  100. Hasturk, H. et al. Resolvin E1 regulates inflammation at the cellular and tissue level and restores tissue homeostasis in vivo. J. Immunol. 179, 7021–7029 (2007).

    CAS  PubMed  Google Scholar 

  101. Abe, T. et al. Local complement-targeted intervention in periodontitis: proof-of-concept using a C5a receptor (CD88) antagonist. J. Immunol. 189, 5442–5448 (2012).

    CAS  PubMed  Google Scholar 

  102. Hajishengallis, G. et al. Low-abundance biofilm species orchestrates inflammatory periodontal disease through the commensal microbiota and complement. Cell Host Microbe 10, 497–506 (2011). This study substantiates the concept of a keystone pathogen by providing in vivo evidence that a specific microorganism instigates quantitative and qualitative alterations to the commensal microbiota, which is thereby remodelled into a dysbiotic community driving periodontitis.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Eskan, M. A. et al. The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss. Nat. Immunol. 13, 465–473 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Moutsopoulos, N. M. et al. Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17–driven inflammatory bone loss. Sci. Transl Med. 6, 229ra240 (2014).

    Google Scholar 

  105. Lee, C.-T. et al. Resolvin E1 reverses experimental periodontitis and dysbiosis. J. Immunol. 197, 2796–2806 (2016).

    CAS  PubMed  Google Scholar 

  106. Hajishengallis, G. The inflammophilic character of the periodontitis-associated microbiota. Mol. Oral Microbiol. 29, 248–257 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Duran-Pinedo, A. E. et al. Community-wide transcriptome of the oral microbiome in subjects with and without periodontitis. ISME J. 8, 1659–1672 (2014).

    PubMed  PubMed Central  Google Scholar 

  108. Herrero, E. R. et al. Dysbiotic biofilms deregulate the periodontal inflammatory response. J. Dent. Res. 97, 547–555 (2018).

    CAS  PubMed  Google Scholar 

  109. Yost, S., Duran-Pinedo, A. E., Teles, R., Krishnan, K. & Frias-Lopez, J. Functional signatures of oral dysbiosis during periodontitis progression revealed by microbial metatranscriptome analysis. Genome Med. 7, 27 (2015).

    PubMed  PubMed Central  Google Scholar 

  110. Bang, J., Cimasoni, G., Rosenbusch, C. & Duckert, A. Sodium, potassium and calcium contents of crevicular exudate: their relations to gingivitis and periodontitis. J. Periodontol. 44, 770–774 (1973).

    CAS  PubMed  Google Scholar 

  111. Yost, S., Duran-Pinedo, A. E., Krishnan, K. & Frias-Lopez, J. Potassium is a key signal in host-microbiome dysbiosis in periodontitis. PLOS Pathog. 13, e1006457 (2017). This investigation identifies key metabolic changes in the periodontal microbial community associated with dysbiosis initiation and concludes that disease progression is mediated by the collective virulence of the entire community rather than by the action of a select few pathogens.

    PubMed  PubMed Central  Google Scholar 

  112. Winter, S. E. & Baumler, A. J. Dysbiosis in the inflamed intestine: chance favors the prepared microbe. Gut Microbes 5, 71–73 (2014).

    PubMed  PubMed Central  Google Scholar 

  113. Nassar, M. et al. GAS6 is a key homeostatic immunological regulator of host-commensal interactions in the oral mucosa. Proc. Natl Acad. Sci. USA 114, E337–E346 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Dalal, S. R. & Chang, E. B. The microbial basis of inflammatory bowel diseases. J. Clin. Invest. 124, 4190–4196 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Hajishengallis, G., Darveau, R. P. & Curtis, M. A. The keystone-pathogen hypothesis. Nat. Rev. Microbiol. 10, 717–725 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Finlay, B. B. & McFadden, G. Anti-immunology: evasion of the host immune system by bacterial and viral pathogens. Cell 124, 767–782 (2006).

    CAS  PubMed  Google Scholar 

  117. Maekawa, T. et al. Porphyromonas gingivalis manipulates complement and TLR signaling to uncouple bacterial clearance from inflammation and promote dysbiosis. Cell Host Microbe 15, 768–778 (2014). This study shows that a keystone periodontal pathogen manipulates complement–TLR crosstalk to block bactericidal mechanisms while fostering a nutritionally favourable inflammatory response; this uncoupling of immune bacterial clearance from inflammation promotes dysbiosis and periodontitis.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Makkawi, H. et al. Porphyromonas gingivalis stimulates TLR2-PI3K signaling to escape immune clearance and induce bone resorption independently of MyD88. Front. Cell. Infect. Microbiol. 7, 359 (2017).

    PubMed  PubMed Central  Google Scholar 

  119. Wang, M. et al. Microbial hijacking of complement-toll-like receptor crosstalk. Sci. Signal. 3, ra11 (2010).

    PubMed  PubMed Central  Google Scholar 

  120. Liang, S. et al. The C5a receptor impairs IL-12-dependent clearance of Porphyromonas gingivalis and is required for induction of periodontal bone loss. J. Immunol. 186, 869–877 (2011).

    CAS  PubMed  Google Scholar 

  121. Tonetti, M. S., Cortellini, D. & Lang, N. P. In situ detection of apoptosis at sites of chronic bacterially induced inflammation in human gingiva. Infect. Immun. 66, 5190–5195 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Zenobia, C. et al. Commensal bacteria-dependent select expression of CXCL2 contributes to periodontal tissue homeostasis. Cell. Microbiol. 15, 1419–1426 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Jauregui, C. E. et al. Suppression of T cell chemokines by Porphyromonas gingivalis. Infect. Immun. 81, 2288–2295 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Takeuchi, H. et al. The serine phosphatase SerB of Porphyromonas gingivalis suppresses IL-8 production by dephosphorylation of NF-kappaB RelA/p65. PLOS Pathog. 9, e1003326 (2013). References 123–125 establish the concept of P. gingivalis-induced local ‘chemokine paralysis’, originally shown to affect innate immunity and later expanded to include suppression of T cell-specific chemokines.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Hajishengallis, G. Immunomicrobial pathogenesis of periodontitis: keystones, pathobionts, and host response. Trends Immunol. 35, 3–11 (2014).

    CAS  PubMed  Google Scholar 

  127. Moutsopoulos, N. M. et al. Subgingival microbial communities in leukocyte adhesion deficiency and their relationship with local immunopathology. PLOS Pathog. 11, e1004698 (2015).

    PubMed  PubMed Central  Google Scholar 

  128. Darveau, R. P., Hajishengallis, G. & Curtis, M. A. Porphyromonas gingivalis as a potential community activist for disease. J. Dent. Res. 91, 816–820 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Koo, H., Allan, R. N., Howlin, R. P., Stoodley, P. & Hall-Stoodley, L. Targeting microbial biofilms: current and prospective therapeutic strategies. Nat. Rev. Microbiol. 15, 740–755 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Liu, Y., Ren, Z., Hwang, G. & Koo, H. Therapeutic strategies targeting cariogenic biofilm microenvironment. Adv. Dent. Res. 29, 86–92 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Marquis, R. E., Clock, S. A. & Mota-Meira, M. Fluoride and organic weak acids as modulators of microbial physiology. FEMS Microbiol. Rev. 26, 493–510 (2003).

    CAS  PubMed  Google Scholar 

  132. Kolderman, E. et al. L-Arginine destabilizes oral multi-species biofilm communities developed in human saliva. PLOS ONE 10, e0121835 (2015).

    PubMed  PubMed Central  Google Scholar 

  133. Nascimento, M. M. et al. The effect of arginine on oral biofilm communities. Mol. Oral Microbiol. 29, 45–54 (2014).

    CAS  PubMed  Google Scholar 

  134. Pleszczynska, M., Wiater, A., Janczarek, M. & Szczodrak, J. (1-→3)-alpha-D-Glucan hydrolases in dental biofilm prevention and control: a review. Int. J. Biol. Macromol. 79, 761–778 (2015).

    CAS  PubMed  Google Scholar 

  135. Fabbri, S. et al. High-velocity microsprays enhance antimicrobial activity in Streptococcus mutans biofilms. J. Dent. Res. 95, 1494–1500 (2016).

    CAS  PubMed  Google Scholar 

  136. Liu, Y. et al. Topical delivery of low-cost protein drug candidates made in chloroplasts for biofilm disruption and uptake by oral epithelial cells. Biomaterials 105, 156–166 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Gao, L. et al. Nanocatalysts promote Streptococcus mutans biofilm matrix degradation and enhance bacterial killing to suppress dental caries in vivo. Biomaterials 101, 272–284 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Horev, B. et al. pH-activated nanoparticles for controlled topical delivery of farnesol to disrupt oral biofilm virulence. ACS Nano 9, 2390–2404 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Liu, Y. et al. Surface-adaptive, antimicrobially loaded, micellar nanocarriers with enhanced penetration and killing efficiency in Staphylococcal biofilms. ACS Nano 10, 4779–4789 (2016). References 137–139 demonstrate the feasibility of using pH-activated nanotechnologies for enhanced drug delivery, release or activation within the biofilm microenvironment to amplify the precision and efficacy of antibiofilm effects.

    CAS  PubMed  Google Scholar 

  140. Paula, A. J. & Koo, H. Nanosized building blocks for customizing novel antibiofilm approaches. J. Dent. Res. 96, 128–136 (2017).

    CAS  PubMed  Google Scholar 

  141. Maekawa, T. et al. Inhibition of pre-existing natural periodontitis in non-human primates by a locally administered peptide inhibitor of complement C3. J. Clin. Periodontol. 43, 238–249 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. 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 

  143. Hasturk, H., Kantarci, A. & Van Dyke, T. E. Paradigm shift in the pharmacological management of periodontal diseases. Front. Oral Biol. 15, 160–176 (2012).

    PubMed  Google Scholar 

  144. Moutsopoulos, N. M. et al. Interleukin-12 and interleukin-23 blockade in leukocyte adhesion deficiency type 1. N. Engl. J. Med. 376, 1141–1146 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Chen, X. et al. Advanced biomaterials and their potential applications in the treatment of periodontal disease. Crit. Rev. Biotechnol. 36, 760–775 (2016).

    CAS  PubMed  Google Scholar 

  146. Goyal, G., Garg, T., Rath, G. & Goyal, A. K. Current nanotechnological strategies for an effective delivery of drugs in treatment of periodontal disease. Crit. Rev. Ther. Drug Carrier Syst. 31, 89–119 (2014).

    CAS  PubMed  Google Scholar 

  147. Morris, J. J., Lenski, R. E. & Zinser, E. R. The black queen hypothesis: evolution of dependencies through adaptive gene loss. mBio 3, e00036–12 (2012).

    PubMed  PubMed Central  Google Scholar 

  148. Mastellos, D. C., Ricklin, D., Hajishengallis, E., Hajishengallis, G. & Lambris, J. D. Complement therapeutics in inflammatory diseases: promising drug candidates for C3-targeted intervention. Mol. Oral Microbiol. 31, 3–17 (2016).

    CAS  PubMed  Google Scholar 

  149. Van Dyke, T. E. Pro-resolving mediators in the regulation of periodontal disease. Mol. Aspects Med. 58, 21–36 (2017).

    PubMed  PubMed Central  Google Scholar 

  150. Gatej, S., Gully, N., Gibson, R. & Bartold, P. M. Probiotics and periodontitis – a literature review. J. Int. Acad. Periodontol. 19, 42–50 (2017).

    PubMed  Google Scholar 

  151. Rosier, B. T., Marsh, P. D. & Mira, A. Resilience of the oral microbiota in health: mechanisms that prevent dysbiosis. J. Dent. Res. 97, 371–380 (2017).

    PubMed  Google Scholar 

  152. Garza, D. R., van Verk, M. C., Huynen, M. A. & Dutilh, B. E. Towards predicting the environmental metabolome from metagenomics with a mechanistic model. Nat. Microbiol. 3, 456–460 (2018).

    CAS  PubMed  Google Scholar 

  153. Surana, N. K. & Kasper, D. L. Moving beyond microbiome-wide associations to causal microbe identification. Nature 552, 244–247 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Brown, S. A. & Whiteley, M. A novel exclusion mechanism for carbon resource partitioning in Aggregatibacter actinomycetemcomitans. J. Bacteriol. 189, 6407–6414 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Brown, S. A. & Whiteley, M. Characterization of the L-lactate dehydrogenase from Aggregatibacter actinomycetemcomitans. PLOS ONE 4, e7864 (2009).

    PubMed  PubMed Central  Google Scholar 

  156. Ramsey, M. M., Rumbaugh, K. P. & Whiteley, M. Metabolite cross-feeding enhances virulence in a model polymicrobial infection. PLOS Pathog. 7, e1002012 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Stacy, A., Fleming, D., Lamont, R. J., Rumbaugh, K. P. & Whiteley, M. A. Commensal bacterium promotes virulence of an opportunistic pathogen via cross-respiration. mBio 7, e00782–16 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Stacy, A., Abraham, N., Jorth, P. & Whiteley, M. Microbial community composition impacts pathogen iron availability during polymicrobial infection. PLOS Pathog. 12, e1006084 (2016).

    PubMed  PubMed Central  Google Scholar 

  159. Chawla, A. et al. Community signalling between Streptococcus gordonii and Porphyromonas gingivalis is controlled by the transcriptional regulator CdhR. Mol. Microbiol. 78, 1510–1522 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Maeda, K. et al. A Porphyromonas gingivalis tyrosine phosphatase is a multifunctional regulator of virulence attributes. Mol. Microbiol. 69, 1153–1164 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Burns, E., Eliyahu, T., Uematsu, S., Akira, S. & Nussbaum, G. TLR2-dependent inflammatory response to Porphyromonas gingivalis is MyD88 independent, whereas MyD88 is required to clear infection. J. Immunol. 184, 1455–1462 (2010).

    CAS  PubMed  Google Scholar 

  162. Brzezinska, A. A., Johnson, J. L., Munafo, D. B., Ellis, B. A. & Catz, S. D. Signalling mechanisms for Toll-like receptor-activated neutrophil exocytosis: key roles for interleukin-1-receptor-associated kinase-4 and phosphatidylinositol 3-kinase but not Toll/IL-1 receptor (TIR) domain-containing adaptor inducing IFNβ (TRIF). Immunology 127, 386–397 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Bainbridge, B. et al. Role of Porphyromonas gingivalis phosphoserine phosphatase enzyme SerB in inflammation, immune response, and induction of alveolar bone resorption in rats. Infect. Immun. 78, 4560–4569 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The work of the authors is supported by US National Institutes of Health grants DE01111, DE012505, DE017921 and DE0130585 (R.J.L.); DE018023, DE025220 and DE025848 (H.K.); and DE015254, DE024153, DE024716, DE026152 and AI068730 (G.H.).

Reviewer information

Nature Reviews Microbiology thanks G. Belibasakis, B. Keijser and other anonymous reviewers for their contributions to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

R.J.L., H.K. and G.H. researched the data for the article, substantially contributed to discussion of content, wrote the article and reviewed and edited the manuscript before submission.

Corresponding authors

Correspondence to Richard J. Lamont, Hyun Koo or George Hajishengallis.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's note

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

Glossary

Subgingival

Relating to the area under the gum margins.

Gingival crevicular fluid

(GCF). A serum exudate that contains immune and inflammatory mediators, along with large numbers of neutrophils recruited along a chemokine gradient.

Extracellular polymeric substances

(EPS). Extracellular biomolecules including exopolysaccharides, fibrous and globular proteins in addition to extracellular enzymes, lipids and nucleic acids.

Gingivitis

Mild and reversible inflammation of the gum often accompanied by bleeding upon toothbrushing. Tissue destruction does not occur. Gingivitis results from an accumulation of the plaque biofilm around the gingival margin and resolves after removal of the plaque.

Acidogenic

Organisms capable of producing acidic metabolites and reducing environmental pH.

Dysbiosis

An imbalanced interaction that can be among bacteria in a community or between the microbiome and the host, and is detrimental to the host. The imbalance can be in the amount and/or the influence of individual microbial species relative to their abundance or influence in health. Alternatively, the imbalance can be caused by a poorly controlled immune response.

Periodontitis

An episodic, slowly progressing inflammatory disease of the periodontal tissues that usually occurs in adults, although aggressive, rapidly progressing forms exist and can occur in adolescents.

Dental caries

A polymicrobial and diet-dependent disease that is characterized by the development of pathogenic biofilms (dental plaque) within which acid production from bacterial metabolism of dietary carbohydrates causes demineralization of the mineralized tooth tissues (enamel, dentin and cementum), eventually leading to the clinical onset of cavitation or tooth decay.

Aciduric

Organisms capable of growth at acidic pH levels that are often toxic to other bacteria.

Polymicrobial synergy

Interactions among organisms that increase microbial fitness in the local environment.

Dentin

Calcified tissue, predominantly hydroxyapatite, forming the bulk of the tooth, which is beneath and is softer (less mineralized with more organic material) than enamel.

Periodontal diseases

A collection of conditions in which poorly controlled inflammatory responses induced by the microbiota cause destruction of the supporting structures of the tooth.

Red complex

The triad of P. gingivalis, T. forsythia and T. denticola — organisms that are often isolated together and were classically considered to be the predominant pathogens in chronic periodontitis.

Gingival crevice

The compartment between the tooth root and the gingival (gum) tissue. The gingival crevice deepens into a periodontal pocket as periodontal disease progresses and tissue is destroyed.

Citrullinate

Post-translational modification of a protein involving deamination of arginine by the enzyme peptidylarginine deiminase PPAD to produce citrulline.

Cross feeding

The utilization of a metabolic by-product of one organism as a nutrient source by another organism.

Nososymbiocity

The potential for a microbial community to contribute to disease; this recognizes the community rather than a single species as the aetiological agent.

Salivary pellicle

A layer of salivary proteins and glycoproteins adsorbed to the enamel surface and to which adhesins of initial colonizers of the oral surface can attach. Pellicle can also contain molecules of microbial origin and those derived from epithelial cells.

Accessory pathogen

Organisms that act synergistically with more pathogenic species (keystone pathogens or pathobionts) to elevate community nososymbiocity. Accessory pathogens can provide an attachment substratum for colonization and metabolic support, and can increase virulence gene expression in other organisms through physical interactions or small-molecule-dependent communication.

Keystone pathogen

Species that exert an influence on their communities that is disproportionate relative to their abundance and therefore form the ‘keystone’ of the community’s structure.

Homeostatic commensals

Species that act to maintain a host–microbiota equilibrium by mitigating the action of more pathogenic species. Mechanisms include reducing the impact of pathogens on host cell signalling pathways or production of metabolites that favour a homeostatic inflammatory response.

Homeostasis

A state of equilibrium or stability in a system that is maintained by adjusting physiological processes to counteract external changes.

Ecological plaque (biofilm) hypothesis

A model encompassing microbiological, biochemical and ecological properties of oral biofilms and their association with disease. The model also accommodates host-derived changes (for example, frequent dietary sugar exposure) that trigger changes in the nososymbiocity of biofilm communities.

Saccharolytic potential

The ability of an organism to metabolize carbohydrates.

Emergent properties

Novel structures, activities, patterns and properties that arise during self-organization into complex systems. In the context of biofilm communities, these include surface adhesion and interbacterial cohesion, spatial organization, physical and social interactions, chemical heterogeneity and increased tolerance to antimicrobials.

Pathobionts

Organisms that are generally benign or commensal within an indigenous community but transition to pathogenic upon the breakdown of host–microbiota homeostasis (for example, as a result of antibiotic treatment, tissue damage, dietary shifts and especially immune deficiencies). These conditions promote pathobiont outgrowth and disrupt the symbiotic microbiota, causing further dysbiosis and inflammation.

Asaccharolytic

A property of organisms incapable of breaking down carbohydrates for energy and thus reliant on the degradation of proteins and the generation of amino acids for metabolic energy and growth.

Inflammophilic

A property of bacteria that thrive on inflammation and utilize inflammatory tissue breakdown products for nutrition.

Hemin

An iron-containing porphyrin compound released from red blood cells; exploited by bacteria to obtain iron for growth.

Localized chemokine paralysis

Precise and targeted suppression of specific chemokines by microbial community participants, superseding the otherwise stimulatory activity of other community inhabitants.

Black queen hypothesis

A theory of reductive evolution to account for co-dependency.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lamont, R.J., Koo, H. & Hajishengallis, G. The oral microbiota: dynamic communities and host interactions. Nat Rev Microbiol 16, 745–759 (2018). https://doi.org/10.1038/s41579-018-0089-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41579-018-0089-x

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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