Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Dec 1.
Published in final edited form as: Mol Oral Microbiol. 2012 Sep 3;27(6):409–419. doi: 10.1111/j.2041-1014.2012.00663.x

Beyond the red complex and into more complexity: the polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology

G Hajishengallis 1, RJ Lamont 2
PMCID: PMC3653317  NIHMSID: NIHMS467095  PMID: 23134607

SUMMARY

Recent advancements in the periodontal research field are consistent with a new model of pathogenesis according to which periodontitis is initiated by a synergistic and dysbiotic microbial community rather than by select ‘periopathogens’, such as the ‘red complex’. In this polymicrobial synergy, different members or specific gene combinations within the community fulfill distinct roles that converge to shape and stabilize a disease-provoking microbiota. One of the core requirements for a potentially pathogenic community to arise involves the capacity of certain species, termed ‘keystone pathogens’, to modulate the host response in ways that impair immune surveillance and tip the balance from homeostasis to dysbiosis. Keystone pathogens also elevate the virulence of the entire microbial community through interactive communication with accessory pathogens. Other important core functions for pathogenicity require the expression of diverse molecules (e.g. appropriate adhesins, cognate receptors, proteolytic enzymes and proinflammatory surface structures/ligands), which in combination act as community virulence factors to nutritionally sustain a heterotypic, compatible and proinflammatory microbial community that elicits a non-resolving and tissue-destructive host response. On the basis of the fundamental concepts underlying this model of periodontal pathogenesis, that is, polymicrobial synergy and dysbiosis, we term it the PSD model.

Keywords: accessory pathogen, dysbiosis, homeostasis, keystone pathogen, periodontitis, polymicrobial infections

THE ‘RED COMPLEX’ AND THE MICROBIAL ETIOLOGY OF PERIODONTITIS

There is an old story of a woman out walking when she encounters a boy with his dog. ‘That’s a nice dog’, says the woman, ‘what’s his name?’ ‘We call him Rover’ the boy replies, ‘but we don’t know his real name’. The notion that individual organisms or even species may have properties that elude our classification systems is also applicable to bacteria, which may have inherent characteristics that are independent of our attempts to label them as ‘pathogens’ or ‘commensals’. Indeed, the distinction between pathogens and commensals is becoming increasingly blurred, especially in diseases that ensue from the action of bacteria that are also present in health and that involve complex host–microbe interactions. Such a case, par excellence, is human periodontal disease. Not surprisingly, therefore, periodontitis has a long and rich history of proposed microbial etiologies ranging from the ‘non-specific plaque hypothesis’ to implication of specific and varied microorganisms, including the oral protozoan Entamoeba gingivalis proposed a century ago (see Table 1) (Socransky & Haffajee, 1994; Wade, 2011).

Table 1.

Milestones and hypotheses in the microbial etiology of periodontitis1

Chronology Etiology or key concepts Remarks References
Late 19th to early 20th century Specific microorganisms (amebae, spirochetes, fusiforms or streptococci) Based on apparent association with periodontal lesions; heavily biased by technique used (wet-mount microscopy, stained smear microscopy, and limited cultural methods) Meyer (1917)
Mid-1920s to 1930s Decline in the interest in bacteria as primary agents in the etiology of periodontitis Disease primarily due to defects in the patient (e.g. trauma from occlusion); bacteria are simply secondary invaders or at best contributors to the disease process Bruske (1928) and Belding & Belding (1933)
Late 1950s ‘Non-specific plaque hypothesis’ and its variant, ‘Mixed anaerobic infections’ Sufficient accumulation of any microorganisms at or below the gingival margin can cause destructive inflammation through the local production of ‘irritants’ Bacteriologically non-specific but biochemically specific mixed anaerobic infections are capable of producing destructive metabolites Schultz-Haudt et al. (1954) and Macdonald et al. (1956)
Late 1970s to early 1980s Microbial shift in periodontitis Dramatic compositional changes to the microbiota in disease as compared with health Slots (1977a,b), Socransky (1977), Tanner et al. (1979) and Moore et al. (1982, 1983)
‘Specific plaque hypothesis’ Periodontitis results, at least in significant part, from the overgrowth of specific pathogenic species Loesche (1979, 1992)
Late 1980s to 1990s ‘Red complex’ Periodontal microbial communities characterized on the basis of a color-coded system reflecting cluster analysis, community ordination, and associated disease severity. A group of ‘red complex’ bacteria (Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia) are strongly associated with each other and with diseased sites. Porphyromonas gingivalis causes periodontitis in nonhuman primates upon its oral implantation Holt et al. (1988) and Socransky et al. (1998)
2003 ‘Ecological catastrophe hypothesis’ Environmental factors drive the selection and enrichment of specific pathogenic bacteria Marsh (2003)
2010 Disruption of periodontal tissue homeostasis Periodontitis fundamentally represents disruption of tissue homeostasis; inflammation is critical but a secondary event. Red complex bacteria are key species for disease, although the polymicrobial nature of periodontitis is acknowledged; commensal bacteria probably induce a protective response Darveau (2010)
2011 ‘Keystone pathogen’ concept Low-abundance keystone species can disrupt tissue homeostasis through quantitative and qualitative changes to the commensal microbiota (resulting, at least in great part, from host modulation by the keystone pathogen). Inflammatory bone loss is mediated by the altered microbiota Hajishengallis et al. (2011)
2012 ‘Polymicrobial synergy and dysbiosis’ (PSD) Periodontitis is initiated by a synergistic and dysbiotic microbiota, within which different members, or specific gene combinations thereof, fulfill distinct roles that converge to shape and stabilize a disease- provoking microbiota. Combines the concepts of ‘disrupted homeostasis’ and ‘keystone pathogen’ but questions the primary importance of the red complex This paper
1

For a detailed historic description of the various hypotheses, the reader is referred to Socransky & Haffajee (1994).

Much of our current appreciation of the microbial etiology of periodontitis derives from detailed cultural characterization of the periodontal microbiota in the late 1970s and early 1980s. These studies revealed dramatic compositional changes to the microbiota in disease as compared with health (Slots, 1977a,b; Socransky, 1977; Tanner et al., 1979; Moore et al., 1982, 1983). One plausible interpretation was that these key findings pointed to bacterial specificity in the etiology of periodontitis, in that the disease-associated microbiota contained novel pathogenic species that were either absent or hardly detectable in health. The quest to identify specific periodontal pathogens led to significant progress, including the identification of a number of candidates and the characterization of putative virulence factors thereof (Socransky et al., 1998; Holt & Ebersole, 2005). Using whole genomic DNA probes and checkerboard DNA–DNA hybridization, Socransky and colleagues characterized periodontal microbial communities on the basis of a color-coded system that reflected cluster analysis, community ordination and associated disease severity (Socransky et al., 1998). Foremost among these groups was the so-called ‘red complex’, a group of three species including Porphyromonas gingivalis, Treponema denticola and Tannerella forsythia, the detection of which was strongly associated with each other and with diseased sites (Socransky et al., 1998).

Partly because P. gingivalis was the easiest of the red complex bacteria to grow and genetically manipulate, it became the most widely studied periodontal bacterium. Virulence of the organism was attributed to an array of molecules, including colonization factors (fimbriae and hemagglutinins) and functionally versatile proteolytic enzymes (gingipains) (Lamont & Jenkinson, 1998). In a landmark study, P. gingivalis was shown to cause periodontitis in non-human primates upon its oral implantation, which was interpreted as evidence for a specific microbial etiology in periodontitis (Holt et al., 1988). More recently, longitudinal human studies demonstrated that the progression of chronic periodontitis can be predicted by the levels of P. gingivalis and T. denticola in subgingival plaque (Byrne et al., 2009). The implication of specific periodontal bacteria as putative etiologic agents provided rationale and impetus for experimental vaccination against periodontal disease (Persson, 2005). Additional work focused primarily on understanding cellular and molecular pathogenic mechanisms of the red complex and certain other suspected pathogens (Cutler et al., 1995; Sela, 2001; Holt & Ebersole, 2005; Sharma, 2010). As a note of caution, however, most of this research was performed in the context of a conventional host–pathogen interaction, as exemplified by diseases with defined infective etiology, and the extent to which these virulence factors are operational in vivo remains to be determined.

The convenience and appeal of the concept of red, and other color-coded, complex(es) led to widespread adoption up to the present day. However, as molecular-based approaches to microbe detection became increasingly facile, and as studies using culture-independent methodology for analysis of the periodontal microbiota became abundant, two newer concepts emerged. First, red complex organisms such as P. gingivalis can be found in the absence of disease, further argument against the organisms as classical exogenous pathogens (Ximenez-Fyvie et al., 2000a, b; Mayanagi et al., 2004; Diaz et al., 2006). Second, the periodontal microbiota is more heterogeneous and diverse than previously thought (Dewhirst et al., 2010; Curtis et al., 2011b; Griffen et al., 2011), with over 700 organisms recognized as possible components, of which around 200 can be present in any one individual and about 50 are present at any one site (Aas et al., 2005). Many of these newly recognized organisms show as good or better a correlation with disease as the classical red complex (Kumar et al., 2005, 2006; Griffen et al., 2012). Novel disease-associated species include the gram-positive Filifactor alocis and Peptostreptococcus stomatis and other species from the genera Prevotella, Megasphaera, Selenomonas, Desulfobulbus, Dialister and Synergistetes (Paster et al., 2001; Kumar et al., 2003, 2005; Dewhirst et al., 2010; Griffen et al., 2011, 2012). Moreover, contrary to the ‘dogma’ of gram-negative bacterial dominance in periodontitis, gram-positive anaerobic species exhibit a significant increase in deep diseased sites relative to healthy sites, and can be detected in greater abundance than gram-negative species in some studies (Kumar et al., 2005). Many of these species are as yet to be cultivated, however, study of the more tractable organisms is underway and F. alocis, for example, has the potential to withstand oxidative stress (Aruni et al., 2011) and to induce strong proinflammatory responses (Moffatt et al., 2011).

Although the pathogenic credentials of most of these newly recognized organisms remain to be established, it is evident that the concept of a three-species red complex as representing the primary etiologic unit in periodontitis requires refinement. Not only are there more potential pathogens to consider, but these organisms can exhibit polymicrobial synergy (discussed in more detail below). Furthermore, a disease-associated shift in the composition of the periodontal microbiota could have alternative explanations. The findings could be interpreted to imply that periodontitis is caused by a dysbiotic change in the periodontal microbiota, that is, a change in the relative abundance of individual members of the microbiota relative to their abundance in health. The altered microbiota could in turn lead to changes in the host–microbe crosstalk sufficient to initiate chronic, non-resolving inflammatory disease. Disease can then be seen in terms of an ecological catastrophe as defined by Marsh (2003). In the following text we shall attempt to integrate these concepts into a new model of periodontitis that accommodates the data derived from sequencing projects, the interactions that occur among organisms and their differential pathogenicity (Fig. 1).

Figure 1.

Figure 1

The polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology. The gingival crevice is colonized by a diverse microbiota, and compatible organisms assemble into heterotypic communities. These communities are in equilibrium with the host. Although they are proinflammatory and can produce toxic products such as proteases, overgrowth and overt pathogenicity are controlled by the host. The microbial constituents of the communities can vary over time, and from person to person and site to site. Colonization by keystone pathogens such as Porphyromonas gingivalis elevates the virulence of the entire community following interactive communication with accessory pathogens such as mitis group streptococci. Host immune surveillance is impaired and the dysbiotic community increases in number eventually disrupting tissue homeostasis and causing destruction of periodontal tissues.

THE PORPHYROMONAS GINGIVALIS PARADOX

To understand basic principles of periodontal microbial pathogenicity, it is instructive to first consider the case of P. gingivalis. This organism can cause alveolar bone loss in animal models of periodontal disease, although, as we shall see, pathogenicity is context dependent. The low abundance of P. gingivalis in periodontitis-associated dental plaque was noted in early bacteriological studies (Moore et al., 1982). This, together with findings that P. gingivalis is not a potent inducer of inflammation by itself, presented an apparent paradox that could not be readily reconciled with a proactive role in an inflammatory disease. For instance, P. gingivalis expresses an atypical lipopolysaccharide with a 4-acyl monophosphorylated lipid A moiety that can potently antagonize Toll-like receptor 4, unlike the highly proinflammatory lipopolysaccharides of most other gram-negative bacteria (Coats et al., 2009; Darveau, 2010). Porphyromonas gingivalis can invade and survive within gingival epithelial cells; however, rather than causing host cell damage, internalized bacteria induce an anti-apoptotic, prosurvival phenotype in host cells (Mao et al., 2007; Kuboniwa et al., 2008) and downregulate expression of bacterial proinflammatory components such as fimbrial proteins and proteases (Xia et al., 2007; Hendrickson et al., 2009). Moreover, in contrast to other periodontal bacteria that stimulate interleukin-8 release from gingival epithelial cells, P. gingivalis actually inhibits the production of this proinflammatory chemokine by means of a secreted serine phosphatase (SerB) (Darveau et al., 1998; Hasegawa et al., 2008). The release of interleukin-8 by the junctional gingival epithelium is regarded as an important feature of the healthy periodontium because it generates a gradient for neutrophil recruitment into the gingival crevice (Tonetti et al., 1998; Darveau, 2010).

A recent study in mice has offered an explanation for this apparent paradox and proposed a disease model that is consistent with the alternative interpretation of the periodontitis-associated microbial shift. This study showed that P. gingivalis can impair innate immunity in ways that enhance the growth and alter the composition of the periodontal microbiota (Hajishengallis et al., 2011). Specifically, P. gingivalis, at low colonization levels (<0.01% of the total microbiota), remodeled a symbiotic community into a dysbiotic state that triggered inflammatory bone loss. Porphyromonas gingivalis failed to cause periodontitis in the absence of commensal bacteria, i.e. in germfree mice, despite its ability to colonize this host (Hajishengallis et al., 2011). The capacity of P. gingivalis to exert a community-wide impact that tipped the balance towards dysbiosis, while being a quantitatively minor constituent of this microbial community, has prompted its characterization as a keystone pathogen, in analogy to the crucial role of a single keystone in an arch (Hajishengallis et al., 2011).

The keystone effects of P. gingivalis are likely exerted via both host modulation and bacterial synergy (discussed below). In terms of host modulation, P. gingivalis can transiently inhibit the induction of gingival interleukin-8-like chemokines, which could delay the recruitment of neutrophils and, thereby, facilitate its initial colonization and promote the fitness of other organisms (Hajishengallis et al., 2011). However, the ability of P. gingivalis to persist in the periodontium appears to depend on the complement C5 convertase-like activity of its gingipains and the instigation of a subversive crosstalk between C5a receptor (C5aR) and Toll-like receptor 2 (Hajishengallis et al., 2011), earlier shown to impede the killing capacity of leukocytes (Wang et al., 2010; Liang et al., 2011). Consistent with this, P. gingivalis fails to cause dysbiosis and periodontitis in mice lacking C5aR.

Although established in the mouse model, the keystone pathogen concept is consistent with observations in other animal models and in humans (reviewed by Darveau et al., 2012). Briefly, in rabbits, P. gingivalis causes a shift to a more anaerobic microbiota and an overall increase in the bacterial load of the dental biofilm (Hasturk et al., 2007). In non-human primates (Macaca fascicularis), where P. gingivalis is a natural inhabitant of the periodontal biofilm, a gingipain-based vaccine causes a reduction in P. gingivalis numbers and in the total subgingival bacterial load (Page et al., 2007), suggesting that the presence of P. gingivalis benefits the entire community. A significant increase in the total microbial load is also observed in human periodontitis compared with health (Socransky & Haffajee, 1994; Darveau et al., 1997); however, it has not been yet addressed whether this is causally linked to the presence of P. gingivalis or other bacteria acting as keystone pathogens. Nevertheless, consistent with a keystone pathogen role, P. gingivalis is a quantitatively minor constituent of periodontitis-associated biofilms (Moore et al., 1982; Doungudomdacha et al., 2000; Kumar et al., 2006), despite its high prevalence and association with progressive bone loss in periodontitis patients (Moore et al., 1991; Chaves et al., 2000).

The subversion of recruited leukocytes by P. gingivalis likely facilitates uncontrolled growth of other species in the same biofilm. This disruption of tissue homeostasis, in turn may allow other community members to trigger destructive inflammation. Such a host response, apart from its adverse impact on the integrity of the periodontium, leads to tissue breakdown products that serve the nutritional needs of the community at large (Gaffen & Hajishengallis, 2008). Consequently, the transition to a disease-provoking microbiota may be stabilized and, conversely, those species that cannot thrive under inflammatory environmental conditions, or for which host inflammation is detrimental, may be outcompeted or eliminated. It should be clarified, however, that disruption of tissue homeostasis followed by dysbiosis can also be caused by host regulatory defects in the absence of a keystone pathogen. For instance, genetic deficiency of Del-1, an endothelial cell-derived protein that regulates neutrophil recruitment, causes inflammatory bone loss and both quantitative and compositional alterations to the murine commensal microbiota in the absence of P. gingivalis (Eskan et al., 2012). This implies that, at least in principle, periodontitis could be initiated in the absence of bacteria belonging to the ‘red complex’ or acting as keystone pathogens.

As mentioned, P. gingivalis can also be detected, albeit less frequently, in periodontally healthy individuals (Haffajee et al., 1998; Ximenez-Fyvie et al., 2000a,b; Mayanagi et al., 2004; Diaz et al., 2006), which begs the question why its presence does not always lead to periodontitis. In other words, why is the organism a pathogen in some instances and a commensal in others. Several not mutually exclusive explanations may involve variability in the status of the host or the bacterium. There may be individuals who can either resist or tolerate the conversion of the periodontal microbiota from a symbiotic to a dysbiotic state, by virtue of their intrinsic immuno-inflammatory status (e.g. hyporesponsive or lack-of-function polymorphisms that attenuate inflammation or microbial immune subversion). Moreover, strain and virulence diversity within the population structure of P. gingivalis may affect its capacity to act as a keystone pathogen. Additionally, local environmental changes may influence the capacity of P. gingivalis to disrupt host–bacteria homeostasis. In this regard, the production of P. gingivalis gingipains and fimbriae is regulated by local environmental conditions (Xie et al., 1997, 2000; Curtis et al., 2001), which can therefore influence the capacity of this organism to modulate complement activity and subvert leukocytes (Hajishengallis et al., 2011). In the same vein, the structure of P. gingivalis lipopolysaccharide is regulated by temperature fluctuations and hemin concentrations (Al-Qutub et al., 2006; Curtis et al., 2011a). Therefore, at least theoretically, there are conditions under which P. gingivalis (or any other single bacterium whether from the ‘red complex’ or not) may not act as a pathogen but rather behave as a commensal.

THE REAL CULPRIT: A SYNERGISTIC MICROBIAL COMMUNITY

It is becoming evident, therefore, that the virulence of periodontal pathogens such as P. gingivalis acquires importance only in the context of a synergistic microbial community, which is required for the expression of pathogenicity. This model is consistent with the participation of both gram-negative and gram-positive bacteria in periodontal pathogenesis, as long as they can provoke or tolerate inflammation, or provide other useful service to the community. Mixed microbial communities provide opportunities for competitive and co-operative interspecies interactions, and such interactions shape the nature and function of the entire assemblage (Jenkinson & Lamont, 2005; Hansen et al., 2007). Furthermore, interspecies signaling within communities provides the opportunity to collectively regulate activities including gene expression, nutrient acquisition and DNA exchange. In this manner communities of bacteria can exhibit polymicrobial synergy, defined as an increase in the ability of a bacterium to colonize/persist or elevate disease symptoms when in the presence of other bacteria (Fig. 1). Indeed, there are numerous examples of mixed infections with oral organisms exhibiting increased pathogenicity compared with either organism alone in animal models (Kesavalu et al., 2007; Orth et al., 2011; Settem et al., 2012). To some degree this will reflect nutritional cross-feeding that enhances the growth rate of compatible organisms in combination (Grenier, 1992; Nilius et al., 1993), and, moreover, closely associated organisms can compile a communal suite of enzymes to sequentially degrade complex substrates into constituents that can be metabolized by individual members of the community. However, as mentioned above, P. gingivalis can have a community-wide pathogenic influence on the microbiota in animals, at least in part via host modulation (Hajishengallis et al., 2011). Moreover, the introduction of P. gingivalis into a healthy multispecies biofilm alters the pattern of microbial community gene expression (Frias-Lopez & Duran-Pinedo, 2012), suggesting that this keystone pathogen could additionally modulate the commensal oral microbiota through direct, host-independent effects. Conversely, when in a community with Streptococcus gordonii and Fusobacterium nucleatum, P. gingivalis differentially expresses around 500 proteins, indicating that the organism undergoes profound phenotypic changes in response to common oral species (Kuboniwa et al., 2009). Polymicrobial synergy among periodontal pathogens therefore extends beyond growth rate effects and involves interspecies signaling and response interactions.

The molecular mechanisms that underlie the polymicrobial synergy of oral bacteria reveal hierarchical, temporally distinct communication systems whereby organisms can integrate multiple signals of various forms and function during the process of heterotypic community development. Oral bacteria can communicate through contact-dependent systems, and short range diffusible signals, including metabolic products and autoinducer-2 (Kolenbrander et al., 2002, 2010), all of which can influence pathogenic potential (Ramsey & Whiteley, 2009; Ramsey et al., 2011). For example, P. gingivalis displays numerous specific interactions with oral streptococci, suggesting that interspecies co-operation has evolved to enhance the fitness of these organisms. Specific adherence of P. gingivalis to S. gordonii is mediated by the minor fimbrial subunit protein (Mfa1) which interacts with discrete domains on the streptococcal surface proteins SspA/B (Demuth et al., 2001; Park et al., 2005; Daep et al., 2008). Coadhesion between the organisms initiates a signal transduction pathway within P. gingivalis based on protein tyrosine (de)phosphorylation that converges on regulation of genes encoding community effectors such as Mfa1 and LuxS (Maeda et al., 2008; Chawla et al., 2010). The level of community accumulation is tightly controlled possibly to maintain an optimal surface area : volume ratio of the dual species microcolony. The P. gingivalisS. gordonii communities exhibit mutualistic growth (Periasamy & Kolenbrander, 2009) and induce greater alveolar bone loss in mice compared with either species alone (Daep et al., 2011). Moreover, interference with P. gingivalisS. gordonii binding by blocking the Mfa1–SspA/B interaction abrogates bone loss, demonstrating the importance of coadhesion and subsequent signaling in the development of a pathogenic community (Daep et al., 2011).

Communication relevant to virulence also occurs between the oral streptococci and Aggregatibacter actinomycetemcomitans which is closely associated with localized aggressive periodontitis. Aggregatibacter actinomycetemcomitans displays resource partitioning to favor carbon sources such as lactate generated by streptococcal metabolism (Brown & Whiteley, 2007). Additionally, A. actinomycetemcomitans can sense hydrogen peroxide, a metabolic byproduct of streptococci, and respond by upregulation of genes that enhance resistance to killing by neutrophils (Ramsey & Whiteley, 2009). The relevance of these interactions is demonstrated by co-culture of A. actinomycetemcomitans with S. gordonii, which enhances the pathogenicity of A. actinomycetemcomitans in a mouse abscess model (Ramsey et al., 2011).

It is interesting to note that both of the above examples involve the mitis group oral streptococci, traditionally seen as commensals in the oral cavity. However, although the relative amounts decrease, oral streptococci still comprise a significant portion of the microbial population of subgingival plaque (Moore & Moore, 1994; Kroes et al., 1999; Paster et al., 2001; Kumar et al., 2005; Quirynen et al., 2005; Colombo et al., 2009), and indeed some studies have shown a higher proportion of S. gordonii in the subgingival biofilm of periodontitis subjects compared with healthy individuals (Abiko et al., 2010). It has been proposed, therefore, that organisms such as the mitis group streptococci be considered ‘accessory pathogens’, organisms whose pathogenic potential only becomes evident in the context of a heterotypic microbial community (Whitmore & Lamont, 2011). Periodontal pathogenicity would therefore appear to require significant interspecies co-operation.

THE PSD MODEL

The recent advancements discussed above are consistent with a new model of pathogenesis according to which periodontitis is initiated by a broadly-based dysbiotic, synergistic, microbiota (Fig. 1), as opposed to the traditional view of a conventional infectious disease caused by a single or even several select periopathogens, such as the ‘red complex’. The situation can be likened, perhaps, to that of a crew team. All the oars need to be manned but the identities of individual crew members, provided they are capable of rowing, are not important for forward progression. Some functions, however, such as cox, are more important for coordinating characteristics such as direction and speed. In the periodontal ecosystem diverse bacteria (or specific combinations of genes within the community) may be able to fulfill distinct roles that converge to form and stabilize a disease-provoking microbiota. Hence, there will be a number of core requirements for a potentially pathogenic community to arise. (i) Bacterial constituents will express the relevant adhesins and receptors to allow assembly of a heterotypic community. (ii) Individual members of the community will be physiologically compatible or at least non-antagonistic. (iii) The combined activities of the community will resist the host innate and acquired immune responses and contribute to tissue inflammation through, for example, proteolytic activity and cytokine induction. On the basis of the diversity of organisms associated with periodontal lesions, it is likely that potentially pathogenic communities occur frequently and that there are a variety of organisms that can contribute the genes necessary for these conditions to be satisfied. In that regard, it is relevant that dental plaque obtained from healthy sites shares the capacity of disease-associated plaque to induce strong inflammatory responses through Toll-like receptor activation (Yoshioka et al., 2008). However, for pathogenic potential to be realized, the activities of a keystone species such as P. gingivalis are required. These organisms engage in two-way communication with the community inhabitants, in particular the accessory pathogens, to both disrupt host immune surveillance and elevate the pathogenicity of the entire group. This more specialized dysbiotic role will be restricted to fewer organisms. The identification of keystone and accessory pathogens from the catalog of organisms generated by microbiome projects will present the next major challenge in periodontal disease research and, perhaps, in other inflammatory diseases with a complex polymicrobial etiology (Hajishengallis et al., 2012). Moreover, an in-depth understanding of periodontal pathogenesis on the basis of the PSD model may offer new targets for therapeutic intervention.

Acknowledgments

The authors’ research is supported by DE15254, DE17138, DE18292, DE21580, DE21685 (GH) and DE11111, DE12505, DE16690, DE17921, DE22867 (RJL).

References

  1. Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining the normal bacterial flora of the oral cavity. J Clin Microbiol. 2005;43:5721–5732. doi: 10.1128/JCM.43.11.5721-5732.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Abiko Y, Sato T, Mayanagi G, Takahashi N. Profiling of subgingival plaque biofilm microflora from periodontally healthy subjects and from subjects with periodontitis using quantitative real-time PCR. J Periodontal Res. 2010;45:389–395. doi: 10.1111/j.1600-0765.2009.01250.x. [DOI] [PubMed] [Google Scholar]
  3. Al-Qutub MN, Braham PH, Karimi-Naser LM, Liu X, Genco CA, Darveau RP. Hemin-dependent modulation of the lipid A structure of Porphyromonas gingivalis lipopolysaccharide. Infect Immun. 2006;74:4474–4485. doi: 10.1128/IAI.01924-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Aruni AW, Roy F, Fletcher HM. Filifactor alocis has virulence attributes that can enhance its persistence under oxidative stress conditions and mediate invasion of epithelial cells by Porphyromonas gingivalis. Infect Immun. 2011;79:3872–3886. doi: 10.1128/IAI.05631-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Belding LJ, Belding PH. An evaluation of various theories treating on the etiology of periodontoclasis. Dent Cosmos. 1933;75:140–145. [Google Scholar]
  6. Brown SA, Whiteley M. A novel exclusion mechanism for carbon resource partitioning in Aggregatibacter actinomycetemcomitans. J Bacteriol. 2007;189:6407– 6414. doi: 10.1128/JB.00554-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bruske JS. Periodontoclasia – a working hypothesis. J Am Dent Assoc. 1928;15:723–728. [Google Scholar]
  8. Byrne SJ, Dashper SG, Darby IB, Adams GG, Hoffmann B, Reynolds EC. Progression of chronic periodontitis can be predicted by the levels of Porphyromonas gingivalis and Treponema denticola in subgingival plaque. Oral Microbiol Immunol. 2009;24:469–477. doi: 10.1111/j.1399-302X.2009.00544.x. [DOI] [PubMed] [Google Scholar]
  9. Chaves ES, Jeffcoat MK, Ryerson CC, Snyder B. Persistent bacterial colonization of Porphyromonas gingivalis, Prevotella intermedia, and Actinobacillus actinomycetemcomitans in periodontitis and its association with alveolar bone loss after 6 months of therapy. J Clin Periodontol. 2000;27:897–903. doi: 10.1034/j.1600-051x.2000.027012897.x. [DOI] [PubMed] [Google Scholar]
  10. Chawla A, Hirano T, Bainbridge BW, Demuth DR, Xie H, Lamont RJ. Community signalling between Streptococcus gordonii and Porphyromonas gingivalis is controlled by the transcriptional regulator CdhR. Mol Microbiol. 2010;78:1510–1522. doi: 10.1111/j.1365-2958.2010.07420.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Coats SR, Jones JW, Do CT, et al. Human Toll-like receptor 4 responses to P. gingivalis are regulated by lipid A 1- and 4′-phosphatase activities. Cell Microbiol. 2009;11:1587–1599. doi: 10.1111/j.1462-5822.2009.01349.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Colombo AP, Boches SK, Cotton SL, et al. Comparisons of subgingival microbial profiles of refractory periodontitis, severe periodontitis, and periodontal health using the human oral microbe identification microarray. J Periodontol. 2009;80:1421–1432. doi: 10.1902/jop.2009.090185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Curtis MA, Aduse-Opoku J, Rangarajan M. Cysteine proteases of Porphyromonas gingivalis. Crit Rev Oral Biol Med. 2001;12:192–216. doi: 10.1177/10454411010120030101. [DOI] [PubMed] [Google Scholar]
  14. Curtis MA, Percival RS, Devine D, et al. Temperature dependent modulation of Porphyromonas gingivalis lipid A structure and interaction with the innate host defences. Infect Immun. 2011a;79:1187–1193. doi: 10.1128/IAI.00900-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Curtis MA, Zenobia C, Darveau RP. The relationship of the oral microbiotia to periodontal health and disease. Cell Host Microbe. 2011b;10:302–306. doi: 10.1016/j.chom.2011.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cutler CW, Kalmar JR, Genco CA. Pathogenic strategies of the oral anaerobe, Porphyromonas gingivalis. Trends Microbiol. 1995;3:45–51. doi: 10.1016/s0966-842x(00)88874-5. [DOI] [PubMed] [Google Scholar]
  17. Daep CA, Lamont RJ, Demuth DR. Interaction of Porphyromonas gingivalis with oral streptococci requires a motif that resembles the eukaryotic nuclear receptor box protein–protein interaction domain. Infect Immun. 2008;76:3273–3280. doi: 10.1128/IAI.00366-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Daep CA, Novak EA, Lamont RJ, Demuth DR. Structural dissection and in vivo effectiveness of a peptide inhibitor of Porphyromonas gingivalis adherence to Streptococcus gordonii. Infect Immun. 2011;79:67–74. doi: 10.1128/IAI.00361-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Darveau RP. Periodontitis: a polymicrobial disruption of host homeostasis. Nat Rev Microbiol. 2010;8:481–490. doi: 10.1038/nrmicro2337. [DOI] [PubMed] [Google Scholar]
  20. Darveau RP, Tanner A, Page RC. The microbial challenge in periodontitis. Periodontol 2000. 1997;14:12–32. doi: 10.1111/j.1600-0757.1997.tb00190.x. [DOI] [PubMed] [Google Scholar]
  21. Darveau RP, Belton CM, Reife RA, Lamont RJ. Local chemokine paralysis, a novel pathogenic mechanism for Porphyromonas gingivalis. Infect Immun. 1998;66:1660–1665. doi: 10.1128/iai.66.4.1660-1665.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Darveau RP, Hajishengallis G, Curtis MA. Porphyromonas gingivalis as a community activist for disease. J Dent Res. 2012;91 doi: 10.1177/0022034512453589. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Demuth DR, Irvine DC, Costerton JW, Cook GS, Lamont RJ. Discrete protein determinant directs the species-specific adherence of Porphyromonas gingivalis to oral streptococci. Infect Immun. 2001;69:5736–5741. doi: 10.1128/IAI.69.9.5736-5741.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Dewhirst FE, Chen T, Izard J, et al. The human oral microbiome. J Bacteriol. 2010;192:5002–5017. doi: 10.1128/JB.00542-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Diaz PI, Chalmers NI, Rickard AH, et al. Molecular characterization of subject-specific oral microflora during initial colonization of enamel. Appl Environ Microbiol. 2006;72:2837–2848. doi: 10.1128/AEM.72.4.2837-2848.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Doungudomdacha S, Rawlinson A, Douglas CW. Enumeration of Porphyromonas gingivalis, Prevotella intermedia and Actinobacillus actinomycetemcomitans in subgingival plaque samples by a quantitative-competitive PCR method. J Med Microbiol. 2000;49:861– 874. doi: 10.1099/0022-1317-49-10-861. [DOI] [PubMed] [Google Scholar]
  27. Eskan MA, Jotwani R, Abe T, et al. The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss. Nat Immunol. 2012;13:465–473. doi: 10.1038/ni.2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Frias-Lopez J, Duran-Pinedo A. Effect of periodontal pathogens on the metatranscriptome of a healthy multispecies biofilm model. J Bacteriol. 2012;194:2082–2095. doi: 10.1128/JB.06328-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Gaffen SL, Hajishengallis G. A new inflammatory cytokine on the block: re-thinking periodontal disease and the Th1/Th2 paradigm in the context of Th17 cells and IL-17. J Dent Res. 2008;87:817–828. doi: 10.1177/154405910808700908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Grenier D. Nutritional interactions between two suspected periodontopathogens, Treponema denticola and Porphyromonas gingivalis. Infect Immun. 1992;60:5298–5301. doi: 10.1128/iai.60.12.5298-5301.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Griffen AL, Beall CJ, Firestone ND, et al. CORE: a phylogenetically-curated 16S rDNA database of the core oral microbiome. PLoS ONE. 2011;6:e19051. doi: 10.1371/journal.pone.0019051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Griffen AL, Beall CJ, Campbell JH, et al. Distinct and complex bacterial profiles in human periodontitis and health revealed by 16S pyrosequencing. ISME J. 2012;6:1176–1185. doi: 10.1038/ismej.2011.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Haffajee AD, Cugini MA, Tanner A, et al. Subgingival microbiota in healthy, well-maintained elder and periodontitis subjects. J Clin Periodontol. 1998;25:346–353. doi: 10.1111/j.1600-051x.1998.tb02454.x. [DOI] [PubMed] [Google Scholar]
  34. Hajishengallis G, Liang S, Payne MA, et al. Low-abundance biofilm species orchestrates inflammatory periodontal disease through the commensal microbiota and complement. Cell Host Microbe. 2011;10:497–506. doi: 10.1016/j.chom.2011.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hajishengallis G, Darveau RP, Curtis MA. The keystone pathogen hypothesis. Nat Rev Microbiol. 2012;10 doi: 10.1038/nrmicro2873. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Hansen SK, Rainey PB, Haagensen JA, Molin S. Evolution of species interactions in a biofilm community. Nature. 2007;445:533–536. doi: 10.1038/nature05514. [DOI] [PubMed] [Google Scholar]
  37. Hasegawa Y, Tribble GD, Baker HV, Mans JJ, Handfield M, Lamont RJ. Role of Porphyromonas gingivalis SerB in gingival epithelial cell cytoskeletal remodeling and cytokine production. Infect Immun. 2008;76:2420–2427. doi: 10.1128/IAI.00156-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Hasturk H, Kantarci A, Goguet-Surmenian E, et al. Resolvin E1 regulates inflammation at the cellular and tissue level and restores tissue homeostasis in vivo. J Immunol. 2007;179:7021–7029. doi: 10.4049/jimmunol.179.10.7021. [DOI] [PubMed] [Google Scholar]
  39. Hendrickson EL, Xia Q, Wang T, Lamont RJ, Hackett M. Pathway analysis for intracellular Porphyromonas gingivalis using a strain ATCC 33277 specific database. BMC Microbiol. 2009;9:185. doi: 10.1186/1471-2180-9-185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Holt SC, Ebersole JL. Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia: the “red complex”, a prototype polybacterial pathogenic consortium in periodontitis. Periodontol 2000. 2005;38:72–122. doi: 10.1111/j.1600-0757.2005.00113.x. [DOI] [PubMed] [Google Scholar]
  41. Holt SC, Ebersole J, Felton J, Brunsvold M, Kornman KS. Implantation of Bacteroides gingivalis in nonhuman primates initiates progression of periodontitis. Science. 1988;239:55–57. doi: 10.1126/science.3336774. [DOI] [PubMed] [Google Scholar]
  42. Jenkinson HF, Lamont RJ. Oral microbial communities in sickness and in health. Trends Microbiol. 2005;13:589–595. doi: 10.1016/j.tim.2005.09.006. [DOI] [PubMed] [Google Scholar]
  43. Kesavalu L, Sathishkumar S, Bakthavatchalu V, et al. Rat model of polymicrobial infection, immunity, and alveolar bone resorption in periodontal disease. Infect Immun. 2007;75:1704–1712. doi: 10.1128/IAI.00733-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Kolenbrander PE, Andersen RN, Blehert DS, Egland PG, Foster JS, Palmer RJ., Jr Communication among oral bacteria. Microbiol Mol Biol Rev. 2002;66:486–505. doi: 10.1128/MMBR.66.3.486-505.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Kolenbrander PE, Palmer RJ, Jr, Periasamy S, Jakubovics NS. Oral multispecies biofilm development and the key role of cell-cell distance. Nat Rev Microbiol. 2010;8:471–480. doi: 10.1038/nrmicro2381. [DOI] [PubMed] [Google Scholar]
  46. Kroes I, Lepp PW, Relman DA. Bacterial diversity within the human subgingival crevice. Proc Natl Acad Sci USA. 1999;96:14547–14552. doi: 10.1073/pnas.96.25.14547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Kuboniwa M, Hasegawa Y, Mao S, et al. P. gingivalis accelerates gingival epithelial cell progression through the cell cycle. Microbes Infect. 2008;10:122–128. doi: 10.1016/j.micinf.2007.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Kuboniwa M, Hendrickson EL, Xia Q, et al. Proteomics of Porphyromonas gingivalis within a model oral microbial community. BMC Microbiol. 2009;9:98. doi: 10.1186/1471-2180-9-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Kumar PS, Griffen AL, Barton JA, Paster BJ, Moeschberger ML, Leys EJ. New bacterial species associated with chronic periodontitis. J Dent Res. 2003;82:338–344. doi: 10.1177/154405910308200503. [DOI] [PubMed] [Google Scholar]
  50. Kumar PS, Griffen AL, Moeschberger ML, Leys EJ. Identification of candidate periodontal pathogens and beneficial species by quantitative 16S clonal analysis. J Clin Microbiol. 2005;43:3944–3955. doi: 10.1128/JCM.43.8.3944-3955.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Kumar PS, Leys EJ, Bryk JM, Martinez FJ, Moeschberger ML, Griffen AL. Changes in periodontal health status are associated with bacterial community shifts as assessed by quantitative 16S cloning and sequencing. J Clin Microbiol. 2006;44:3665–3673. doi: 10.1128/JCM.00317-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Lamont RJ, Jenkinson HF. Life below the gum line: pathogenic mechanisms of Porphyromonas gingivalis. Microbiol Mol Biol Rev. 1998;62:1244–1263. doi: 10.1128/mmbr.62.4.1244-1263.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Liang S, Krauss JL, Domon H, et al. The C5a receptor impairs IL-12-dependent clearance of Porphyromonas gingivalis and is required for induction of periodontal bone loss. J Immunol. 2011;186:869–877. doi: 10.4049/jimmunol.1003252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Loesche WJ. Clinical and microbiological aspects of chemotherapeutic agents used according to the specific plaque hypothesis. J Dent Res. 1979;58:2404–2412. doi: 10.1177/00220345790580120905. [DOI] [PubMed] [Google Scholar]
  55. Loesche WJ. The specific plaque hypothesis and the antimicrobial treatment of periodontal disease. Dent Update. 1992;19:68, 70–62, 74. [PubMed] [Google Scholar]
  56. Macdonald JB, Sutton RM, Knoll ML, Madlener EM, Grainger RM. The pathogenic components of an experimental fusospirochetal infection. J Infect Dis. 1956;98:15–20. doi: 10.1093/infdis/98.1.15. [DOI] [PubMed] [Google Scholar]
  57. Maeda K, Tribble GD, Tucker CM, et al. A Porphyromonas gingivalis tyrosine phosphatase is a multifunctional regulator of virulence attributes. Mol Microbiol. 2008;69:1153–1164. doi: 10.1111/j.1365-2958.2008.06338.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Mao S, Park Y, Hasegawa Y, et al. Intrinsic apoptotic pathways of gingival epithelial cells modulated by Porphyromonas gingivalis. Cell Microbiol. 2007;9:1997– 2007. doi: 10.1111/j.1462-5822.2007.00931.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Marsh PD. Are dental diseases examples of ecological catastrophes? Microbiology. 2003;149:279–294. doi: 10.1099/mic.0.26082-0. [DOI] [PubMed] [Google Scholar]
  60. Mayanagi G, Sato T, Shimauchi H, Takahashi N. Detection frequency of periodontitis-associated bacteria by polymerase chain reaction in subgingival and supragingival plaque of periodontitis and healthy subjects. Oral Microbiol Immunol. 2004;19:379–385. doi: 10.1111/j.1399-302x.2004.00172.x. [DOI] [PubMed] [Google Scholar]
  61. Meyer KF. The present status of dental bacteriology. J Am Dent Assoc. 1917;4:966–996. [Google Scholar]
  62. Moffatt CE, Whitmore SE, Griffen AL, Leys EJ, Lamont RJ. Filifactor alocis interactions with gingival epithelial cells. Mol Oral Microbiol. 2011;26:365–373. doi: 10.1111/j.2041-1014.2011.00624.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Moore WE, Moore LV. The bacteria of periodontal diseases. Periodontol 2000. 1994;5:66–77. doi: 10.1111/j.1600-0757.1994.tb00019.x. [DOI] [PubMed] [Google Scholar]
  64. Moore WE, Holdeman LV, Smibert RM, Hash DE, Burmeister JA, Ranney RR. Bacteriology of severe periodontitis in young adult humans. Infect Immun. 1982;38:1137–1148. doi: 10.1128/iai.38.3.1137-1148.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Moore WE, Holdeman LV, Cato EP, Smibert RM, Burmeister JA, Ranney RR. Bacteriology of moderate (chronic) periodontitis in mature adult humans. Infect Immun. 1983;42:510–515. doi: 10.1128/iai.42.2.510-515.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Moore WE, Moore LH, Ranney RR, Smibert RM, Burmeister JA, Schenkein HA. The microflora of periodontal sites showing active destructive progression. J Clin Periodontol. 1991;18:729–739. doi: 10.1111/j.1600-051x.1991.tb00064.x. [DOI] [PubMed] [Google Scholar]
  67. Nilius AM, Spencer SC, Simonson LG. Stimulation of in vitro growth of Treponema denticola by extracellular growth factors produced by Porphyromonas gingivalis. J Dent Res. 1993;72:1027–1031. doi: 10.1177/00220345930720060601. [DOI] [PubMed] [Google Scholar]
  68. Orth RK, O’Brien-Simpson NM, Dashper SG, Reynolds EC. Synergistic virulence of Porphyromonas gingivalis and Treponema denticola in a murine periodontitis model. Mol Oral Microbiol. 2011;26:229– 240. doi: 10.1111/j.2041-1014.2011.00612.x. [DOI] [PubMed] [Google Scholar]
  69. Page RC, Lantz MS, Darveau R, et al. Immunization of Macaca fascicularis against experimental periodontitis using a vaccine containing cysteine proteases purified from Porphyromonas gingivalis. Oral Microbiol Immunol. 2007;22:162–168. doi: 10.1111/j.1399-302X.2007.00337.x. [DOI] [PubMed] [Google Scholar]
  70. Park Y, Simionato MR, Sekiya K, et al. Short fimbriae of Porphyromonas gingivalis and their role in coadhesion with Streptococcus gordonii. Infect Immun. 2005;73:3983–3989. doi: 10.1128/IAI.73.7.3983-3989.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Paster BJ, Boches SK, Galvin JL, et al. Bacterial diversity in human subgingival plaque. J Bacteriol. 2001;183:3770–3783. doi: 10.1128/JB.183.12.3770-3783.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Periasamy S, Kolenbrander PE. Mutualistic biofilm communities develop with Porphyromonas gingivalis and initial, early, and late colonizers of enamel. J Bacteriol. 2009;191:6804–6811. doi: 10.1128/JB.01006-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Persson GR. Immune responses and vaccination against periodontal infections. J Clin Periodontol. 2005;32 (Suppl 6):39–53. doi: 10.1111/j.1600-051X.2005.00800.x. [DOI] [PubMed] [Google Scholar]
  74. Quirynen M, Vogels R, Pauwels M, et al. Initial subgingival colonization of ‘pristine’ pockets. J Dent Res. 2005;84:340–344. doi: 10.1177/154405910508400409. [DOI] [PubMed] [Google Scholar]
  75. Ramsey MM, Whiteley M. Polymicrobial interactions stimulate resistance to host innate immunity through metabolite perception. Proc Natl Acad Sci USA. 2009;106:1578–1583. doi: 10.1073/pnas.0809533106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Ramsey MM, Rumbaugh KP, Whiteley M. Metabolite cross-feeding enhances virulence in a model polymicrobial infection. PLoS Pathog. 2011;7:e1002012. doi: 10.1371/journal.ppat.1002012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Schultz-Haudt S, Bruce MA, Bibby BG. Bacterial factors in nonspecific gingivitis. J Dent Res. 1954;33:454–458. doi: 10.1177/00220345540330040301. [DOI] [PubMed] [Google Scholar]
  78. Sela MN. Role of Treponema denticola in periodontal diseases. Crit Rev Oral Biol Med. 2001;12:399–413. doi: 10.1177/10454411010120050301. [DOI] [PubMed] [Google Scholar]
  79. Settem RP, El-Hassan AT, Honma K, Stafford GP, Sharma A. Fusobacterium nucleatum and Tannerella forsythia induce synergistic alveolar bone loss in a mouse periodontitis model. Infect Immun. 2012;80:2436–2443. doi: 10.1128/IAI.06276-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Sharma A. Virulence mechanisms of Tannerella forsythia. Periodontol 2000. 2010;54:106–116. doi: 10.1111/j.1600-0757.2009.00332.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Slots J. Microflora in the healthy gingival sulcus in man. Scand J Dent Res. 1977a;85:247–254. doi: 10.1111/j.1600-0722.1977.tb00560.x. [DOI] [PubMed] [Google Scholar]
  82. Slots J. The predominant cultivable microflora of advanced periodontitis. Scand J Dent Res. 1977b;85:114–121. doi: 10.1111/j.1600-0722.1977.tb00541.x. [DOI] [PubMed] [Google Scholar]
  83. Socransky SS. Microbiology of periodontal disease – present status and future considerations. J Periodontol. 1977;48:497–504. doi: 10.1902/jop.1977.48.9.497. [DOI] [PubMed] [Google Scholar]
  84. Socransky SS, Haffajee AD. Evidence of bacterial etiology: a historical perspective. Periodontol 2000. 1994;5:7–25. doi: 10.1111/j.1600-0757.1994.tb00016.x. [DOI] [PubMed] [Google Scholar]
  85. Socransky SS, Haffajee AD, Cugini MA, Smith C, Kent RL., Jr Microbial complexes in subgingival plaque. J Clin Periodontol. 1998;25:134–144. doi: 10.1111/j.1600-051x.1998.tb02419.x. [DOI] [PubMed] [Google Scholar]
  86. Tanner ACR, Haffer C, Bratthall GT, Visconti RA, Socransky SS. Study of the bacteria associated with advancing periodontitis in man. J Clin Periodontol. 1979;6:278–307. doi: 10.1111/j.1600-051x.1979.tb01931.x. [DOI] [PubMed] [Google Scholar]
  87. Tonetti MS, Imboden MA, Lang NP. Neutrophil migration into the gingival sulcus is associated with transepithelial gradients of interleukin-8 and ICAM- 1. J Periodontol. 1998;69:1139–1147. doi: 10.1902/jop.1998.69.10.1139. [DOI] [PubMed] [Google Scholar]
  88. Wade WG. Has the use of molecular methods for the characterization of the human oral microbiome changed our understanding of the role of bacteria in the pathogenesis of periodontal disease? J Clin Periodontol. 2011;38(Suppl 11):7–16. doi: 10.1111/j.1600-051X.2010.01679.x. [DOI] [PubMed] [Google Scholar]
  89. Wang M, Krauss JL, Domon H, et al. Microbial hijacking of complement-toll-like receptor crosstalk. Sci Signal. 2010;3:ra11. doi: 10.1126/scisignal.2000697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Whitmore SE, Lamont RJ. The pathogenic persona of community-associated oral streptococci. Mol Microbiol. 2011;81:305–314. doi: 10.1111/j.1365-2958.2011.07707.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Xia Q, Wang T, Taub F, et al. Quantitative proteomics of intracellular Porphyromonas gingivalis. Proteomics. 2007;7:4323–4337. doi: 10.1002/pmic.200700543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Xie H, Cai S, Lamont RJ. Environmental regulation of fimbrial gene expression in Porphyromonas gingivalis. Infect Immun. 1997;65:2265–2271. doi: 10.1128/iai.65.6.2265-2271.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Xie H, Chung WO, Park Y, Lamont RJ. Regulation of the Porphyromonas gingivalis fimA (Fimbrillin) gene. Infect Immun. 2000;68:6574–6579. doi: 10.1128/iai.68.12.6574-6579.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Ximenez-Fyvie LA, Haffajee AD, Socransky SS. Comparison of the microbiota of supra- and subgingival plaque in health and periodontitis. J Clin Periodontol. 2000a;27:648–657. doi: 10.1034/j.1600-051x.2000.027009648.x. [DOI] [PubMed] [Google Scholar]
  95. Ximenez-Fyvie LA, Haffajee AD, Socransky SS. Microbial composition of supra- and subgingival plaque in subjects with adult periodontitis. J Clin Periodontol. 2000b;27:722–732. doi: 10.1034/j.1600-051x.2000.027010722.x. [DOI] [PubMed] [Google Scholar]
  96. Yoshioka H, Yoshimura A, Kaneko T, Golenbock DT, Hara Y. Analysis of the activity to induce toll-like receptor (TLR)2- and TLR4-mediated stimulation of supragingival plaque. J Periodontol. 2008;79:920–928. doi: 10.1902/jop.2008.070516. [DOI] [PubMed] [Google Scholar]

RESOURCES