Next Article in Journal
YycH and YycI Regulate Expression of Staphylococcus aureus Autolysins by Activation of WalRK Phosphorylation
Next Article in Special Issue
A Novel Small Molecule, 1,3-di-m-tolyl-urea, Inhibits and Disrupts Multispecies Oral Biofilms
Previous Article in Journal
Temporal Dynamics of the Gut Bacteriome and Mycobiome in the Weanling Pig
Previous Article in Special Issue
Characterization of Scardovia wiggsiae Biofilm by Original Scanning Electron Microscopy Protocol
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Porphyromonas gingivalis, a Long-Range Pathogen: Systemic Impact and Therapeutic Implications

1
Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
2
Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
3
Faculté de Chirurgie-Dentaire, Université de Strasbourg, 8 rue Sainte-Elisabeth, 67000 Strasbourg, France
4
INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
*
Author to whom correspondence should be addressed.
Microorganisms 2020, 8(6), 869; https://doi.org/10.3390/microorganisms8060869
Submission received: 27 May 2020 / Revised: 4 June 2020 / Accepted: 5 June 2020 / Published: 9 June 2020
(This article belongs to the Special Issue Oral Biofilms)

Abstract

:
Periodontitis is an inflammatory disease associated with a dysbiosis of the oral flora characterized by a chronic sustained inflammation leading to destruction of tooth-supporting tissues. Over the last decade, an association between periodontitis and systemic disorders such as cardiovascular diseases, rheumatoid arthritis and obesity has been demonstrated. The role of periodontal pathogens, notably Porphyromonas gingivalis (P. gingivalis), in the onset or exacerbation of systemic diseases has been proposed. P. gingivalis expresses several virulence factors that promote its survival, spreading, and sustaining systemic inflammation. Recently, the impact of periodontitis on gut dysbiosis has also been suggested as a potential mechanism underlying the systemic influence of periodontitis. New therapeutic strategies for periodontitis and other dysbiotic conditions, including the use of beneficial microbes to restore healthy microbial flora, may pave the way to improved therapeutic outcomes and more thorough patient management.

1. Introduction

Periodontitis is an oral inflammatory disease of infectious origin affecting periodontium, characterized by gingival swelling and bleeding, periodontal pocket formation and clinical attachment loss, destruction of tooth-supporting tissue and ultimately to tooth loss [1,2]. Periodontitis is among the most common diseases worldwide, with its severe form affecting approximately 11% of the global population [3]. This disease represents a major public health issue as periodontitis has been correlated with decreased oral health and related quality of life [4]. Furthermore, an association with several systemic diseases such as cardiovascular diseases, diabetes and rheumatoid arthritis was established [5]. The oral microbiome is a highly complex ecosystem with thousands of microbial phylotypes detected [6], and its understanding presents a challenge. In the context of periodontal inflammation, the contribution of microbiome dysbiosis is under investigation due to the molecular mechanisms underlying this microbiome state being poorly understood.
Microbiome dysbiosis is characterized by an imbalance between microbial species and their environment. The imbalance seems to be driven by the subversion of host immunity due to keystone pathogens even when they are present in low abundance [7,8]. Typically, an inflammatory host response would lead to the clearance of pathogenic bacteria. However, in the case of periodontal disease, it promotes the growth of keystone pathogens due to gingival inflammatory exudate being a rich source of nutrients [9]. Keystone pathogens, such as Porphyromonas gingivalis (P. gingivalis), facilitate the subversion of the host immune response through polymicrobial synergy allowing for the growth of both themselves and other dysbiotic species [10,11,12]. Polymicrobial synergy within the pathogenic oral biofilm is critical for the establishment and spread of periodontal disease. It has been demonstrated in vitro that there is a cooperative biofilm formation involving Streptococcus gordonii, Fusobacterium nucleatum (F. nucleatum) and P. gingivalis [12]. When cocultured, these bacteria elicited extensive changes on each other’s biochemical signatures to support cohabitation within the biofilm [13]. During colonization, F. nucleatum triggers the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in the host cells which provides a more favorable environment for P. gingivalis attachment [14]. Furthermore, this leads to the upregulation of virulence factor, mfa1 fimbriae, in both F. nucleatum and P. gingivalis preventing dendritic cell maturation allowing the disruption of the innate immune response [12].
P. gingivalis deploys an extensive arsenal of virulence factors such as lipopolysaccharide, proteases, fimbriae and a CRISPR-Cas system [8,15,16]; enabling it to modulate the host immune response to promote its survival through cellular colonization and spreading [17,18]. During the initial phase of infection, this bacterium manipulates the immune system through inhibition of cytokines and chemokines secretion [19,20]. Neutrophil homing to the gingival tissues is critical for maintaining homeostasis between the host and the microbiome. P. gingivalis creates a chemokine paralysis by degrading IL-8 through its secreted gingipain proteases and prevents IL-8 transcription through SerB, a haloacid dehalogenase (HAD) family serine phosphatase [21,22]. Furthermore, the ability of P. gingivalis to persist in the periodontal tissue after chemokine paralysis may depend to its ability to hijack the complement system, preventing its clearance from the oral cavity [11,23,24]. In addition to paralyzing the immune response, P. gingivalis also targets other periodontal cell types such as gingival epithelial cells, fibroblasts, periodontal ligament cells, and osseous cells, leading to the establishment of an inflammatory environment [17,18,25,26,27]. This hijacking of the host immune response hinders immune cell recruitment, allowing P. gingivalis to spread and colonize the periodontal pocket.

2. Distant Dissemination of P. gingivalis

Over the last decade, there has been increased interest on the links between periodontitis and systemic diseases [5]. Among them, an association has been demonstrated with major chronic diseases such as cardiovascular diseases [28], diabetes [29], metabolic syndrome [30], rheumatoid arthritis [31] and more recently obesity [32] (Figure 1). Interestingly, periodontitis has been documented as contributing to the risk of all-cause mortality in an older European population (subjects 60–70 years of age; hazard ratio = 1.57 (1.04–2.36) after adjustment for confounding variables) [33]. Therefore, the systemic effects of oral pathogens and the role they play in chronic diseases has become a major research focus.
Among the oral bacteria that exhibit systemic effects, P. gingivalis has stood out. It has been detected in several diseased tissues and organs in both humans and animal models. The translocation of P. gingivalis to the distant tissues such as the liver or joint after oral administration [34,35] and its detection in brains of patients with Alzheimer’s disease [36] has led to an increased interest in determining its role in chronic inflammatory diseases.

3. Pathogenicity of P. gingivalis: The Case of Atherothrombosis

Chronic infection has been identified as a potential contributor to the development of atherosclerotic lesions independent of classical risk factors such as unfavorable lipid profile [37,38]. Chlamydia pneumoniae, Helicobacter pylori, P. gingivalis and certain viruses have been detected within atheromatous plaque [39,40,41]. However, their impact remains under investigation. Several periodontal pathogens have been detected in both atherosclerotic plaque and healthy vessels [41,42]. P. gingivalis is among the most commonly found organism in these studies due to its ability to persist within vascular tissue through cell-to-cell transmission [43].
The severity of periodontitis in experimental models has been shown to correlate with the magnitude of the systemic inflammation as well as atheromatous plaque formation. Oral administration of P. gingivalis has been reported to accelerate the development of atherosclerosis in apolipoprotein E knock out (Apoe−/−) mice [38,44,45]. In the experimental models of periodontitis, it has been demonstrated that there is an increased systemic inflammation, potentially contributing to vascular lesion development [46,47]. Signaling through TLR-2 and TLR-4 is critical for the development of periodontitis, as well as atherosclerotic plaque progression [48]. It has been demonstrated that P. gingivalis is able to activate these membrane receptors on the endothelial level [49,50] triggering the secretion of cytokines such as TNF-α, IL-1, IL-18 and M-CSF [51] thus, contributing to a persistent inflammation. Furthermore, in these mice, P. gingivalis DNA can be found in the aortic tissue along with an abundance of activated macrophages [44,45]. Interestingly, when nonsurgical periodontal therapy is performed in these models, there is a reduction in systemic inflammation as well as aortic inflammation supporting the causative role oral microbiome dysbiosis [52].
Oxidized low density lipoproteins (OxLDLs) are believed to initiate the immunological response found in atherosclerosis [53]. It has been demonstrated in patients with stable coronary artery disease, as well as acute coronary disease, that antibodies directed against key virulence factors of P. gingivalis can cross-react with OxLDLs, malondialdehyde-modified low-density lipoprotein (MDA-LDL) and to malondialdehyde acetaldehyde-modified low-density lipoprotein (MAA-LDL) [54]. In vitro, the synergic effects between risk factors have also been observed. Indeed, P. gingivalis-induced effects can be influenced by the cellular environment and other proinflammatory triggers, such as oxidized low-density lipoproteins, (Ox-LDL) [55] highlighting the fact that vascular inflammation could arise from different mechanisms and insults. The antibody cross-reaction between bacterial components and host molecules may also contribute to the inflammatory environment in the atherosclerotic plaque. In mouse models, immunization with P. gingivalis or its gingipain, Rgp44, seems to display athero-protective effects in the modulation of plaque size, and anti-inflammatory cytokines IL-10 and IL-5 [54].
Based on the cumulative evidence strengthening the role of bacterial infection in atherosclerosis, several clinical trials have been conducted assessing the impact of antibiotic administration on atherosclerosis-related parameters in patients with stable coronary artery disease (WIZARD, ACES, CLARICOR), acute coronary syndrome (PROVE-IT-TIMI) and peripheral artery disease (PROVIDENCE-1) [56,57,58,59,60]. However, none of these trials demonstrated long term benefit of such a therapeutic strategy. These results might be explained by the fact that 1. antibiotics never penetrate a biofilm unless mechanically disrupted; 2. antibiotics must be able to address host cell invasion by pathogens; 3. the poorly designed studies focus mainly on late stages of atherothrombosis, reducing the ability to detect a benefit of such an approach [61]. Future trials testing the effect of antibiotics on earlier stage disease might be worth consideration.

4. P. gingivalis and Rheumatoid Arthritis

Rheumatoid arthritis (RA) is a systemic autoimmune inflammatory disease that primary affects the joints, however, the condition can damage a wide variety of body systems including the skin, eyes, lungs, heart and blood vessels [62]. The notion that RA had an infectious origin is not a new one. Since the early 19th century, investigators have claimed to identify different bacteria in RA synovial fluid and tissue. Although these results were not reproducible, the evidence supported the theory that RA was a result of microbial action either within the joint or systemically [63]. The association between periodontal disease and RA has long been established [64]. Initially, it was thought that the alveolar bone destruction and gingival inflammation was part of the same RA-induced damage. It was not until recently that periodontitis was seen more as a cause than a consequence of RA. Like periodontitis, the tissue damage seen in RA is due to a host driven inflammation [65,66]. The cytokine signature of each disease is similar with an increased expression of proinflammatory cytokines, TNF-α and IL-6. In murine models of experimental arthritis, it was found that there is an increase in the serum levels of these inflammatory mediators following oral administration of periodontal pathogens, supporting their role in exacerbating the local and systemic inflammation associated with RA [67,68,69]. Additionally, auto-antibodies such as rheumatoid factor and anticollagen antibodies common to RA are often found in diseased periodontal tissues [70,71]. Specific periodontal pathogens have even been identified as risk factors for the development of RA, and P. gingivalis in particular has received a lot of attention. P. gingivalis secretes a petidylarginine deiminase (PAD)-like enzyme (PPAD), which can contribute to the generation of citrullinated proteins, a major target of autoantibodies in RA [72,73]. PPAD is secreted in a soluble form and in association with the outer membrane vesicles of P. gingivalis [74]. As a major virulence factor, PPAD helps to maintain the presence of P. gingivalis in the mouth by preventing neutrophil function as well as the production of ammonia [75,76,77]. Clinical isolates of P. gingivalis have been shown to be highly enriched in citrullinated proteins due to the functioning of PPAD as well as its gingipain Rgp [73,78]. In murine models of collagen-induced arthritis, inoculation with P. gingivalis lead to an increase in citrullinated peptides as well as an increased expression of proteins targeted for citrullination [78]. The production of bacterial and host citrullinated proteins by PPAD may further exacerbate the loss of tolerance to citrullinated autoantigens in RA. In murine models of collagen-induced arthritis PPAD-null mutants, P. gingivalis does not elicit the same level of inflammation or autoantibodies to collagen type II, highlighting its potential for therapeutic targeting [73]. In addition to autoreactive antibodies, patients with RA often have high levels of antibodies to P. gingivalis gingipain domains [79]. A majority of the antigingipain antibodies target the hemagglutinin (HA2-4) as well as the catalytic domains [80]. Interestingly, in the murine model of collagen-induced arthritis, rats that received a pretreatment vaccination of the catalytic domain of RgpA had less severe cartilage erosive changes when compared to controls. Furthermore, rats that received the catalytic domain of RgpA demonstrated a reduction in inflammatory mediators (IL-1b and TNF-α) both at the mRNA and protein level in the synovial tissues [79].
It is difficult to clearly define periodontitis and periodontal pathogens as microbial cause of RA, given 11% of the global adult population present periodontitis and RA affects only 0.5 to 1% [3,81]. However, there is overwhelming evidence to support that periodontal pathogens, specifically P. gingivalis, play a role in the progression of the disease by allowing for the accumulation of inflammatory cytokines and autoreactive antibodies, leading to the tissue destruction associated with RA [69,82,83,84]. Furthermore, it has been demonstrated in numerous animal models of experimental arthritis that arthritis can be treated by targeting P. gingivalis and its numerous virulence factors, supporting the need for further investigation into the effects of nonsurgical periodontal treatment and the progression of RA in human trials.

5. Impact of P. gingivalis on Obesity, Metabolism, and Gut Microbiota Homeostasis

The amount of data illustrating a relationship between obesity and periodontitis [85] has led to interest in the potential role played by P. gingivalis on both sustained inflammation in adipose tissue and gut microbiome-associated changes. It has been suggested that bacterial insult may contribute to obesity-related chronic inflammation and oxidative stress [86]. Endotoxemia characterized by high plasma level of bacterial lipopolysaccharides (LPS) has been shown to stimulate adipocyte proliferation and inflammation in adipose tissue [87]. Interestingly, infection of macrophages by P. gingivalis induced a switch in macrophage polarization leading to the inhibition of M1 phenotype, illustrating an alteration of immune response with effects on a cascade of pathways associated with immune cell proliferation and angiogenesis. These effects are potential causes of the delay in the response to infection observed in obese individuals [88]. However, it should be mentioned that a sole bacterium cannot cause metabolic endotoxemia and that the establishment of the low-grade inflammation is the result of a complex process that involves the whole microbiota, including butyrate-producing bacteria, mucin-degrading bacteria and LPS-containing bacteria such as P. gingivalis [87,89,90,91].
The gut microbiome has recently come to be viewed as an organ system in its own right [92]. It is the most diverse commensal ecosystem in the human body that continues to evolve throughout the human lifespan [93]. Microbes residing in the gut are considered key contributors to host metabolism as they interact with virtually all human cells and induce significant changes in host metabolism [89]. Metabolic disorders, such as obesity, are associated with shifts in the microbiota at the phylum level, especially in the Firmicutes/Bacteroidetes ratio [94]. Saliva is an important transporter of oral bacteria to the gut as daily saliva production is approximately 0.6 L [95]. Clinical trials have demonstrated that bacterial counts of periodontal pathogens in saliva (Campylobacter rectus, F. nucleatum, P. gingivalis, Prevotella intermedia and Tannerella forsythia) were increased with the severity of periodontal disease (gingivitis/periodontitis) [96]. It has been hypothesized that swallowed oral pathogens can lead to disturbances in the gut microbiota causing metabolic endotoxemia and metabolic disorders [97]. Patients with chronic periodontitis have been shown to have less diversity in their gut microbiomes [98]. Several studies have investigated the influence of P. gingivalis oral gavage on the gut microbiome, mostly in animal models. All studies indicate a significant modification of gut microbiome composition after P. gingivalis oral administration [99,100]. In C57BL/6 mice infected with P. gingivalis strain W83, Kato et al. showed that the proportion of Bacteroidetes was significantly reduced compared to sham-administered mice, indicating a dysbiosis of the gut microbiome. Interestingly, the proportions of other genera such as Lactobacillus and Desulfovibrio were also affected by P. gingivalis administration [99]. Due to the changes in the gut microbiome, P. gingivalis administration leads to increased systemic inflammation, endotoxin levels, and intestinal permeability, even after a single exposure [100]. The increased systemic inflammation contributes to the changes in gene expression profiles of epididymal adipose tissue, increasing proinflammatory genes while decreasing insulin sensitivity [97], as well as changes in expression of genes associated with glucose and lipid metabolism [99,101]. However, the limitations associated with the use of mouse models should be mentioned. Mouse models lack some human-specific gut bacteria and proportion of bacterial phyla is different compared to humans. Furthermore, the immune responses also differ [102]. Altogether, these limitations indicate the need of human studies. In humans, the Human Microbiome Project demonstrated that several genera, including Bacteroides, Faecalibacterium, Parabacteroides, Eubacteium, Alistipes, Dialister, Streptococcus, Prevotella, Rosburia, Coprococcus, Veillonella and Oscilibacter could be detected in both the oral cavity and stool in more than 45% of the included patients [103], highlighting a potential role for oral bacteria in the disruption of the gut microbiome. It was therefore hypothesized that saliva is a key driver of microbial composition in the habitats above the stomach. However, the role of the continuous epithelial lining of the upper gastrointestinal mucosal surfaces has also been suggested [103].
Obesity is also associated with metabolic alterations such as glucose homeostasis including glucose intolerance, diabetes and insulin resistance [104], all of which are associated with low-grade inflammation. Metabolic endotoxemia induced by LPS has been associated with insulin resistance [105]. Interestingly, it was demonstrated that mice fed a high-fat diet and treated with antibiotics exhibited reduced inflammation, insulin resistance and fat mass gain, emphasizing a role of the gut microbiota [106]. In the context of periodontitis and experimental periodontitis induced by P. gingivalis, obese animals exhibited greater periodontal bone loss that in lean animals; a process attributed to a severe immune dysfunction [88,107,108,109]. Experimental periodontitis induced by multispecies (P. gingivalis, F. nucleatum, P. intermedia) oral gavage was associated with increased insulin resistance [110]. This modulation of the insulin resistance was linked to the recruitment of cells involved in the adaptive immune response, and a decrease in antibodies against P. gingivalis was associated with impaired glucose metabolism [110].
Conventional periodontal treatments including scaling and root planning with adjuvant therapies such as topical antibiotics have been demonstrated to alleviate some of the symptoms of metabolic disorders. In fact, such treatments have led to a reduction in serum glucose and glycated hemoglobin, insulin resistance, and fasting glucose levels [111,112,113]. There is little understanding of the impact of periodontal treatment on the composition of the gut microbiome. Due to the ability of periodontal treatment to change the serum metabolome, the effects on the composition of the microbiome should be investigated.

6. Clinical Implications

Periodontal treatment aims to achieve resolution of gingival inflammation and a decrease in the pathogenic bacterial load [114]. It includes nonsurgical periodontal therapy consisting of oral hygiene instructions, scaling and root planning of affected teeth. In cases of severe lesions, anti-infective treatment may be used as an adjunctive to mechanical treatment. Accordingly, antiseptics, antibiotics, and local anti-infective treatments such as photodynamic therapy have been proposed to reduce the need for additional surgical treatment [115,116,117]. Such therapeutic approaches are effective in most clinical situations. However, in some patients, refractory lesions and recurrence leading ultimately to tooth loss could be observed [118,119], emphasizing the need for pharmacological approaches such as antibiotics.
Recently, the modulation of the biofilm composition has also been proposed as a therapeutic approach through the administration of beneficial microbes [120,121]. In this aspect, Lactobacillus strains, especially L.reuteri, were the most evaluated in human clinical settings. They showed a significant impact with a reduction in clinical attachment loss (−0.42 mm, p = 0.002) and gingival inflammation, as well as a significant reduction in bleeding on probing (−14.66%, p = 0.03) [121]. The mechanisms associated with oral administration of probiotics at the periodontal level are not fully elucidated yet. However, probiotic treatment has demonstrated an inhibitory effect towards bacteria present within periodontal biofilms such as Streptococcus mutans, Streptococcus gordonii, Tannerella forsythia and Actinomyces naeslundi. Interestingly, it was also demonstrated that the antimicrobial activity of L. reuteri was not associated with its viability, as even heat-killed bacteria or culture supernatant were effective, indicating a likely role for substances produced by the beneficial bacteria such as reutericyclin [122]. Interestingly, in rodent models, the restoration of a healthy gut flora concomitant with administration of probiotics such as L. plantarum was associated to cardioprotective effects, improved cardiac function and decreased inflammatory markers [123].
The gut symbiont Akkermansia muciniphila (A. muciniphila) has been evaluated as a new potential therapeutic tool for periodontitis. A. muciniphila has attracted growing interest due to its host-beneficial properties. In several rodent models, supplementation with A. muciniphila reduces obesity, insulin resistance, glucose intolerance, steatosis and gut permeability [124,125,126]. Such properties suggest A. muciniphila as a next-generation probiotic [127]. Furthermore, there has been additional focus on one of its membrane proteins, Amuc_1100 [128], demonstrating its ability to recapitulate the effect induced by live bacteria. Amuc_1100 interacts with TLR- 2 and -4 resulting in the improvement of gut barrier function and anti-inflammatory IL-10 secretion in mouse models [125,129]. In humans, a recent proof-of-concept exploratory study shows that the supplementation with A. muciniphila was safe and well tolerated. Most importantly, the administration of this gut commensal administration in human obese volunteers, notably in pasteurized form, contributed to body weight reduction in comparison with placebo treated patients, but also reduced the level of markers associated with liver dysfunction and inflammation such as fasting plasma insulin, total cholesterol and γGT [130]. In the context of periodontitis, administration of A.muciniphila in a murine model of experimental periodontitis was associated with improved periodontal healing [120]. This effect was associated with a modulation of P. gingivalis virulence by A. muciniphila in cocultures. As observed for L. reuteri, some studies showed the role of a secreted bacterial byproduct, Amuc_1100 in the anti-inflammatory properties [125]. The impact of the oral administration of A. muciniphila on the gut microbiome was also observed and associated with changes in the ratio of Firmicutes/Bacteroidetes, highlighting a potential influence of gut ecology on systemic inflammation levels [120].

7. Conclusions

The detrimental effect of periodontitis on other major systemic diseases with high social and financial consequences has been demonstrated. Modulation of the microbiome has emerged as an exciting potential treatment that could pave the way for a new approach to managing periodontal diseases, as well as their systemic effects. New research is needed to establish the molecular mechanisms involved, and to elucidate effects related to both local antimicrobial properties and systemic anti-inflammatory effects associated with the transition from a dysbiotic to a healthy gut microbiome. Additionally, studies are needed to determine the most effective probiotic organisms, strains, dosage, and routes of administration necessary to achieve optimal outcomes in each respective disease. With further research into the effects of modulation of the oral microbiome on systemic disease, promising new treatments for these chronic inflammatory diseases will emerge.

Author Contributions

Conceptualization, O.H. and S.A.; methodology, O.H, S.A.; data curation, H.M. and O.H.; writing—original draft preparation, H.M., O.H. and S.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH/NIDCR, grant #R01DE014079 and NIH/NHLBI grant #R01HL076801.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Caton, J.G.; Armitage, G.; Berglundh, T.; Chapple, I.L.C.; Jepsen, S.; Kornman, K.S.; Mealey, B.L.; Papapanou, P.N.; Sanz, M.; Tonetti, M.S. A new classification scheme for periodontal and peri-implant diseases and conditions—Introduction and key changes from the 1999 classification. J. Periodontol. 2018, 89, S1–S8. [Google Scholar] [CrossRef] [PubMed]
  2. Kinane, D.F.; Stathopoulou, P.G.; Papapanou, P.N. Periodontal diseases. Nat. Rev. Dis. Primer 2017, 3, 17038. [Google Scholar] [CrossRef] [PubMed]
  3. Kassebaum, N.J.; Bernabe, E.; Dahiya, M.; Bhandari, B.; Murray, C.J.L.; Marcenes, W. Global Burden of Severe Periodontitis in 1990-2010: A Systematic Review and Meta-regression. J. Dent. Res. 2014, 93, 1045–1053. [Google Scholar] [CrossRef] [PubMed]
  4. Sulaiman, L.; Saub, R.; Baharuddin, N.A.; Safii, S.H.; Gopal Krishna, V.; Bartold, P.M.; Vaithilingam, R.D. Impact of Severe Chronic Periodontitis on Oral Health-related Quality of Life. Oral Health Prev. Dent. 2019, 17, 365–373. [Google Scholar] [CrossRef] [PubMed]
  5. Linden, G.J.; Lyons, A.; Scannapieco, F.A. Periodontal systemic associations: Review of the evidence. J. Clin. Periodontol. 2013, 40, 8–19. [Google Scholar] [CrossRef]
  6. Keijser, B.J.F.; Zaura, E.; Huse, S.M.; van der Vossen, J.M.B.M.; Schuren, F.H.J.; Montijn, R.C.; ten Cate, J.M.; Crielaard, W. Pyrosequencing analysis of the oral microflora of healthy adults. J. Dent. Res. 2008, 87, 1016–1020. [Google Scholar] [CrossRef]
  7. Lamont, R.J.; Koo, H.; Hajishengallis, G. The oral microbiota: Dynamic communities and host interactions. Nat. Rev. Microbiol. 2018, 16, 745–759. [Google Scholar] [CrossRef]
  8. Zenobia, C.; Hajishengallis, G. Porphyromonas gingivalis virulence factors involved in subversion of leukocytes and microbial dysbiosis. Virulence 2015, 6, 236–243. [Google Scholar] [CrossRef] [Green Version]
  9. Darveau, R.P. Periodontitis: A polymicrobial disruption of host homeostasis. Nat. Rev. Microbiol. 2010, 8, 481–490. [Google Scholar] [CrossRef]
  10. Zenobia, C.; Hasturk, H.; Nguyen, D.; Dyke, T.E.V.; Kantarci, A.; Darveau, R.P. Porphyromonas gingivalis Lipid A Phosphatase Activity Is Critical for Colonization and Increasing the Commensal Load in the Rabbit Ligature Model. Infect. Immun. 2014, 82, 650–659. [Google Scholar] [CrossRef] [Green Version]
  11. Hajishengallis, G.; Liang, S.; Payne, M.A.; Hashim, A.; Jotwani, R.; Eskan, M.A.; McIntosh, M.L.; Alsam, A.; Kirkwood, K.L.; Lambris, J.D.; et al. Low-Abundance Biofilm Species Orchestrates Inflammatory Periodontal Disease through the Commensal Microbiota and Complement. Cell Host Microbe 2011, 10, 497–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. El-Awady, A.; de Sousa Rabelo, M.; Meghil, M.M.; Rajendran, M.; Elashiry, M.; Stadler, A.F.; Foz, A.M.; Susin, C.; Romito, G.A.; Arce, R.M.; et al. Polymicrobial synergy within oral biofilm promotes invasion of dendritic cells and survival of consortia members. Npj Biofilms Microbiomes 2019, 5, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hendrickson, E.L.; Wang, T.; Dickinson, B.C.; Whitmore, S.E.; Wright, C.J.; Lamont, R.J.; Hackett, M. Proteomics of Streptococcus gordonii within a model developing oral microbial community. BMC Microbiol. 2012, 12, 211. [Google Scholar] [CrossRef] [Green Version]
  14. Ahn, S.H.; Song, J.-E.; Kim, S.; Cho, S.-H.; Lim, Y.K.; Kook, J.-K.; Kook, M.-S.; Lee, T.-H. NOX1/2 activation in human gingival fibroblasts by Fusobacterium nucleatum facilitates attachment of Porphyromonas gingivalis. Arch. Microbiol. 2016, 198, 573–583. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, T.; Olsen, I. Porphyromonas gingivalis and its CRISPR-Cas system. J. Oral Microbiol. 2019, 11, 1638196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. 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. 2013, 5, 19804. [Google Scholar] [CrossRef]
  17. Bugueno, I.M.; Batool, F.; Korah, L.; Benkirane-Jessel, N.; Huck, O. Porphyromonas gingivalis Differentially Modulates Apoptosome Apoptotic Peptidase Activating Factor 1 in Epithelial Cells and Fibroblasts. AJPA 2018, 188, 404–416. [Google Scholar] [CrossRef] [Green Version]
  18. Bugueno, I.M.; Batool, F.; Keller, L.; Kuchler-Bopp, S.; Benkirane-Jessel, N.; Huck, O. Porphyromonas gingivalis bypasses epithelial barrier and modulates fibroblastic inflammatory response in an in vitro 3D spheroid model. Sci. Rep. 2018, 8, 30. [Google Scholar] [CrossRef] [Green Version]
  19. Huck, O.; Elkaim, R.; Davideau, J.-L.; Tenenbaum, H. Porphyromonas gingivalis-impaired innate immune response via NLRP3 proteolysis in endothelial cells. Innate Immun. 2014, 21, 1–8. [Google Scholar] [CrossRef] [Green Version]
  20. Yu, S.; Ding, L.; Liang, D.; Luo, L. Porphyromonas gingivalis inhibits M2 activation of macrophages by suppressing α-ketoglutarate production in mice. Mol. Oral Microbiol. 2018, 33, 388–395. [Google Scholar] [CrossRef]
  21. Sochalska, M.; Potempa, J. Manipulation of Neutrophils by Porphyromonas gingivalis in the Development of Periodontitis. Front. Cell. Infect. Microbiol. 2017, 7, 197. [Google Scholar] [CrossRef] [Green Version]
  22. Takeuchi, H.; Hirano, T.; Whitmore, S.E.; Morisaki, I.; Amano, A.; Lamont, R.J. The Serine Phosphatase SerB of Porphyromonas gingivalis Suppresses IL-8 Production by Dephosphorylation of NF-κB RelA/p65. PLoS Pathog. 2013, 9, e1003326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Maekawa, T.; Krauss, J.L.; Abe, T.; Jotwani, R.; Triantafilou, M.; Triantafilou, K.; Hashim, A.; Hoch, S.; Curtis, M.A.; Nussbaum, G.; et al. Porphyromonas gingivalis manipulates complement and TLR signaling to uncouple bacterial clearance from inflammation and promote dysbiosis. Cell Host Microbe 2014, 15, 768–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Popadiak, K.; Potempa, J.; Riesbeck, K.; Blom, A.M. Biphasic Effect of Gingipains from Porphyromonas gingivalis on the Human Complement System. J. Immunol. 2007, 178, 7242–7250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Han, E.-C.; Choi, S.-Y.; Lee, Y.; Park, J.-W.; Hong, S.-H.; Lee, H.-J. Extracellular RNAs in periodontopathogenic outer membrane vesicles promote TNF-α production in human macrophages and cross the blood-brain barrier in mice. FASEB J. 2019. [Google Scholar] [CrossRef] [Green Version]
  26. Lapérine, O.; Cloitre, A.; Caillon, J.; Huck, O.; Bugueno, I.M.; Pilet, P.; Sourice, S.; Le Tilly, E.; Palmer, G.; Davideau, J.-L.; et al. Interleukin-33 and RANK-L Interplay in the Alveolar Bone Loss Associated to Periodontitis. PLoS ONE 2016, 11, e0168080-17. [Google Scholar] [CrossRef] [Green Version]
  27. Yuan, H.; Gupte, R.; Zelkha, S.; Amar, S. Receptor activator of nuclear factor kappa B ligand antagonists inhibit tissue inflammation and bone loss in experimental periodontitis. J. Clin. Periodontol. 2011, 38, 1029–1036. [Google Scholar] [CrossRef]
  28. Huck, O.; Saadi-Thiers, K.; Tenenbaum, H.; Davideau, J.-L.; Romagna, C.; Laurent, Y.; Cottin, Y.; Roul, J.G. Evaluating periodontal risk for patients at risk of or suffering from atherosclerosis: Recent biological hypotheses and therapeutic consequences. Arch. Cardiovasc. Dis. 2017, 1–7. [Google Scholar] [CrossRef] [Green Version]
  29. Preshaw, P.M.; Alba, A.L.; Herrera, D.; Jepsen, S.; Konstantinidis, A.; Makrilakis, K.; Taylor, R. Periodontitis and diabetes: A two-way relationship. Diabetologia 2012, 55, 21–31. [Google Scholar] [CrossRef] [Green Version]
  30. Nibali, L.; Tatarakis, N.; Needleman, I.; Tu, Y.-K.; D’Aiuto, F.; Rizzo, M.; Donos, N. Association between Metabolic Syndrome and Periodontitis: A Systematic Review and Meta-analysis. J. Clin. Endocrinol. Metab. 2013, 98, 913–920. [Google Scholar] [CrossRef] [Green Version]
  31. Chou, Y.-Y.; Lai, K.-L.; Chen, D.-Y.; Lin, C.-H.; Chen, H.-H. Rheumatoid Arthritis Risk Associated with Periodontitis Exposure: A Nationwide, Population-Based Cohort Study. PLoS ONE 2015, 10, e0139693. [Google Scholar] [CrossRef] [PubMed]
  32. Nascimento, G.G.; Peres, M.A.; Mittinty, M.N.; Peres, K.G.; Do, L.G.; Horta, B.L.; Gigante, D.P.; Corrêa, M.B.; Demarco, F.F. Diet-Induced Overweight and Obesity and Periodontitis Risk: An Application of the Parametric G-Formula in the 1982 Pelotas Birth Cohort. Am. J. Epidemiol. 2017, 185, 442–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Linden, G.J.; Linden, K.; Yarnell, J.; Evans, A.; Kee, F.; Patterson, C.C. All-cause mortality and periodontitis in 60–70-year-old men: A prospective cohort study. J. Clin. Periodontol. 2012, 39, 940–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ishikawa, M.; Yoshida, K.; Okamura, H.; Ochiai, K.; Takamura, H.; Fujiwara, N.; Ozaki, K. Oral Porphyromonas gingivalis Translocates to the Liver and Regulates Hepatic Glycogen Synthesis through the Akt/GSK-3β Signaling Pathway. Biochim. Biophys. Acta 2013, 1832, 2035–2043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Jeong, S.H.; Nam, Y.; Jung, H.; Kim, J.; Rim, Y.A.; Park, N.; Lee, K.; Choi, S.; Jang, Y.; Kim, Y.; et al. Interrupting Oral Infection of Porphyromons gingivalis with anti-FimA Antibody Attenuates Bacterial Dissemination to the Arthritic Joint and Improves Experimental Arthritis. Exp. Mol. Med. 2018, 50, 460. [Google Scholar] [CrossRef] [PubMed]
  36. Panza, F.; Lozupone, M.; Solfrizzi, V.; Watling, M.; Imbimbo, B.P. Time to test antibacterial therapy in Alzheimer’s disease. Brain 2019, 142, 2905–2929. [Google Scholar] [CrossRef]
  37. Rosenfeld, M.E.; Campbell, L.A. Pathogens and atherosclerosis: Update on the potential contribution of multiple infectiou organisms to the pathogenesis of atherosclerosis. Thromb. Haemost. 2011, 106, 858–867. [Google Scholar] [CrossRef] [PubMed]
  38. Li, L.; Messas, E.; Batista, E.L.; Levine, R.A.; Amar, S. Porphyromonas gingivalis infection accelerates the progression of atherosclerosis in a heterozygous apolipoprotein E-deficient murine model. Circulation 2002, 105, 861–867. [Google Scholar]
  39. Sessa, R.; Di Pietro, M.; Schiavoni, G.; Petrucca, A.; Cipriani, P.; Zagaglia, C.; Nicoletti, M.; Santino, I.; del Piano, M. Measurement of Chlamydia pneumoniae bacterial load in peripheral blood mononuclear cells may be helpful to assess the state of chlamydial infection in patients with carotid atherosclerotic disease. Atherosclerosis 2007, 195, 224–230. [Google Scholar] [CrossRef]
  40. Izadi, M.; Fazel, M.; Sharubandi, S.H.; Saadat, S.H.; Farahani, M.M.; Nasseri, M.H.; Dabiri, H.; SafiAryan, R.; Esfahani, A.A.; Ahmadi, A.; et al. Helicobacter Species in the Atherosclerotic Plaques of Patients with Coronary Artery Disease. Cardiovasc Pathol. 2012, 21, 307–311. [Google Scholar] [CrossRef]
  41. Elkaim, R.; Dahan, M.; Kocgozlu, L.; Werner, S.; Kanter, D.; Kretz, J.G.; Tenenbaum, H. Prevalence of periodontal pathoges in subgingival lesions, atherosclerotic plaques and healthy blood vessels: A preliminary study. J. Periodontal Res. 2008, 43, 224–231. [Google Scholar] [CrossRef] [PubMed]
  42. Rangé, H.; Labreuche, J.; Louedec, L.; Rondeau, P.; Planesse, C.; Sebbag, U.; Bourdon, E.; Michel, J.-B.; Bouchard, P.; Meilhac, O. Periodontal bacteria in human carotid atherothrombosis as a potential trigger for neutrophil activation. Atherosclerosis 2014, 236, 448–455. [Google Scholar] [CrossRef] [PubMed]
  43. Li, L.; Michel, R.; Cohen, J.; Decarlo, A.; Kozarov, E. Intracellular survival and vascular cell-to-cell transmission of Porphyromonas gingivalis. BMC Microbiol. 2008, 8, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Suh, J.S.; Kim, S.; Boström, K.I.; Wang, C.-Y.; Kim, R.H.; Park, N.-H. Periodontitis-induced systemic inflammation exacerbates atherosclerosis partly via endothelial–mesenchymal transition in mice. Int. J. Oral Sci. 2019, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  45. Lalla, E.; Lamster, I.B.; Hofmann, M.A.; Bucciarelli, L.; Jerud, A.P.; Tucker, S.; Lu, Y.; Papapanou, P.N.; Schmidt, A.M. Oral Infection With a Periodontal Pathogen Accelerates Early Atherosclerosis in Apolipoprotein E–Null Mice. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1405–1411. [Google Scholar] [CrossRef] [PubMed]
  46. Saadi-Thiers, K.; Huck, O.; Simonis, P.; Tilly, P.; Fabre, J.-E.; Tenenbaum, H.; Davideau, J.-L. Periodontal and systemic responses in various mice models of experimental periodontitis: Respective roles of inflammation duration and Porphyromonas gingivalis infection. J. Periodontol. 2013, 84, 396–406. [Google Scholar] [CrossRef]
  47. Chi, H.; Messas, E.; Levine, R.A.; Graves, D.T.; Amar, S. Interleukin-1 Receptor Signaling Mediates Atherosclerosis Associated with Bacterial Exposure and/or a High-Fat Diet in a Murine Apolipoprotein E Heterozygote Model. Circulation 2004, 110, 1678–1685. [Google Scholar] [CrossRef] [Green Version]
  48. Chukkapalli, S.S.; Ambadapadi, S.; Varkoly, K.; Jiron, J.; Aguirre, J.I.; Bhattacharyya, I.; Morel, L.M.; Lucas, A.R.; Kesavalu, L. Impaired innate immune signaling due to combined Toll-like receptor 2 and 4 deficiency affects both periodontitis and atherosclerosis in response to polybacterial infection. Pathog. Dis. 2018, 76. [Google Scholar] [CrossRef]
  49. Huck, O.; Elkaim, R.; Davideau, J.-L.; Tenenbaum, H. Porphyromonas gingivalis and its lipopolysaccharide differentially regulate the expression of cathepsin B in endothelial cells. Mol. Oral Microbiol. 2012. [Google Scholar] [CrossRef]
  50. Madan, M.; Amar, S. Toll-like receptor-2 mediates diet and/or pathogen associated atherosclerosis: Proteomic findings. PLoS ONE 2008, 3, e3204. [Google Scholar] [CrossRef] [Green Version]
  51. Amar, S.; Wu, S.; Madan, M. Is Porphyromonas gingivalis cell invasion required for atherogenesis? Pharmacotherapeutic implications. J. Immunol. 2009, 182, 1584–1592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Cui, D.; Li, H.; Lei, L.; Chen, C.; Yan, F. Nonsurgical periodontal treatment reduced aortic inflammation in ApoE−/− mice with periodontitis. SpringerPlus 2016, 5, 940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Akhi, R.; Wang, C.; Nissinen, A.E.; Kankaanpää, J.; Bloigu, R.; Paju, S.; Mäntylä, P.; Buhlin, K.; Sinisalo, J.; Pussinen, P.J.; et al. Salivary IgA to MAA-LDL and Oral Pathogens Are Linked to Coronary Disease. J. Dent. Res. 2019, 98, 296–303. [Google Scholar] [CrossRef] [PubMed]
  54. Kyrklund, M.; Kummu, O.; Kankaanpää, J.; Akhi, R.; Nissinen, A.; Turunen, S.P.; Pussinen, P.; Wang, C.; Hörkkö, S. Immunization with gingipain A hemagglutinin domain of Porphyromonas gingivalis induces IgM antibodies binding to malondialdehyde-acetaldehyde modified low-density lipoprotein. PLoS ONE 2018, 13, e0191216. [Google Scholar] [CrossRef] [Green Version]
  55. Bugueno, I.M.; Khelif, Y.; Seelam, N.; Morand, D.-N.; Tenenbaum, H.; Davideau, J.-L.; Huck, O. Porphyromonas gingivalis Differentially Modulates Cell Death Profile in Ox-LDL and TNF-α Pre-Treated Endothelial Cells. PLoS ONE 2016, 11, e0154590-18. [Google Scholar] [CrossRef]
  56. O’Connor, C.M.; Dunne, M.W.; Pfeffer, M.A.; Muhlestein, J.B.; Yao, L.; Gupta, S.; Benner, R.J.; Fisher, M.R.; Cook, T.D. Investigators in the WIZARD Study. Azithromycin for the Secondary Prevention of Coronary Heart Disease Events: The WIZARD Study: A Randomized Controlled Trial. JAMA 2003, 290, 1459–1466. [Google Scholar] [CrossRef] [Green Version]
  57. Grayston, J.T.; Kronmal, R.A.; Jackson, L.A.; Parisi, A.F.; Muhlestein, J.B.; Cohen, J.D.; Rogers, W.J.; Crouse, J.R.; Borrowdale, S.L.; Schron, E.; et al. ACES Investigators. Azithromycin for the Secondary Prevention of Coronary Events. N. Engl. J. Med. 2005, 352, 1637–1645. [Google Scholar] [CrossRef] [Green Version]
  58. Cannon, C.P.; Braunwald, E.; McCabe, C.H.; Grayston, J.T.; Muhlestein, B.; Giugliano, R.P.; Cairns, R.; Skene, A.M. Antibiotic treatment of Chlamydia pneumoniae after acute coronary syndrome. N. Engl. J. Med. 2005, 352, 1646–1654. [Google Scholar] [CrossRef] [Green Version]
  59. Jespersen, C.M.; Als-Nielsen, B.; Damgaard, M.; Hansen, J.F.; Hansen, S.; Helø, O.H.; Hildebrandt, P.; Hilden, J.; Jensen, G.B.; Kastrup, J.; et al. Randomised placebo controlled multicentre trial to assess short term clarithromycin for patients with stable coronary heart disease: CLARICOR trial. BMJ 2006, 332, 22–27. [Google Scholar] [CrossRef] [Green Version]
  60. Joensen, J.B.; Juul, S.; Henneberg, E.; Thomsen, G.; Ostergaard, L.; Lindholt, J.S. Can long-term antibiotic treatment prevent progression of peripheral arterial occlusive disease? A large, randomized, double-blinded, placebo-controlled trial. Atherosclerosis 2008, 196, 937–942. [Google Scholar] [CrossRef]
  61. Campbell, L.A.; Rosenfeld, M.E. Infection and Atherosclerosis Development. Arch. Med. Res. 2015, 46, 339–350. [Google Scholar] [CrossRef] [Green Version]
  62. Grassi, W.; Angelis, R.D.; Lamanna, G.; Cervini, C. The clinical features of rheumatoid arthritis. Eur. J. Radiol. 1998, 7, S18–S24. [Google Scholar] [CrossRef]
  63. Mantle, A. The Etiology of Rheumatism Considered from a Bacterial Point of View. Br. Med. J. 1887, 1, 1381–1384. [Google Scholar] [CrossRef]
  64. Li, S.; Yu, Y.; Yue, Y.; Zhang, Z.; Su, K. Microbial Infection and Rheumatoid Arthritis. J. Clin. Cell. Immunol. 2013, 4. [Google Scholar] [CrossRef]
  65. Bingham, C.O., 3rd; Moni, M. Periodontal disease and rheumatoid arthritis: The evidence accumulates for complex pathobiologic interactions. Curr. Opin. Rheumatol. 2013, 25, 345–353. [Google Scholar] [CrossRef] [Green Version]
  66. Rutger, G.P. Rheumatoid arthritis and periodontitis—Inflammatory and infectious connections. Review of the literature. J. Oral Microbiol. 2012, 4. [Google Scholar] [CrossRef]
  67. Chukkapalli, S.; Rivera-Kweh, M.; Gehlot, P.; Velsko, I.; Bhattacharyya, I.; Calise, S.J.; Satoh, M.; Chan, E.K.L.; Holoshitz, J.; Kesavalu, L. Periodontal bacterial colonization in synovial tissues exacerbates collagen-induced arthritis in B10.RIII mice. Arthritis Res. Ther. 2016, 18, 161. [Google Scholar] [CrossRef] [Green Version]
  68. Okumura, G.; Kondo, N.; Sato, K.; Yamazaki, K.; Ohshima, H.; Kawashima, H.; Ogose, A.; Endo, N. Experimental arthritis and Porphyromonas gingivalis administration synergistically decrease bone regeneration in femoral cortical defects. Sci. Rep. 2019, 9, 1–12. [Google Scholar] [CrossRef]
  69. Huck, O.; You, J.; Han, X.; Cai, B.; Panek, J.; Amar, S. Reduction of Articular and Systemic Inflammation by Kava-241 in Porphyromonas gingivalis-induced Arthritis Murine Model. Infect. Immun. 2018, 86. [Google Scholar] [CrossRef] [Green Version]
  70. Sugawara, M.; Yamashita, K.; Yoshie, H.; Hara, K. Detection of, and anti-collagen antibody produced by, CD5-positive B cells in inflamed gingival tissues. J. Periodontal Res. 1992, 27, 489–498. [Google Scholar] [CrossRef]
  71. Harre, U.; Georgess, D.; Bang, H.; Bozec, A.; Axmann, R.; Ossipova, E.; Jakobsson, P.-J.; Baum, W.; Nimmerjahn, F.; Szarka, E.; et al. Induction of osteoclastogenesis and bone loss by human autoantibodies against citrullinated vimentin. J. Clin. Investig. 2012, 122, 1791–1802. [Google Scholar] [CrossRef]
  72. Elkaim, R.; Bugueno-Valdebenito, I.M.; Benkirane-Jessel, N.; Tenenbaum, H. Porphyromonas gingivalis and its LPS differentially regulate the expression of peptidyl arginine deiminases in human chondrocytes. Innate Immun. 2017, 23, 468–475. [Google Scholar] [CrossRef]
  73. Maresz, K.J.; Hellvard, A.; Sroka, A.; Adamowicz, K.; Bielecka, E.; Koziel, J.; Gawron, K.; Mizgalska, D.; Marcinska, K.A.; Benedyk, M.; et al. Porphyromonas gingivalis facilitates the development and progression of destructive arthritis through its unique bacterial peptidylarginine deiminase (PAD). PLoS Pathog. 2013, 9, e1003627. [Google Scholar] [CrossRef]
  74. Larsen, D.N.; Mikkelsen, C.E.; Kierkegaard, M.; Bereta, G.P.; Nowakowska, Z.; Kaczmarek, J.Z.; Potempa, J.; Højrup, P. Citrullinome of Porphyromonas gingivalis Outer Membrane Vesicles: Confident Identification of Citrullinated Peptides. Mol. Cell. Proteom. 2020, 19, 167–180. [Google Scholar] [CrossRef]
  75. Casiano-Colón, A.; Marquis, R.E. Role of the arginine deiminase system in protecting oral bacteria and an enzymatic basis for acid tolerance. Appl. Environ. Microbiol. 1988, 54, 1318–1324. [Google Scholar] [CrossRef] [Green Version]
  76. Niederman, R.; Brunkhorst, B.; Smith, S.; Weinreb, R.N.; Ryder, M.I. Ammonia as a potential mediator of adult human periodontal infection: Inhibition of neutrophil function. Arch. Oral Biol. 1990, 35, S205–S209. [Google Scholar] [CrossRef]
  77. Dahlen, G.; Basic, A.; Bylund, J. Importance of Virulence Factors for the Persistence of Oral Bacteria in the Inflamed Gingival Crevice and in the Pathogenesis of Periodontal Disease. J. Clin. Med. 2019, 8, 1339. [Google Scholar] [CrossRef] [Green Version]
  78. Jung, H.; Jung, S.M.; Rim, Y.A.; Park, N.; Nam, Y.; Lee, J.; Park, S.-H.; Ju, J.H. Arthritic role of Porphyromonas gingivalis in collagen-induced arthritis mice. PLoS ONE 2017, 12, e0188698. [Google Scholar] [CrossRef] [Green Version]
  79. Peng, H.-Y.; Chen, S.-Y.; Siao, S.-H.; Chang, J.T.; Xue, T.-Y.; Lee, Y.-H.; Jan, M.-S.; Tsay, G.J.; Zouali, M. Targeting a cysteine protease from a pathobiont alleviates experimental arthritis. Arthritis Res. Ther. 2020, 22, 114. [Google Scholar] [CrossRef]
  80. Kharlamova, N.; Jiang, X.; Sherina, N.; Potempa, B.; Israelsson, L.; Quirke, A.M.; Eriksson, K.; Yucel-Lindberg, T.; Venables, P.J.; Potempa, J.; et al. Antibodies to Porphyromonas gingivalis Indicate Interaction between Oral Infection, Smoking, and Risk Genes in Rheumatoid Arthritis Etiology. Arthritis Rheumatol. 2016, 68, 604–613. [Google Scholar] [CrossRef] [Green Version]
  81. Carmona, L.; Cross, M.; Williams, B.; Lassere, M.; March, L. Rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2010, 24, 733–745. [Google Scholar] [CrossRef] [PubMed]
  82. Kononoff, A.; Elfving, P.; Pussinen, P.; Hörkkö, S.; Kautiainen, H.; Arstila, L.; Laasonen, L.; Savolainen, E.; Niinisalo, H.; Rutanen, J.; et al. Association of rheumatoid arthritis disease activity and antibodies to periodontal bacteria with serum lipoprotein profile in drug naive patients. Ann. Med. 2020, 52, 32–42. [Google Scholar] [CrossRef]
  83. de Molon, R.S.; Rossa, C., Jr.; Thurlings, R.M.; Cirelli, J.A.; Koenders, M.I. Linkage of Periodontitis and Rheumatoid Arthritis: Current Evidence and Potential Biological Interactions. Int. J. Mol. Sci. 2019, 20, 4541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Białowąs, K.; Radwan-Oczko, M.; Duś-Ilnicka, I.; Korman, L.; Świerkot, J. Periodontal disease and influence of periodontal treatment on disease activity in patients with rheumatoid arthritis and spondyloarthritis. Rheumatol. Int. 2020, 40, 455–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Keller, A.; Rohde, J.F.; Raymond, K.; Heitmann, B.L. The Association between Periodontal Disease and Overweight and Obesity: A Systematic Review. J. Periodontol. 2015, 1–15. [Google Scholar] [CrossRef] [PubMed]
  86. Le Sage, F.; Meilhac, O.; Gonthier, M.-P. Porphyromonas gingivalis lipopolysaccharide induces pro-inflammatory adipokine secretion and oxidative stress by regulating Toll-like receptor-mediated signaling pathways and redox enzymes in adipocytes. Mol. Cell. Endocrinol. 2017, 446, 102–110. [Google Scholar] [CrossRef]
  87. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  88. Richard, G.; Trivedi, N.; Belta, C.; Amar, S. Partial restoration of macrophage alteration from diet-induced obesity in response to Porphyromonas gingivalis infection. PLoS ONE 2013, 8, e70320. [Google Scholar] [CrossRef] [Green Version]
  89. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [Google Scholar] [CrossRef]
  90. Tilg, H.; Moschen, A.R. Microbiota and diabetes: An evolving relationship. Gut 2014, 63, 1513–1521. [Google Scholar] [CrossRef] [PubMed]
  91. Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia Muciniphila Protects against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation 2016, 133, 2434–2446. [Google Scholar] [CrossRef] [Green Version]
  92. Baquero, F.; Nombela, C. The microbiome as a human organ. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2012, 18, 2–4. [Google Scholar] [CrossRef] [Green Version]
  93. Kundu, P.; Blacher, E.; Elinav, E.; Pettersson, S. Our Gut Microbiome: The Evolving Inner. Self. Cell 2017, 171, 1481–1493. [Google Scholar] [CrossRef] [PubMed]
  94. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
  95. Pedersen, A.M.L.; Sørensen, C.E.; Proctor, G.B.; Carpenter, G.H.; Ekström, J. Salivary secretion in health and disease. J. Oral Rehabil. 2018, 45, 730–746. [Google Scholar] [CrossRef] [PubMed]
  96. Saygun, I.; Nizam, N.; Keskiner, I.; Bal, V.; Kubar, A.; Açıkel, C.; Serdar, M.; Slots, J. Salivary infectious agents and periodontal disease status. J. Periodontal Res. 2011, 46, 235–239. [Google Scholar] [CrossRef] [PubMed]
  97. Arimatsu, K.; Yamada, H.; Miyazawa, H.; Minagawa, T.; Nakajima, M.; Ryder, M.I.; Gotoh, K.; Motooka, D.; Nakamura, S.; Iida, T.; et al. Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota. Sci. Rep. 2014, 4, 4828. [Google Scholar] [CrossRef] [Green Version]
  98. Lourenςo, T.G.B.; Spencer, S.J.; Alm, E.J.; Colombo, A.P. Defining the gut microbiota in individuals with periodontal diseases: An exploratory study. J. Oral Microbiol. 2018, 10, 1487741. [Google Scholar] [CrossRef] [Green Version]
  99. Kato, T.; Yamazaki, K.; Nakajima, M.; Date, Y.; Kikuchi, J.; Hase, K.; Ohno, H.; Yamazaki, K.; D’Orazio, S.E.F. Oral Administration of Porphyromonas gingivalis Alters the Gut Microbiome and Serum Metabolome. mSphere 2018, 3, e00460-18. [Google Scholar] [CrossRef] [Green Version]
  100. Nakajima, M.; Arimatsu, K.; Kato, T.; Matsuda, Y.; Minagawa, T.; Takahashi, N.; Ohno, H.; Yamazaki, K. Oral Administration of P. gingivalis Induces Dysbiosis of Gut Microbiota and Impaired Barrier Function Leading to Dissemination of Enterobacteria to the Liver. PLoS ONE 2015, 10, e0134234. [Google Scholar] [CrossRef] [Green Version]
  101. Matsuda, Y.; Kato, T.; Takahashi, N.; Nakajima, M.; Arimatsu, K.; Minagawa, T.; Sato, K.; Ohno, H.; Yamazaki, K. Ligature-induced periodontitis in mice induces elevated levels of circulating interleukin-6 but shows only weak effects on adipose and liver tissues. J. Periodontal. Res. 2016, 51, 639–646. [Google Scholar] [CrossRef] [PubMed]
  102. Nguyen, T.L.; Vieira-Silva, S.; Liston, A.; Raes, J. How informative is the mouse for human gut microbiota research? Dis. Model. Mech. 2015, 8, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Segata, N.; Haake, S.K.; Mannon, P.; Lemon, K.P.; Waldron, L.; Gevers, D.; Huttenhower, C.; Izard, J. Composition of the adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and stool samples. Genome Biol. 2012, 13, R42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Eckel, R.H.; Grundy, S.M.; Zimmet, P.Z. The metabolic syndrome. Lancet 2005, 365, 1415–1428. [Google Scholar] [CrossRef]
  105. Cani, P.D.; Osto, M.; Geurts, L.; Everard, A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 2012, 3, 279–288. [Google Scholar] [CrossRef] [Green Version]
  106. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
  107. Amar, S.; Zhou, Q.; Shaik-Dasthagirisaheb, Y.; Leeman, S. Diet-induced obesity in mice causes changes in immune responses and bone loss manifested by bacterial challenge. Proc. Natl. Acad. Sci. USA 2007, 104, 20466–20471. [Google Scholar] [CrossRef] [Green Version]
  108. Zhou, Q.; Leeman, S.E.; Amar, S. Signaling mechanisms in the restoration of impaired immune function due to diet-induced obesity. Proc. Natl. Acad. Sci. USA 2011, 108, 2867–2872. [Google Scholar] [CrossRef] [Green Version]
  109. Zhou, Q.; Leeman, S.E.; Amar, S. Signaling mechanisms involved in altered function of macrophages from diet-induced obese mice affect immune responses. Proc. Natl. Acad. Sci. USA 2009, 106, 10740–10745. [Google Scholar] [CrossRef] [Green Version]
  110. Blasco-Baque, V.; Garidou, L.; Pomié, C.; Escoula, Q.; Loubieres, P.; Le Gall-David, S.; Lemaitre, M.; Nicolas, S.; Klopp, P.; Waget, A.; et al. Periodontitis induced by Porphyromonas gingivalis drives periodontal microbiota dysbiosis and insulin resistance via an impaired adaptive immune response. Gut 2017, 66, 872–885. [Google Scholar] [CrossRef] [Green Version]
  111. Grossi, S.G.; Skrepcinski, F.B.; DeCaro, T.; Robertson, D.C.; Ho, A.W.; Dunford, R.G.; Genco, R.J. Treatment of Periodontal Disease in Diabetics Reduces Glycated Hemoglobin. J. Periodontol. 1997, 68, 713–719. [Google Scholar] [CrossRef] [PubMed]
  112. Kıran, M.; Arpak, N.; Ünsal, E.; Erdoğan, M.F. The effect of improved periodontal health on metabolic control in type 2 diabetes mellitus. J. Clin. Periodontol. 2005, 32, 266–272. [Google Scholar] [CrossRef] [PubMed]
  113. Altay, U.; Gürgan, C.A.; Ağbaht, K. Changes in Inflammatory and Metabolic Parameters after Periodontal Treatment in Patients with and without Obesity. J. Periodontol. 2013, 84, 13–23. [Google Scholar] [CrossRef] [PubMed]
  114. Lang, N.P.; Salvi, G.E.; Sculean, A. Nonsurgical therapy for teeth and implants-When and why? Periodontology 2019, 79, 15–21. [Google Scholar] [CrossRef] [PubMed]
  115. Almeida, M.L.; Duarte, P.M.; Figueira, E.A.; Lemos, J.C.; Nobre, C.M.G.; Miranda, T.S.; de Vasconcelos Gurgel, B.C. Effects of a full-mouth disinfection protocol on the treatment of type-2 diabetic and non-diabetic subjects with mild-to-moderate periodontitis: One-year clinical outcomes. Clin. Oral Investig. 2019. [Google Scholar] [CrossRef]
  116. Harmouche, L.; Courval, A.; Mathieu, A.; Petit, C.; Huck, O.; Séverac, F.; Davideau, J.-L. Impact of tooth-related factors on photodynamic therapy effectiveness during active periodontal therapy: A 6-months split-mouth randomized clinical trial. Photodiagnosis Photodyn. Ther. 2019, 27, 167–172. [Google Scholar] [CrossRef]
  117. Mombelli, A.; Almaghlouth, A.; Cionca, N.; Courvoisier, D.S.; Giannopoulou, C. Differential benefits of amoxicillin-metronidazole in different phases of periodontal therapy in a randomized controlled crossover clinical trial. J. Periodontol. 2015, 86, 367–375. [Google Scholar] [CrossRef] [Green Version]
  118. Leininger, M.; Tenenbaum, H.; Davideau, J.-L. Modified periodontal risk assessment score: Long-term predictive value of treatment outcomes. Retrosp. Study 2010, 37, 427–435. [Google Scholar] [CrossRef]
  119. Petit, C.; Schmeltz, S.; Burgy, A.; Tenenbaum, H.; Huck, O.; Davideau, J.-L. Risk factors associated with long-term outcomes after active and supporting periodontal treatments: Impact of various compliance definitions on tooth loss. Clin. Oral Investig. 2019, 62, 218–219. [Google Scholar] [CrossRef]
  120. Huck, O.; Mulhall, H.; Rubin, G.; Kizelnik, Z.; Iyer, R.; Perpich, J.D.; Haque, N.; Cani, P.D.; de Vos, W.M.; Amar, S. Akkermansia muciniphila reduces Porphyromonas gingivalis-induced inflammation and periodontal bone destruction. J. Clin. Periodontol. 2019. [Google Scholar] [CrossRef]
  121. Martin-Cabezas, R.; Davideau, J.-L.; Tenenbaum, H.; Huck, O. Clinical efficacy of probiotic as an adjunctive therapy to non-surgical periodontal treatment of chronic periodontitis: A systematic review and meta-analysis. J. Clin. Periodontol. 2016, 43. [Google Scholar] [CrossRef] [PubMed]
  122. Geraldo, B.M.C.; Batalha, M.N.; Milhan, N.V.M.; Rossoni, R.D.; Scorzoni, L.; Anbinder, A.L. Heat-killed Lactobacillus reuteri and cell-free culture supernatant have similar effects to viable probiotics during interaction with Porphyromonas gingivalis. J. Periodontal Res. 2019. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, M.; Liu, Y.; Song, Y.; Gao, Y.; Zhao, F.; Luo, Y.; Qian, F.; Mu, G.; Tuo, Y. The ameliorative effect of Lactobacillus plantarum-12 on DSS-induced murine colitis. Food Funct. 2020. [Google Scholar] [CrossRef] [PubMed]
  124. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Plovier, H.; Everard, A.; Druart, C.; Depommier, C.; Van Hul, M.; Geurts, L.; Chilloux, J.; Ottman, N.; Duparc, T.; Lichtenstein, L.; et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 2017, 23, 107–113. [Google Scholar] [CrossRef] [Green Version]
  126. Everard, A.; Plovier, H.; Rastelli, M.; Van Hul, M.; de Wouters d’Oplinter, A.; Geurts, L.; Druart, C.; Robine, S.; Delzenne, N.M.; Muccioli, G.G.; et al. Intestinal epithelial N-acylphosphatidylethanolamine phospholipase D links dietary fat to metabolic adaptations in obesity and steatosis. Nat. Commun. 2019, 10, 457. [Google Scholar] [CrossRef]
  127. Zhai, R.; Xue, X.; Zhang, L.; Yang, X.; Zhao, L.; Zhang, C. Strain-Specific Anti-inflammatory Properties of Two Akkermansia muciniphila Strains on Chronic Colitis in Mice. Front. Cell. Infect. Microbiol. 2019, 9, 239. [Google Scholar] [CrossRef]
  128. Cani, P.D.; de Vos, W.M. Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef]
  129. Ottman, N.; Reunanen, J.; Meijerink, M.; Pietilä, T.E.; Kainulainen, V.; Klievink, J.; Huuskonen, L.; Aalvink, S.; Skurnik, M.; Boeren, S.; et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS ONE 2017, 12, e0173004. [Google Scholar] [CrossRef]
  130. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
Figure 1. Porphyromonas gingivalis influences the development of multiple chronic inflammatory conditions. Through the cross-reactive antibodies (atherothrombosis, rheumatoid arthritis), increased levels of systemic inflammation (atherothrombosis, rheumatoid arthritis, gut microbiome dysbiosis, metabolic disorders), as well as overall microbiome dysbiosis. (↑ = increase ↓= decrease).
Figure 1. Porphyromonas gingivalis influences the development of multiple chronic inflammatory conditions. Through the cross-reactive antibodies (atherothrombosis, rheumatoid arthritis), increased levels of systemic inflammation (atherothrombosis, rheumatoid arthritis, gut microbiome dysbiosis, metabolic disorders), as well as overall microbiome dysbiosis. (↑ = increase ↓= decrease).
Microorganisms 08 00869 g001

Share and Cite

MDPI and ACS Style

Mulhall, H.; Huck, O.; Amar, S. Porphyromonas gingivalis, a Long-Range Pathogen: Systemic Impact and Therapeutic Implications. Microorganisms 2020, 8, 869. https://doi.org/10.3390/microorganisms8060869

AMA Style

Mulhall H, Huck O, Amar S. Porphyromonas gingivalis, a Long-Range Pathogen: Systemic Impact and Therapeutic Implications. Microorganisms. 2020; 8(6):869. https://doi.org/10.3390/microorganisms8060869

Chicago/Turabian Style

Mulhall, Hannah, Olivier Huck, and Salomon Amar. 2020. "Porphyromonas gingivalis, a Long-Range Pathogen: Systemic Impact and Therapeutic Implications" Microorganisms 8, no. 6: 869. https://doi.org/10.3390/microorganisms8060869

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop