Local and systemic mechanisms linking periodontal disease and inflammatory comorbidities

George Hajishengallis, Triantafyllos Chavakis, George Hajishengallis, Triantafyllos Chavakis

Abstract

Periodontitis, a major inflammatory disease of the oral mucosa, is epidemiologically associated with other chronic inflammation-driven disorders, including cardio-metabolic, neurodegenerative and autoimmune diseases and cancer. Emerging evidence from interventional studies indicates that local treatment of periodontitis ameliorates surrogate markers of comorbid conditions. The potential causal link between periodontitis and its comorbidities is further strengthened by recent experimental animal studies establishing biologically plausible and clinically consistent mechanisms whereby periodontitis could initiate or aggravate a comorbid condition. This multi-faceted 'mechanistic causality' aspect of the link between periodontitis and comorbidities is the focus of this Review. Understanding how certain extra-oral pathologies are affected by disseminated periodontal pathogens and periodontitis-associated systemic inflammation, including adaptation of bone marrow haematopoietic progenitors, may provide new therapeutic options to reduce the risk of periodontitis-associated comorbidities.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Periodontal disease and associated inflammatory…
Fig. 1. Periodontal disease and associated inflammatory comorbidities.
On the basis of epidemiological, clinical intervention and animal model-based studies, periodontitis has been linked with a number of comorbid conditions, such as those indicated. Mechanistically, periodontitis is associated with bacteraemias and systemic inflammation, which can induce acute-phase responses as well as metabolic and inflammatory alterations in the liver and bone marrow, activities that can influence comorbid conditions. Moreover, periodontal bacteria can disseminate by different routes — haematogenous, oro-pharyngeal and oro-digestive — to reach extra-oral sites where they can cause or exacerbate inflammatory pathologies.
Fig. 2. Periodontal bacterial translocation leading to…
Fig. 2. Periodontal bacterial translocation leading to bacteraemia and alterations in the bone marrow that promote osteoclastogenesis in different sites.
Periodontal bacteria may translocate through the ulcerated epithelium of the periodontal pockets into the circulation, causing bacteraemia and systemic inflammation. Blood-borne Porphyromonas gingivalis causes an increase in the serum levels of IL-6, which in turn induces the expansion of an osteoclast precursor population (defined as CD11b+CSF1R+Ly6Chi) in the bone marrow. This osteoclast precursor population displays enhanced osteoclastogenic lineage bias and populates various bone resorption sites, where it can differentiate into mature osteoclasts, in response to locally produced receptor activator of NF-κB ligand (RANKL) (for example, produced by osteoblasts stimulated by T helper 17 (TH17) cell-derived IL-17). This concept may be a mechanism whereby the bone marrow might link periodontitis to other bone loss disorders, such as rheumatoid arthritis.
Fig. 3. Trained myelopoiesis in the bone…
Fig. 3. Trained myelopoiesis in the bone marrow as a mechanistic basis of inflammatory comorbidities.
Periodontitis-associated bacteraemias and systemic inflammation, for example, inflammatory cytokines, such as IL-1β, can be sensed in the bone marrow, leading to induction of long-term metabolic and epigenetic rewiring in haematopoietic stem cells (HSCs). These inflammation-adapted HSCs proliferate and preferentially undergo myeloid-biased differentiation, leading to expansion of multipotent progenitors (MPPs) with myeloid potential and granulocyte–macrophage progenitors (GMPs) and ultimately production of ‘trained’ myeloid cell populations. These hyper-reactive neutrophils or monocytes/macrophages can be recruited to the periodontium and other sites of infection, inflammation or injury, thereby potentially exacerbating periodontitis and promoting inflammatory pathology of comorbid conditions such as cardiovascular disease.
Fig. 4. Oral–gut axis mechanisms that promote…
Fig. 4. Oral–gut axis mechanisms that promote colitis.
Periodontitis-associated pathobionts can reach the gut through the oro-digestive route (owing to constant saliva swallowing) and can promote colitis in susceptible hosts, in part through induction of IL-1β by inflammatory macrophages. Oral pathobiont-reactive T cells (enriched in T helper 17 (TH17) cells), which expand during periodontitis, migrate through the lymphatics to the gut, where they are activated by the ectopically colonized oral pathobionts upon their processing by antigen-presenting cells (APCs). The oral pathobiont-induced elevation of IL-1β contributes to the activation and proliferation of the transmigrated TH17 cells, which become colitogenic and exacerbate intestinal inflammation. Thus, ectopically colonized oral pathobionts may exacerbate colitis by activating both innate immunity (local inflammatory macrophages) and adaptive immunity (transmigrated TH17 cells of oral origin).
Fig. 5. Mechanisms of periodontal bacterial action…
Fig. 5. Mechanisms of periodontal bacterial action in extra-oral pathologies.
a | Porphyromonas gingivalis causes vascular endothelial barrier disruption and increased permeability by disrupting intercellular junctions. This action is mediated by its gingipain proteases, which degrade platelet endothelial cell adhesion molecule (PECAM1) and vascular endothelial cadherin (VE-cadherin), which are crucial for junctional integrity. Endothelial damage and elevated permeability may instigate several processes that potentially induce or exacerbate atherogenesis, including induction of pro-inflammatory cytokines, platelet aggregation and increased leukocyte extravasation to subendothelial areas. b | P. gingivalis-induced pathology in Alzheimer disease. P. gingivalis DNA and gingipains have been detected in brain autopsies of patients with Alzheimer disease as well as in the brain of mice orally infected with this pathogen. The presence of P. gingivalis in the mouse brain is associated with increased levels of amyloid-β, complement activation and neuroinflammation. The gingipains of the pathogen cleave the microtubule-associated protein tau, an activity that promotes aberrant phosphorylation of tau and accumulation of misfolded insoluble tau in Alzheimer disease. c | Role of periodontal bacteria in the generation of anti-citrullinated protein antibodies (ACPAs). P. gingivalis expresses a unique (among prokaryotic organisms) peptidyl-arginine deiminase (PPAD), which can citrullinate proteins including host proteins. The PPAD activity is facilitated by the pathogen’s arginine-specific gingipains, which cleave proteins and expose C-terminal arginine residues for citrullination by PPAD. Alternatively, Aggregatibacter actinomycetemcomitans indirectly causes host protein citrullination by secreting leukotoxin A (LtxA), a pore-forming toxin that induces calcium influx and hyperactivation of PAD enzymes in neutrophils, as well as cytolysis (by NETosis, a form of cell death typified by the release of decondensed chromatin and granular contents to the extracellular milieu), thereby releasing the generated citrullinated autoantigens. Thus, through distinct mechanisms, both pathogens can contribute to the generation of the rheumatoid arthritis-specific ACPAs that promote disease in individuals with HLA-DRB1 shared epitope (SE) alleles. APC, antigen-presenting cell.

References

    1. Genco RJ, Sanz M. Clinical and public health implications of periodontal and systemic diseases: an overview. Periodontol. 2000. 2020;83:7–13.
    1. Potempa J, Mydel P, Koziel J. The case for periodontitis in the pathogenesis of rheumatoid arthritis. Nat. Rev. Rheumatol. 2017;13:606–620.
    1. Hajishengallis G. Periodontitis: from microbial immune subversion to systemic inflammation. Nat. Rev. Immunol. 2015;15:30–44.
    1. Acharya C, Sahingur SE, Bajaj JS. Microbiota, cirrhosis, and the emerging oral-gut-liver axis. JCI Insight. 2017;2:e94416.
    1. Schenkein HA, Papapanou PN, Genco R, Sanz M. Mechanisms underlying the association between periodontitis and atherosclerotic disease. Periodontol. 2000. 2020;83:90–106.
    1. Kitamoto S, et al. The intermucosal connection between the mouth and gut in commensal pathobiont-driven colitis. Cell. 2020;182:447–462.
    1. Abed J, et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc. Cell Host Microbe. 2016;20:215–225.
    1. Atarashi K, et al. Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation. Science. 2017;358:359–365.
    1. Blasco-Baque V, 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.
    1. Gur C, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42:344–355.
    1. Dominy SS, et al. Porphyromonas gingivalis in Alzheimer’s disease brains: evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 2019;5:eaau3333.
    1. Konig MF, et al. Aggregatibacter actinomycetemcomitans-induced hypercitrullination links periodontal infection to autoimmunity in rheumatoid arthritis. Sci. Transl. Med. 2016;8:369ra176.
    1. Meghil MM, et al. Disruption of immune homeostasis in human dendritic cells via regulation of autophagy and apoptosis by Porphyromonas gingivalis. Front. Immunol. 2019;10:2286.
    1. Sato K, et al. Aggravation of collagen-induced arthritis by orally administered Porphyromonas gingivalis through modulation of the gut microbiota and gut immune system. Sci. Rep. 2017;7:6955.
    1. Farrugia C, et al. Mechanisms of vascular damage by systemic dissemination of the oral pathogen Porphyromonas gingivalis. FEBS J. 2020 doi: 10.1111/febs.15486.
    1. Zhao Y, et al. Characterization and regulation of osteoclast precursors following chronic Porphyromonas gingivalis infection. J. Leukoc. Biol. 2020;108:1037–1050.
    1. Netea MG, et al. Defining trained immunity and its role in health and disease. Nat. Rev. Immunol. 2020;20:375–388.
    1. Chavakis T, Mitroulis I, Hajishengallis G. Hematopoietic progenitor cells as integrative hubs for adaptation to and fine-tuning of inflammation. Nat. Immunol. 2019;20:802–811.
    1. D’Aiuto F, et al. Systemic effects of periodontitis treatment in patients with type 2 diabetes: a 12 month, single-centre, investigator-masked, randomised trial. Lancet Diabetes Endocrinol. 2018;6:954–965.
    1. Ishai A, et al. Periodontal disease associates with arterial inflammation via potentiation of a hematopoietic–arterial axis. JACC Cardiovasc. Imaging. 2019;12:2271–2273.
    1. Teles R, Wang C-Y. Mechanisms involved in the association between peridontal diseases and cardiovascular disease. Oral Dis. 2011;17:450–461.
    1. Bajaj JS, et al. Periodontal therapy favorably modulates the oral-gut-hepatic axis in cirrhosis. Am. J. Physiol. Gastrointest. Liver Physiol. 2018;315:G824–G837.
    1. Xiao E, et al. Diabetes enhances IL-17 expression and alters the oral microbiome to increase its pathogenicity. Cell Host Microbe. 2017;22:120–128 e124.
    1. Teles F, Wang Y, Hajishengallis G, Hasturk H, Marchesan J. Impact of systemic factors in shaping the periodontal microbiome. Periodontol. 2000. 2021;85:126–160.
    1. Monsarrat P, et al. Clinical research activity in periodontal medicine: a systematic mapping of trial registers. J. Clin. Periodontol. 2016;43:390–400.
    1. Rydén L, et al. Periodontitis increases the risk of a first myocardial infarction. Circulation. 2016;133:576–583.
    1. Rodríguez-Lozano B, et al. Association between severity of periodontitis and clinical activity in rheumatoid arthritis patients: a case–control study. Arthritis Res. Ther. 2019;21:27.
    1. Genco RJ, Van Dyke TE. Prevention: reducing the risk of CVD in patients with periodontitis. Nat. Rev. Cardiol. 2010;7:479–480.
    1. D’Aiuto F, Orlandi M, Gunsolley JC. Evidence that periodontal treatment improves biomarkers and CVD outcomes. J. Clin. Periodontol. 2013;40:S85–S105.
    1. H. Bokhari SA, et al. Non-surgical periodontal therapy reduces coronary heart disease risk markers: a randomized controlled trial. J. Clin. Periodontol. 2012;39:1065–1074.
    1. de Oliveira C, Watt R, Hamer M. Toothbrushing, inflammation, and risk of cardiovascular disease: results from Scottish health survey. Br. Med. J. 2010;340:c2451.
    1. Tonetti MS. Periodontitis and risk for atherosclerosis: an update on intervention trials. J. Clin. Periodontol. 2009;36:15–19.
    1. Türer Ç C, Durmuş D, Balli U, Güven B. Effect of non-surgical periodontal treatment on gingival crevicular fluid and serum endocan, vascular endothelial growth factor-A, and tumor necrosis factor-alpha levels. J. Periodontol. 2017;88:493–501.
    1. Mammen MJ, Scannapieco FA, Sethi S. Oral–lung microbiome interactions in lung diseases. Periodontol. 2000. 2020;83:234–241.
    1. Qin N, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature. 2014;513:59–64.
    1. Arimatsu K, et al. Oral pathobiont induces systemic inflammation and metabolic changes associated with alteration of gut microbiota. Sci. Rep. 2014;4:4828.
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–867.
    1. Norata GD, et al. The cellular and molecular basis of translational immunometabolism. Immunity. 2015;43:421–434.
    1. Orlandi M, Graziani F, D’Aiuto F. Periodontal therapy and cardiovascular risk. Periodontol. 2000. 2020;83:107–124.
    1. Genco RJ, Graziani F, Hasturk H. Effects of periodontal disease on glycemic control, complications, and incidence of diabetes mellitus. Periodontol. 2000. 2020;83:59–65.
    1. Jepsen S, Suvan J, Deschner J. The association of periodontal diseases with metabolic syndrome and obesity. Periodontol. 2000. 2020;83:125–153.
    1. Tonetti MS, et al. Treatment of periodontitis and endothelial function. N. Engl. J. Med. 2007;356:911–920.
    1. Desvarieux M, et al. Changes in clinical and microbiological periodontal profiles relate to progression of carotid intima-media thickness: the oral infections and vascular disease epidemiology study. J. Am. Heart Assoc. 2013;2:e000254.
    1. Yoneda M, et al. Involvement of a periodontal pathogen, Porphyromonas gingivalis on the pathogenesis of non-alcoholic fatty liver disease. BMC Gastroenterol. 2012;12:16.
    1. Helenius-Hietala J, et al. Periodontitis is associated with incident chronic liver disease — a population-based cohort study. Liver Int. 2019;39:583–591.
    1. Brito LCW, et al. Experimental periodontitis promotes transient vascular inflammation and endothelial dysfunction. Arch. Oral. Biol. 2013;58:1187–1198.
    1. Matsuda Y, et al. Ligature-induced periodontitis in mice induces elevated levels of circulating interleukin-6 but shows only weak effects on adipose and liver tissues. J. Periodont. Res. 2016;51:639–646.
    1. O’Boyle C, et al. Ligature-induced periodontitis induces systemic inflammation but does not alter acute outcome after stroke in mice. Int. J. Stroke. 2019;15:175–187.
    1. Anbinder AL, et al. Periodontal disease exacerbates systemic ovariectomy-induced bone loss in mice. Bone. 2016;83:241–247.
    1. Miyajima S-i, et al. Periodontitis-activated monocytes/macrophages cause aortic inflammation. Sci. Rep. 2014;4:5171.
    1. Hasturk H, et al. Resolvin E1 (RvE1) attenuates atherosclerotic plaque formation in diet and inflammation-induced atherogenesis. Arterioscl. Thromb. Vasc. Biol. 2015;35:1123–1133.
    1. Tian J, et al. Porphyromonas gingivalis induces insulin resistance by increasing BCAA levels in mice. J. Dent. Res. 2020;99:839–846.
    1. White PJ, Newgard CB. Branched-chain amino acids in disease. Science. 2019;363:582–583.
    1. Chavakis T, et al. The pattern recognition receptor (RAGE) is a counterreceptor for leukocyte integrins: a novel pathway for inflammatory cell recruitment. J. Exp. Med. 2003;198:1507–1515.
    1. Ruiz HH, Ramasamy R, Schmidt AM. Advanced glycation end products: building on the concept of the “Common Soil” in metabolic disease. Endocrinology. 2020;161:1–10.
    1. Lalla E, et al. Blockade of RAGE suppresses periodontitis-associated bone loss in diabetic mice. J. Clin. Invest. 2000;105:1117–1124.
    1. Nakahara T, et al. Involvement of Porphyromonas gingivalis in the progression of non-alcoholic fatty liver disease. J. Gastroenterol. 2018;53:269–280.
    1. Nagasaki A, et al. Odontogenic infection by Porphyromonas gingivalis exacerbates fibrosis in NASH via hepatic stellate cell activation. Sci. Rep. 2020;10:4134.
    1. Vasconcelos DFP, et al. Decrease of pericytes is associated with liver disease caused by ligature-induced periodontitis in rats. J. Periodontol. 2017;88:e49–e57.
    1. Tsukasaki M, et al. Host defense against oral microbiota by bone-damaging T cells. Nat. Commun. 2018;9:701.
    1. Komazaki R, et al. Periodontal pathogenic bacteria, Aggregatibacter actinomycetemcomitans affect non-alcoholic fatty liver disease by altering gut microbiota and glucose metabolism. Sci. Rep. 2017;7:13950.
    1. King KY, Goodell MA. Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response. Nat. Rev. Immunol. 2011;11:685.
    1. Yamamoto R, et al. Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells. Cell. 2013;154:1112–1126.
    1. Tsukasaki M, Takayanagi H. Osteoimmunology: evolving concepts in bone-immune interactions in health and disease. Nat. Rev. Immunol. 2019;19:626–642.
    1. Herrera BS, et al. Peripheral blood mononuclear phagocytes from patients with chronic periodontitis are primed for osteoclast formation. J. Periodontol. 2014;85:e72–e81.
    1. Manz MG, Boettcher S. Emergency granulopoiesis. Nat. Rev. Immunol. 2014;14:302–314.
    1. Arts RJW, et al. BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe. 2018;23:89–100.
    1. Mitroulis I, et al. Modulation of myelopoiesis progenitors is an integral component of trained immunity. Cell. 2018;172:147–161.
    1. Pietras EM, et al. Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal. Nat. Cell Biol. 2016;18:607–618.
    1. Fifer KM, et al. Positron emission tomography measurement of periodontal 18F-fluorodeoxyglucose uptake is associated with histologically determined carotid plaque inflammation. J. Am. Coll. Cardiol. 2011;57:971–976.
    1. Ling MR, Chapple IL, Matthews JB. Peripheral blood neutrophil cytokine hyper-reactivity in chronic periodontitis. Innate Immun. 2015;21:714–725.
    1. Radvar M, Tavakkol-Afshari J, Bajestan MN, Naseh MR, Arab HR. The effect of periodontal treatment on IL-6 production of peripheral blood monocytes in aggressive periodontitis and chronic periodontitis patients. Iran. J. Immunol. 2008;5:100–106.
    1. Kleinnijenhuis J, et al. Bacille Calmette–Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc. Natl Acad. Sci. USA. 2012;109:17537–17542.
    1. Cirovic B, et al. BCG vaccination in humans elicits trained immunity via the hematopoietic progenitor compartment. Cell Host Microbe. 2020;28:322–334.
    1. Bernelot Moens SJ, et al. Unexpected arterial wall and cellular inflammation in patients with rheumatoid arthritis in remission using biological therapy: a cross-sectional study. Arthritis Res. Ther. 2016;18:115.
    1. Schloss MJ, Swirski FK, Nahrendorf M. Modifiable cardiovascular risk, hematopoiesis, and innate immunity. Circ. Res. 2020;126:1242–1259.
    1. Christ A, et al. Western diet triggers NLRP3-dependent innate immune reprogramming. Cell. 2018;172:162–175.
    1. Bekkering S, et al. Trained immunity: linking obesity and cardiovascular disease across the life-course? Trends Endocrinol. Metab. 2020;31:378–389.
    1. Pink C, et al. Longitudinal effects of systemic inflammation markers on periodontitis. J. Clin. Periodontol. 2015;42:988–997.
    1. Wright HJ, Matthews JB, Chapple IL, Ling-Mountford N, Cooper PR. Periodontitis associates with a type 1 IFN signature in peripheral blood neutrophils. J. Immunol. 2008;181:5775–5784.
    1. Kalafati L, et al. Innate immune training of granulopoiesis promotes anti-tumor activity. Cell. 2020;183:771–785.
    1. Rafferty B, et al. Impact of monocytic cells on recovery of uncultivable bacteria from atherosclerotic lesions. J. Intern. Med. 2011;270:273–280.
    1. Kozarov EV, Dorn BR, Shelburne CE, Dunn WA, Jr, Progulske-Fox A. Human atherosclerotic plaque contains viable invasive Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis. Arterioscler. Thromb. Vasc. Biol. 2005;25:e17–e18.
    1. Carrion J, et al. Microbial carriage state of peripheral blood dendritic cells (DCs) in chronic periodontitis influences DC differentiation, atherogenic potential. J. Immunol. 2012;189:3178–3187.
    1. Gevers D, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–392.
    1. Schirmer M, et al. Compositional and temporal changes in the gut microbiome of pediatric ulcerative colitis patients are linked to disease course. Cell Host Microbe. 2018;24:600–610.e604.
    1. Lamont RJ, Koo H, Hajishengallis G. The oral microbiota: dynamic communities and host interactions. Nat. Rev. Microbiol. 2018;16:745–759.
    1. Gimbrone MA, Jr, García-Cardeña G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ. Res. 2016;118:620–636.
    1. Mougeot JC, et al. Porphyromonas gingivalis is the most abundant species detected in coronary and femoral arteries. J. Oral Microbiol. 2017;9:1281562.
    1. Dioguardi M, et al. The role of periodontitis and periodontal bacteria in the onset and progression of Alzheimer’s disease: a systematic review. J. Clin. Med. 2020;9:495.
    1. Beydoun MA, et al. Clinical and bacterial markers of periodontitis and their association with incident all-cause and Alzheimer’s disease dementia in a large national survey. J. Alzheimers Dis. 2020;75:157–172.
    1. Kovacech B, Novak M. Tau truncation is a productive posttranslational modification of neurofibrillary degeneration in Alzheimer’s disease. Curr. Alzheimer Res. 2010;7:708–716.
    1. Long JM, Holtzman DM. Alzheimer disease: an update on pathobiology and treatment strategies. Cell. 2019;179:312–339.
    1. Muñoz SS, Garner B, Ooi L. Understanding the role of ApoE fragments in Alzheimer’s disease. Neurochem. Res. 2019;44:1297–1305.
    1. Lönn J, et al. Lipoprotein modifications by gingipains of Porphyromonas gingivalis. J. Periodont. Res. 2018;53:403–413.
    1. Poole S, et al. Active invasion of Porphyromonas gingivalis and infection-induced complement activation in ApoE–/– mice brains. J. Alzheimers Dis. 2015;43:67–80.
    1. Hajishengallis G, Reis ES, Mastellos DC, Ricklin D, Lambris JD. Novel mechanisms and functions of complement. Nat. Immunol. 2017;18:1288–1298.
    1. Yin C, et al. ApoE attenuates unresolvable inflammation by complex formation with activated C1q. Nat. Med. 2019;25:496–506.
    1. Kantarci A, et al. Combined administration of resolvin E1 and lipoxin A4 resolves inflammation in a murine model of Alzheimer’s disease. Exp. Neurol. 2018;300:111–120.
    1. Kantarci A, et al. Microglial response to experimental periodontitis in a murine model of Alzheimer’s disease. Sci. Rep. 2020;10:18561.
    1. Papageorgiou SN, et al. Inflammatory bowel disease and oral health: systematic review and a meta-analysis. J. Clin. Periodontol. 2017;44:382–393.
    1. Pietropaoli D, et al. Occurrence of spontaneous periodontal disease in the SAMP1/YitFc Murine model of Crohn disease. J. Periodontol. 2014;85:1799–1805.
    1. Moran CJ, et al. IL-10R polymorphisms are associated with very-early-onset ulcerative colitis. Inflamm. Bowel Dis. 2013;19:115–123.
    1. Schmidt TS, et al. Extensive transmission of microbes along the gastrointestinal tract. eLife. 2019;8:e42693.
    1. Yachida S, et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat. Med. 2019;25:968–976.
    1. Thomas AM, et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat. Med. 2019;25:667–678.
    1. Kostic AD, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–215.
    1. Wirbel J, et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat. Med. 2019;25:679–689.
    1. Castellarin M, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012;22:299–306.
    1. Kostic AD, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22:292–298.
    1. Figuero E, Han YW, Furuichi Y. Periodontal diseases and adverse pregnancy outcomes: mechanisms. Periodontol. 2000. 2020;83:175–188.
    1. Rubinstein MR, et al. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14:195–206.
    1. Casasanta MA, et al. Fusobacterium nucleatum host-cell binding and invasion induces IL-8 and CXCL1 secretion that drives colorectal cancer cell migration. Sci. Signal. 2020;13:eaba9157.
    1. Yu T, et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017;170:548–563.
    1. Komiya Y, et al. Patients with colorectal cancer have identical strains of Fusobacterium nucleatum in their colorectal cancer and oral cavity. Gut. 2019;68:1335–1337.
    1. Abed J, et al. Colon cancer-associated Fusobacterium nucleatum may originate from the oral cavity and reach colon tumors via the circulatory system. Front. Cell Infect. Microbiol. 2020;10:400.
    1. Ning Y, Lenz HJ. Targeting IL-8 in colorectal cancer. Expert. Opin. Ther. Targets. 2012;16:491–497.
    1. Rubinstein MR, et al. Fusobacterium nucleatum promotes colorectal cancer by inducing Wnt/β-catenin modulator annexin A1. EMBO Rep. 2019;20:e47638.
    1. Sun J, et al. Role of the oral microbiota in cancer evolution and progression. Cancer Med. 2020;9:6306–6321.
    1. Hashimoto M, et al. Periodontitis and Porphyromonas gingivalis in preclinical stage of arthritis patients. PLoS ONE. 2015;10:e0122121.
    1. Mikuls TR, et al. Periodontitis and Porphyromonas gingivalis in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014;66:1090–1100.
    1. Al-Katma MK, Bissada NF, Bordeaux JM, Sue J, Askari AD. Control of periodontal infection reduces the severity of active rheumatoid arthritis. J. Clin. Rheumatol. 2007;13:134–137.
    1. Khare N, et al. Nonsurgical periodontal therapy decreases the severity of rheumatoid arthritis: a case-control study. J. Contemp. Dent. Pract. 2016;17:484–488.
    1. Ortiz P, et al. Periodontal therapy reduces the severity of active rheumatoid arthritis in patients treated with or without tumor necrosis factor inhibitors. J. Periodontol. 2009;80:535–540.
    1. Maresz KJ, et al. Porphyromonas gingivalis facilitates the development and progression of destructive arthritis through its unique bacterial peptidylarginine deiminase (PAD) PLoS Pathog. 2013;9:e1003627.
    1. Gully N, et al. Porphyromonas gingivalis peptidylarginine deiminase, a key contributor in the pathogenesis of experimental periodontal disease and experimental arthritis. PLoS ONE. 2014;9:e100838.
    1. Wegner N, et al. Peptidylarginine deiminase from Porphyromonas gingivalis citrullinates human fibrinogen and alpha-enolase: implications for autoimmunity in rheumatoid arthritis. Arthritis Rheumatol. 2010;62:2662–2672.
    1. Lübcke PM, et al. Periodontal treatment prevents arthritis in mice and methotrexate ameliorates periodontal bone loss. Sci. Rep. 2019;9:8128.
    1. Flak MB, et al. Inflammatory arthritis disrupts gut resolution mechanisms, promoting barrier breakdown by Porphyromonas gingivalis. JCI Insight. 2019;4:e125191.
    1. Boyaka, P. N. & Fujihashi, K. in Clinical Immunology (Fifth Edition) (eds Rich, R. R. et al.) 285–298 (Elsevier, 2019).
    1. Gill N, Wlodarska M, Finlay BB. The future of mucosal immunology: studying an integrated system-wide organ. Nat. Immunol. 2010;11:558–560.
    1. Scribano ML. Vedolizumab for inflammatory bowel disease: from randomized controlled trials to real-life evidence. World J. Gastroenterol. 2018;24:2457–2467.
    1. Calderón-Gómez E, et al. Commensal-specific CD4+ cells from patients with Crohn’s disease have a T-helper 17 inflammatory profile. Gastroenterology. 2016;151:489–500.
    1. Wang J, et al. Respiratory influenza virus infection induces intestinal immune injury via microbiota-mediated Th17 cell–dependent inflammation. J. Exp. Med. 2014;211:2397–2410.
    1. D’Amico F, Baumgart DC, Danese S, Peyrin-Biroulet L. Diarrhea during COVID-19 infection: pathogenesis, epidemiology, prevention, and management. Clin. Gastroenterol. Hepatol. 2020;18:1663–1672.
    1. Miles B, et al. Secondary lymphoid organ homing phenotype of human myeloid dendritic cells disrupted by an intracellular oral pathogen. Infect. Immun. 2014;82:101–111.
    1. Sainz J, Sata M. CXCR4, a key modulator of vascular progenitor cells. Arterioscler. Thromb. Vasc. Biol. 2007;27:263–265.
    1. Rajendran M, et al. Systemic antibiotic therapy reduces circulating inflammatory dendritic cells and Treg-Th17 plasticity in periodontitis. J. Immunol. 2019;202:2690–2699.
    1. Chen M, et al. Dendritic cell apoptosis in the maintenance of immune tolerance. Science. 2006;311:1160–1164.
    1. Hajishengallis G, Chavakis T, Lambris JD. Current understanding of periodontal disease pathogenesis and targets for host-modulation therapy. Periodontol. 2000. 2020;84:14–34.
    1. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017;377:1119–1131.
    1. Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat. Rev. Cardiol. 2020;17:137–144.
    1. Fuster JJ, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. 2017;355:842–847.
    1. Eskan MA, et al. The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss. Nat. Immunol. 2012;13:465–473.
    1. Shin J, et al. DEL-1 restrains osteoclastogenesis and inhibits inflammatory bone loss in nonhuman primates. Sci. Transl. Med. 2015;7:307ra155.
    1. Kourtzelis I, et al. DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat. Immunol. 2019;20:40–49.
    1. Li X, et al. The DEL-1/β3 integrin axis promotes regulatory T cell responses during inflammation resolution. J. Clin. Invest. 2020;130:6261–6277.
    1. Mitroulis I, et al. Secreted protein Del-1 regulates myelopoiesis in the hematopoietic stem cell niche. J. Clin. Invest. 2017;127:3624–3639.
    1. Adler CJ, et al. Sequencing ancient calcified dental plaque shows changes in oral microbiota with dietary shifts of the Neolithic and Industrial revolutions. Nat. Genet. 2013;45:450–455.
    1. Peres MA, et al. Oral diseases: a global public health challenge. Lancet. 2019;394:249–260.
    1. Dutzan N, et al. A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans. Sci. Transl. Med. 2018;10:eaat0797.
    1. Abe T, Hajishengallis G. Optimization of the ligature-induced periodontitis model in mice. J. Immunol. Methods. 2013;394:49–54.
    1. Hajishengallis G, et al. Low-abundance biofilm species orchestrates inflammatory periodontal disease through the commensal microbiota and complement. Cell Host Microbe. 2011;10:497–506.
    1. Payne MA, et al. Horizontal and vertical transfer of oral microbial dysbiosis and periodontal disease. J. Dent. Res. 2019;98:1503–1510.
    1. Penkov S, Mitroulis I, Hajishengallis G, Chavakis T. Immunometabolic crosstalk: an ancestral principle of trained immunity? Trends Immunol. 2019;40:1–11.
    1. Kaufmann E, et al. BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis. Cell. 2018;172:176–190.
    1. Nahrendorf M. Myeloid cell contributions to cardiovascular health and disease. Nat. Med. 2018;24:711–720.
    1. Murphy AJ, Tall AR. Disordered haematopoiesis and athero-thrombosis. Eur. Heart J. 2016;37:1113–1121.
    1. Barrett TJ, Murphy AJ, Goldberg IJ, Fisher EA. Diabetes-mediated myelopoiesis and the relationship to cardiovascular risk. Ann. N. Y. Acad. Sci. 2017;1402:31–42.
    1. Tall AR, Yvan-Charvet L. Cholesterol, inflammation and innate immunity. Nat. Rev. Immunol. 2015;15:104–116.

Source: PubMed

3
Iratkozz fel