Skip to main content
Hippokratia logoLink to Hippokratia
. 2008 Jul-Sep;12(3):144–149.

Periodontitis as a risk factor for cardiovascular disease: The role of anti-phosphorylcholine and anti-cardiolipin antibodies

K Karnoutsos 1, P Papastergiou 2, S Stefanidis 3, A Vakaloudi 2
PMCID: PMC2504402  PMID: 18923669

Abstract

Available evidence does allow an interpretation of periodontitis as being a risk factor for atherosclerosis and coronary heart disease. There is now a convincing body of evidence that mechanism of atherosclerosis has a major inflammatory component and it is much more than the simple accumulation of lipids on the vascular walls. Studies have shown that certain other mild bacterial infections consist a major risk factor for stroke in young and middle aged patients. Several possible mechanisms could explain the observed association between infection and infraction. The evidence supports the premise that periodontitis leads to systemic exposure to oral bacteria and that the resulting production of inflammatory mediators is capable of initiating or supporting mechanisms associated to development of atherosclerosis and coronary heart disease.

Studies in patients with pathologic concentrations of anti-cardiolipin and anti-phosphorylcholine antibodies demonstrated increased pocket depth and attachment loss, compared to patients with normal levels of the above antibodies. These antibodies could be associated to increased risk for stroke and atherosclerosis in patients with periodontitis.

As we become more familiar to the association between periodontitis and cardiovascular disease it is likely that in the future periodontal disease may be added to the list of the factors which are used to assess patients' risk profile for coronary heart disease and stroke.

Keywords: periodontitis, infection, infraction, cardiolipin, phosphorylcholine


Cardiovascular disease occurs as a result of a complex set of genetic and environmental factors14. The genetic factors include age, hypertension5, diabetes6, marked obesity7, lipid metabolism8, fibrinogen levels9,10 and platelet P1 polymorphism11. The environmental risk factors include diet12, physical inactivity, stress13,14, cigarette smoking15,16, socioeconomic status17, chronic infections18,19, use of non-steroid anti-inflammatory drugs20 and possible endothelial cell injury2123. Cardiovascular mortality rates account for about 30% of all deaths, despite efforts aiming at controlling the conventional risk factors. The identification of factors leading to an increased risk of coronary heard disease is still far from complete. It has been estimated that, to a significant percentage of patients with the disease, no one of the established risk factors aforementioned, does apply24.

Periodontitis is an inflammatory disease of the gums, which affects all the dental supporting tissues. It has been mentioned that severe generalized periodontal disease is present in 8% to 13% of the world's adult population25,26. The prevalence is lower in children and young adults with an estimated rate 2% to 5% between the ages of 11 and 25 being affected27. Periodontitis is a progressive inflammation, leading to the destruction of the supporting tissue and alveolar bone loss28,29. This process is the result of the bacteria - produced toxins and the inflammatory reaction of the gum tissues30.

Today, the available evidence does allow us an interpretation of periodontitis being a risk factor for atherosclerosis coronary heart disease31.

In this paper, we examine the evidence for an association between periodontal infections and increased risk of cardiovascular disease and we also study the connection between periodontal infections and increased concentration of antiphosphorylcholine and anticardiolipin atnibodies in the serum of patients with periodontitis.

Infection, inflammation and atherosclerosis

Infection has been recognized for decades as a risk factor for atherogenesis and thromboembolic events3235. Gram-negative bacteria or the associated lipopolysaccharide (LPS) endotoxin, when presented as a systemic challenge in animal models, can induce inflammatory cell infiltration into major blood vessels, vascular smooth muscle proliferation, vascular fatty degeneration and intravascular coagulation3236. The remarkable similarities of bacteria-induced vascular pathology and the natural history of atherogenesis has to investigators to suggest that, infections of unknown origin may contribute to the observed cardiovascular pathology in addition to genetic and dietary influences3239. It has, already, been suggested the association of certain infections with atherogenesis and thromboembolic events as risk factors for this condition31.

The association with increased coronary heart disease and stroke risk is strongest for Chlamydia pneumoniae (Cp). Cp is believed to disseminate through the blood to infect the vascular endothelium and contribute to the occurrence of atherosclerosis38,39. As shown by immunofluorescent techniques, antigens of Cp localize to atherosclerotic plaques with a high prevalence in individuals with prior coronary bypass surgery. Control specimens of coronary arteries obtained from individuals without clinical signs of coronary artery disease rarely show evidence of Cp infection. Other microbes or viruses, which are implicated in the pathology of atherosclerosis, include Helicobacter pylori38, cytomegalovirus4042, and Herpes virus type 243.

Helicobacter pylori, which is known for its colonization on gastric mucosa, has been related with increased risk of coronary heart disease44. It has been found that there is a 33% prevalence of antibody reactive with β2GPI in patients with Helicobacter pylori infection45. Other bacterial infections that may trigger the production of these antibodies range from spirochetes infections such as Syphilis and Lyme disease to those involving non-periodontal Staphylococcal and Streptococcal organisms46.

As atherosclerotic lesions develop in the coronary arteries, the risk for occurrence of myocardial ischemia and infraction increases47.

Possible role of dental infections in the etiopathogenesis of atherosclerosis, coronary heart disease and stroke

Oral flora is normally confined in the mouth, located on the surfaces of the tongue, gingiva, mucous membranes and teeth. Periodontal pathogens, invade epithelial cells and connective tissue causing periodontal inflammation and bleeding which enables entry of oral flora, including non-invasive organism, into the blood stream and transportation to systemic locales. Procedures such as dental extraction, periodontal surgery, tooth scaling and even the brushing of teeth often lead to the presence of oral bacteria in the blood stream (bacteremia). There is evidence that common oral hygiene practices daily produce low-level bacteremia48.

Certain oral bacteria are known as causative agents associated to infective endocarditis49,50. Furthermore, dental bacteremias associated to periodontitis are considered as risk factors for coronary heart disease and stroke51.

Atherosclerotic plaque samples are often found infected with multiple infectious agents, as Porphyromonas gingivalis and Sstreptococcus sanguis, which are common in periodontal disease. The immunolocalization of these microorganisms within unstable plaque regions and their association with plaque ulceration, thrombosis and apoptosis in vascular cells, are intriguing. Multiple infectious agents may alter vascular cell function and provide the possibility for acute ischemic stroke events51.

Streptococcus sanguis is the most prevalent species in dental plaque52 and frequently identified in the polymicrobial bacteremia form dental foci53. Platelet aggregating strains (Agg+) of Streptococcus sanguis are found to induce aggregation of human platelets in vitro5456. This phenomenon is mediated by the expression of the platelet aggregation-associated protein (PAAP) on the surface of certain strains57. The PAAP is a collagen-like cell surface antigen containing the sequence KPGEPGK58. This sequence forms a structural motif common in all known platelet-interactive domains of collagens5961.

In experimental studies on rabbits, PAAP was found to induce the formation of platelet vegetation during endocarditis62. Following infusion of Agg+ Streptococcus sanguis, platelets and fibrin accumulation on injured heart valves, was observed56. To verify that valvular vegetation was indeed thrombi, rabbits were treated to inhibit formation of digest platelet-associated fibrin63. The eventual mass of the vegetation was reduced by pretreatment with monospecific rabbit antifebrin antibody or by therapy with recombinant human tissue plasminogen activator.

When present in the circulation, Agg+ Streptococcus sanguis may also induce thromboembolic events. The Agg+ phenotype may be associated with the occurrence of disseminated intravascular coagulation-like syndrome in immunocompromised patients64,65. Approximately 60% of human isolates express the Agg+ phenotype66. Studies have shown that infusion of Agg+ colony strains causes a rise in blood pressure, heart rate, increased cardiac contractility and chances in electrocardiograms (ECGs) in rabbits58,67.

Inflammatory cells and the inflammatory mediators such as tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β) and prostaglandin E2 (PGE2) play a key role in human coronary heart disease and atherosclerosis36,6870. Bacterial LPS initiates the expression of IL-1b, which impedes fibrilonysis but facilitates coagulation and thrombosis71. Cytokines enhance both cholesterol accumulation in monocytes and smooth muscle proliferation, which presumably results in thickening of vessel walls36,64. Thus, the pathway LPS, moncyte activation, inflammatory-mediator production is implicated as an important mechanism in the pathogenesis of coronary heart disease and atherosclerosis. Today, the existence of inflammatory response in the process of the periodontal disease has been proved72. Monocytes within the periodontal tissues respond to LPS production of the plaque organisms by secreting important pro-inflammatory mediators, such as TNF-α, IL-1b, PGE2, and thromboxane A2 (TxA2), which not only cause local effects in the periodontal tissues, but may appear systemically9,68. Periodontal disease, like ischemic heart disease involves the above shown pathway31. The systemic response to oral pathogens is a result of bacterial penetration into tissues, loss of integrity of the epithelium in the periodontal sulcus and transient dental bacteremia. It has been shown that endotoxins of plaque microorganisms are capable of penetrating the gingival tissues and entering into the blood stream, in amounts sufficient to bring about a systemic LPS-specific antibody response73.

The role of antiphosphorylcholine and anticardiolipin antibodies in serum of patients with periodontitis

Periodontitis is considered to be the result of the bacteria-host interaction30. A vital component of the etiology of periodontitis is the lipopolysacharite, located on the outer membrane of Gram-negative periodontal bacteria. This molecule has 3 major component parts: the core, the external O-antigen and the lipid A which is embedded within the lipid portion of the outer membrane74 and is responsible for the endotoxin properties. Minor lippopolysaccharide antigenic components have also been identified. One such molecule is phosphorylcholine that is attached to cell wall polysaccharide and lipoteichoid acid75 which has been identified in over 30% of the supragingival and subgingival flora, including Streptococcus oralis, Sterptococcus sanguis, Actinomyces israelii, Actinomyces naeslundii, Fussobacterium mucleatum, Haemophilus aphrophilus and Actinobacillus actinomycetemcomitans76,77. The possible function of phoshporylcholine is that certain bacteria may utilize it to gain access to endothelial cells78 or the circulation7981. Bacterial adherence, colonization and invasion are reliant upon surface phosphorylcholine. It has been shown that Streptococcus pneumoniae and Actinobacillus actinomycetemconimans invasion into endothelial cells is based on the interaction between surface phosphorylcholine and endothelial surface receptors for platelet activating factor79,80. Phosphorylcholine, as a component of the lipopolysaccharide (LPS) motive of many bacteria, plays a role in prompting a host immune response. Studies have shown that Phosphorylcholine influences polyclonal B-cell differentiation and activation82,83. Additionally, there is host production of IgG and IgM antibodies directed against Phosphorylcholine, which can assist to monocyte recognition and phagocystosis of the pathogenic bacteria. Phosphorylcholine - positive strains of Streptococcus pneumoniae and Actinobacillus actinomycetemconimans become opsonized by anti-phosphorylcholine IgG84. Signs of such host-periodontal pathogen interplay are not only detected locally but also systemically. The ability of periodontal pathogen bacteria of producing a systemic response to phosphorylcholine is demonstrated by higher serum levels of antibodies directed toward phosphorylcholine (anti-phosphorylcholine IgG) in patients with attachment loss, in comparison with those with healthy gums76. Furthermore, other researchers suggest that both phosphorylcholine bearing strains of oral bacteria and oxidized low-density lipoproteins (oxLDL) react with anti-phosphorylcholine IgG from human serum85. This suggests that antibodies produced against certain periodontal bacteria would also react to phosphorylcholine-bearing oxLDL86 and, therefore, magnify the uptake of this lipid by foam cells, promoting further progress of atherosclerosis.

Cardiolipin (CL) is a phospholipid found in mammalian tissues and eukaryotic organisms and is also produced by some prokaryotic bacteria. It is located in the inner mitochondrial membrane and it is suggested that it plays an integral role in normal electron transport and energy metabolism87.

Antiphospholipid antibodies consist a group of autoantibodies found in several pathologic conditions, including a variety of infectious diseases and are a hallmark of the anti-phospholipid syndrome (APS). APS is present in about 30 to 40% of patients with systematic lupus erythimatosus (SLE), although there are individuals with the primary form of APS, which do not develop SLE. The major clinical symptoms of APS include recurrent venous or arterial thrombosis and fetal loss. Patients with APS may also demonstrate premature atherosclerosis. Pathogenesis of APS is related to prothrombotic activity of some antiphoshpolipid antibodies88. Among the major groups of antibodies detected in patients with APS are b-2glycoprotein, I-dependent anti-cardiolipin (anti-CL), anti-β-2glycoprotien I (anti-β2GPI) and lupus anticoagulant (LA). β2GPI is a 50kDa plasma phoshpolipid-binding protein which functions as a natural anticoagulant89. Immunoassays measuring pathogenic anti-CL require the incorporation of β2GPI bound to CL for detection of anti-CL autoantibodies that promote procoaguland activity90,91. Autoantibodies directed at β2GPI may also be detected in immunoassays that omit CL, though these subsets of anti-CL and anti-β2GPI may not be identical92. In summary, this group of antibodies is heterogeneous and clinical tests usually involve multiple assays to detect autoimmune anti-CL and anti- β2GPI.

Recent studies strongly implicate bacterial and viral infection in the etiology of APS due to induction of cross-reactivity anti-CL autoantibodies. A hexapeptide (TLRVYK) sequence in β2GPI has been identified to be recognized by some anti- β2GPI monoclonal antibodies. Mice immunized with microbial pathogens such as Hemophilus influenza or Neisseria gonorrheae with homologous sequences related to TLRVYK, produced crossreactive anti- β2GPI that induced APS-like symptoms, when subsequently purified and passively infused into mice. Thus, bacterial infection could lead to production of pathogenic anti-CL and be responsible for a subset of cases of APS93.

Periodontal bacteria including Actiobacillus actinomycetemcomitans, Porphyromonas gingivalis have been found to contain a peptide sequence similar to that on the β2GPI molecule9395. In addition, there is evidence that β2GPI by itself is immunogenic94, possibly compounding this response.

There are more examples suggesting an association between periodontitis and cardiolipin. Reports have demonstrated that the prevalence of patients with chronic periodontitis and generalized aggressive periodontitis positive for anti-CL autoantibodies was greater than in healthy controls and patients with localized aggressive periodontitis. Patients with elevated anticardiolipin had greater mean attachment loss and increased pocket depth96.

A recent clinical study in patients with aggressive periodontitis has observed an association between systemic vascular inflammation markers and elevated levels of anti-CL97.

Conclusions

Epidemiological data indicate that periodontal disease is an independent risk factor for myocardial infarction. Periodontal infections have also been suggested as one of the several factors contributing to the development of coronary heart disease98. Evidence supporting a causative role of chronic infection in coronary heart disease is largely circumstantial.

The evidence supports the premise that periodontitis leads to systemic exposure to oral bacteria and that a potential source of systemic inflammatory mediators, capable of initiating or worsening conditions associated with atherosclerosis and coronary heart disease, are cytokines and LPS produced in the infected periodontal tissues, which enter into the blood stream. Cytokines produce their effects directly, whereas the LPS trigger a systemic cascade of inflammatory cytokines, also capable of eliciting effects associated with atherosclerosis and coronary heart disease.

The continued systemic exposure to Gram negative bacteria and LPS results in a release of cytokines such as tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β) and prostaglandin E2 (PGE2) which may be a significant factor in the pathogenesis of coronary heart disease and stroke.

The suggestion that periodontal disease is a significant risk factor for coronary disease adds a new perspective to oral health and should serve to align it with the other aspects of preventive medicine. It also raises a core question about the effect of periodontal treatment in reducing the risk of heart disease.

Clinical studies on periodontal disease have revealed a positive association with coronary disease and emphasis is now being placed on understanding the relation between periodontal disease and atherosclerosis. It has already been demonstrated that anti-phosphorylcholine directed from pathogenic bacteria in the periodontal pocket may cause a cross-reaction with oxLDL, leading to events that culminate in atherosclerotic plaque formation.

As IgG anticardiolipin is a potential factor in atherosclerotic plaque formation and thrombosis, its association to periodontal bacteria becomes an important issue.

Patients with elevated anticardiolipin antibodies present greater mean attachment loss and increased pocket depth. It appears that the development of periodontitis demonstrating greater extent and severity may lead to the production of anticardiolipin antibodies in those patients.

It is likely that in the future periodontal disease may be added to the list of factors, which are used to assess patients' risk profiles for coronary heart disease and stroke. In addition, treatment of periodontal disease should become a standard part of the therapy for patients with the above diseases.

References

  • 1.Fuster V, Badimon L, Badimon JJ, Chesedro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes [part 1] N Engl J Med. 1992;326:242–250. doi: 10.1056/NEJM199201233260406. [DOI] [PubMed] [Google Scholar]
  • 2.Fuster V, Badimon L, Badimon JJ, Chesedro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes [part 2] N Engl J Med. 1992;326:310–318. doi: 10.1056/NEJM199201303260506. [DOI] [PubMed] [Google Scholar]
  • 3.Hegele RA. The pathogenesis of atherosclerosis. Clin Chim Acta. 1996;246:21–38. doi: 10.1016/0009-8981(96)06224-9. [DOI] [PubMed] [Google Scholar]
  • 4.Higgins M, Province M, Heiss G, et al. NHLBI Family Heart Stydy: objectives and design. Am J E. 1996;143:1219–1228. doi: 10.1093/oxfordjournals.aje.a008709. [DOI] [PubMed] [Google Scholar]
  • 5.Rakugi H, Yu H, Kamitani A, et al. Links between hypertension and myocardial infraction. Am Heart J. 1996;132:213–221. [PubMed] [Google Scholar]
  • 6.Stern MP. Do non-insulin dependent diabetes mellitus and cardiovascular disease share common antecedents? Ann Intern Med. 1996;124:110–116. doi: 10.7326/0003-4819-124-1_part_2-199601011-00006. [DOI] [PubMed] [Google Scholar]
  • 7.Lonnqvist F, Arner P, Nordfors L, Schalling M. Overexpression of the obese [ob] gene in adipose tissue of human objects. Nat Med. 1995;1:950–953. doi: 10.1038/nm0995-950. [DOI] [PubMed] [Google Scholar]
  • 8.Kesaniemi YA. Genetics and cholesterol metabolism. Curr Opin Lipidol. 1996;7:124–131. doi: 10.1097/00041433-199606000-00003. [DOI] [PubMed] [Google Scholar]
  • 9.Eber B, Schumacher M. Fibrinogen: its role in the hemostatic regulation in atherosclerosis. Semin Thromb Hemost. 1993;19:104–107. doi: 10.1055/s-2007-994012. [DOI] [PubMed] [Google Scholar]
  • 10.Heinrich J, Assmann G. Fibrinogen and cardiovascular risk. J Cardiovasc Risk. 1995;2:197–205. [PubMed] [Google Scholar]
  • 11.Weiss EJ, Bray PF, Tayback M, et al. A polymorphism of a platelet glycoprotein receptor as an inherited risk factor for coronary thrombosis. N Engl J Med. 1996;334:1090–1094. doi: 10.1056/NEJM199604253341703. [DOI] [PubMed] [Google Scholar]
  • 12.Stampfer MJ, Sacks FM, Salvini S, Willett WC, Hennekens CH. A prospective study of cholesterol, apolipoproteins and the risk of myocardial infarction. N Engl J Med. 1991;325:373–381. doi: 10.1056/NEJM199108083250601. [DOI] [PubMed] [Google Scholar]
  • 13.Grignani G, Soffiantino F, Zucchella M, et al. Platelet activation by emotional stress in patients with coronary artery disease. Circulation. 1991;83(Suppl 4):128–136. [PubMed] [Google Scholar]
  • 14.Yeung AC, Vekshtein VI, Krantz DS, et al. The effect of atherosclerosis on the vasomotor response of coronary arteries to mental stress. N Engl J Med. 1991;325:1551–1556. doi: 10.1056/NEJM199111283252205. [DOI] [PubMed] [Google Scholar]
  • 15.Hung J, Lam JY, Lacoste L, Letchacovski G. Cigarette smoking acutely increase platelet thrombus formation in patients with coronary artery disease taking aspirin. Circulation. 1995;92:2432–2436. doi: 10.1161/01.cir.92.9.2432. [DOI] [PubMed] [Google Scholar]
  • 16.Kuller LH, Tracy RP, Shaten J, Meilahn EN. Relation of C-reactive protein and coronary heart disease in the MRFIT nested case-control study. Am J Epidemiol. 1996;144:537–547. doi: 10.1093/oxfordjournals.aje.a008963. [DOI] [PubMed] [Google Scholar]
  • 17.Rosengren A, Wilhemsen L, Welin L, Tsipogianni A, Teger-Nillson AC, Wedel H. Social influences and cardiovascular risk factor as determinants of plasma fibrinogen concentration in a general population sample of middle-aged men. Br Med J. 1990;300:634–638. doi: 10.1136/bmj.300.6725.634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nieminen MS, Mattila K, Valtonen V. Infection and inflammation as risk factors for myocardial infarction. Eur Heart J. 1993;14(Suppl):12–16. [PubMed] [Google Scholar]
  • 19.Buja LM. Does atherosclerosis have an infectious etiology? Circulation. 1996;94:872–873. doi: 10.1161/01.cir.94.5.872. [DOI] [PubMed] [Google Scholar]
  • 20.Ridker PM, Cushman M, Stamper MJ, Tracy RP, Hennekens CH. Inflammation, aspirin and the risk of cardiovascular disease in apparently healthy men. N Engl J Med. 1997;336:973–979. doi: 10.1056/NEJM199704033361401. [DOI] [PubMed] [Google Scholar]
  • 21.Riccioni G, De Santis A, Cerasa V, et al. Atherosclerotic plaque formation and risk factors. Int J Immunopathol Pharmacol. 2003;16:25–31. doi: 10.1177/039463200301600104. [DOI] [PubMed] [Google Scholar]
  • 22.Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990's. Nature. 1993;362:801–809. doi: 10.1038/362801a0. [DOI] [PubMed] [Google Scholar]
  • 23.Choy PC, Slow YL, Mymin D, O K. Lipids and atherosclerosis. Biochem Cell Biol. 2004;82:212–224. doi: 10.1139/o03-085. [DOI] [PubMed] [Google Scholar]
  • 24.Branunwald E. Shattuck lecture-cardiovascular medicine at the turn of the millennium: triumphs, concerns and opportunities. N Engl J Med. 1997;337:1360–1369. doi: 10.1056/NEJM199711063371906. [DOI] [PubMed] [Google Scholar]
  • 25.Oliver RL, Brown LJ, Loe H. Periodontal disease in the United States population. J Periodontol. 1998;69:269–278. doi: 10.1902/jop.1998.69.2.269. [DOI] [PubMed] [Google Scholar]
  • 26.Page RC. Critical issues in periodontal research. J Dent Res. 1995;74:1118–1128. doi: 10.1177/00220345950740041301. [DOI] [PubMed] [Google Scholar]
  • 27.Albandar JM, Tinoco EM. Global epidemiology of periodontal disease in children and young persons. Periodontol 2000. 2002;29:153–176. doi: 10.1034/j.1600-0757.2002.290108.x. [DOI] [PubMed] [Google Scholar]
  • 28.Flemmig TF. Periodontitis. Ann Periodontol. 1999;4:32–38. doi: 10.1902/annals.1999.4.1.32. [DOI] [PubMed] [Google Scholar]
  • 29.Suzuki JB. Diagnosis and classification of the periodontal disease. Dent Clin North Am. 1988;32:195–216. [PubMed] [Google Scholar]
  • 30.Kornman KS, Page RC, Tonetti MS. The host response to the microbial challenge in periodontitis: assembling the players. Periodontol 2000. 1997;14:33–53. doi: 10.1111/j.1600-0757.1997.tb00191.x. [DOI] [PubMed] [Google Scholar]
  • 31.Beck JD, Offenbacher S. Oral health and systemic disease: periodontitis and cardiovascular disease. J Dent Educ. 1998;62:859–870. [PubMed] [Google Scholar]
  • 32.Syrjänen J, Peltola J, Valtonen V, Iivanainen M, Kaste M, Huttunen J. Dental infections in association with cerebral infarction in young and middle-aged men. J Intern Med. 1989;225:179–184. doi: 10.1111/j.1365-2796.1989.tb00060.x. [DOI] [PubMed] [Google Scholar]
  • 33.Mackenzie RS, Millarad HD. Interrelated effects of diabetes, arteriosclerosis and calculus on alveolar bone loss. J Am Dent Assoc. 1963;66:192–198. [Google Scholar]
  • 34.Nery EB, Meister F, Ellinger RF, Eslami A, McNamara TJ. Prevalence of medical problems in periodontal patients obtained from three different populations. J Periodontol. 1987;58:564–568. doi: 10.1902/jop.1987.58.8.564. [DOI] [PubMed] [Google Scholar]
  • 35.Osler W. Disease of the arteries. In: Osler W, editor. Modern medicine: its practice and theory. Philadelphia: Lea & Febiger; 1908. pp. 429–447. [Google Scholar]
  • 36.Marcus AJ, Hajjar DP. Vascular transcellular signaling. J Lipid Res. 1993;34:2017–2031. [PubMed] [Google Scholar]
  • 37.Umino M, Nagao M. Systemic disease in elderly dental patients. Int Dent J. 1993;43:213–218. [PubMed] [Google Scholar]
  • 38.Patel P, Mendall MA, Carrington D, et al. Association of Helicobacter pylori and Chlamydia pneumoniae infections with coronary heart disease and cardiovascular risk factors. Br Med J. 1995;311:711–714. doi: 10.1136/bmj.311.7007.711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Muhlestein JB, Hammond EH, Carlquist JF, et al. Increased incidence of Chlamydia species within the coronary arteries of patients with symptomatic atherosclerosclerotic versus other forms of cardiovascular disease. J Am Coll Cardiol. 1996;27:1555–1561. doi: 10.1016/0735-1097(96)00055-1. [DOI] [PubMed] [Google Scholar]
  • 40.Melnic JL, Hu C, Adam E, DeBakey ME. Cytomegalovirus DNA in arterial walls of patients with atherosclerosis. J Med Virol. 1994;42:170–174. doi: 10.1002/jmv.1890420213. [DOI] [PubMed] [Google Scholar]
  • 41.Nieto FJ, Adam E, Sorlie P, et al. Cohort study of cytomegalovirus infection as risk factor for carotid intimal-medial thickening, a measure of subclinical atherosclerosis. Circulation. 1996;94:922–927. doi: 10.1161/01.cir.94.5.922. [DOI] [PubMed] [Google Scholar]
  • 42.Vercellotti GM. Potential role of viruses in thrombosis and atherosclerosis. Trends Cardiovasc Med. 1995;5:128–133. doi: 10.1016/1050-1738(95)00052-B. [DOI] [PubMed] [Google Scholar]
  • 43.Raza-Ahmed A, Klassen GA, Murphy DA, et al. Evidence of type 2 herpes simplex infection in human coronary arteries at the time of coronary artery bypass surgery. Can J Cardiol. 1995;11:1025–1029. [PubMed] [Google Scholar]
  • 44.Pellicano R, Broutet N, Ponzetto A, Megraud F. Helicobacter pylori: from the stomach to the heart. Eur J Gastronetrol Hepatol. 1999;11:1335–1337. [PubMed] [Google Scholar]
  • 45.Sorice M, Pittoni V, Griggi T, et al. Specificity of anti-phospholipid antibodies in infectious mononucleosis: a role for anti-cofactor protein antibodies. Clin Exp Immunol. 2000;120:301–306. doi: 10.1046/j.1365-2249.2000.01206.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cervera R, Asherson RA. Antiphospholipid syndrome associated with infections: clinical and microbiological characteristics. Immunobiology. 2005;210:735–741. doi: 10.1016/j.imbio.2005.10.003. [DOI] [PubMed] [Google Scholar]
  • 47.Ambrose JA, Weinrauch M. Thrombosis in ischemic heart disease. Arch Intern Med. 1996;156:1382–1394. [PubMed] [Google Scholar]
  • 48.Durack DT. Prevention of infective endocarditis. N Engl J Med. 1995;332:38–44. doi: 10.1056/NEJM199501053320107. [DOI] [PubMed] [Google Scholar]
  • 49.Berbari EF, Cockerill FR, Steckelberg JM. Infective endocarditis due to unusual or fastidious microorganisms. Mayo Clin Proc. 1997;72:532–542. doi: 10.4065/72.6.532. [DOI] [PubMed] [Google Scholar]
  • 50.DeStefano F, Anda RF, Kahn S, Williamson DF, Russell CM. Dental disease and risk of coronary heart disease and mortality. BMJ. 1993;306:688–691. doi: 10.1136/bmj.306.6879.688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chiu B. Multiple infections in carotid atherosclerotic plaques. Am Heart J. 1999;138:s5534–s5536. doi: 10.1016/s0002-8703(99)70294-2. [DOI] [PubMed] [Google Scholar]
  • 52.Babb JL, Hamada SH. Microbiology of dental plaque. In: McGhee JR, Michalek SM, Cassell GH, editors. Dental Microbiology. Philadelphia: Harper & Row; 1982. pp. 663–678. [Google Scholar]
  • 53.Watanakunakorn C, Pantelakis J. Alpha-hemolytic streptococcal bacteremia: a review of 203 episodes during 1980-1991. Scand J Infect Dis. 1993;25:403–408. doi: 10.3109/00365549309008519. [DOI] [PubMed] [Google Scholar]
  • 54.Herzberg MC, MacFarlane GD, Delzer PR. Streptococcus sanguis interactions human with platelets. In: Mergenhagen SE, Rosan B, editors. Molecular basis of oral microbial Adhesion. Washington DC: American Society for Microbiology; 1985. pp. 53–60. [Google Scholar]
  • 55.Herzberg MC, Brintzenhofe KL, Clawson CC. Aggregation of human platelets and adhesion of Streptococcus sanguis. Infect Immun. 1983;39:1457–1469. doi: 10.1128/iai.39.3.1457-1469.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Herzberg MC. Platelet-streptococcal interactions in endocarditis. Crit Rev Oral Biol Med. 1996;7:222–236. doi: 10.1177/10454411960070030201. [DOI] [PubMed] [Google Scholar]
  • 57.Herzberg MC, Gong K, MacFarlane GD, et al. Phenotypic characterization of Streptococcus sanguis virulence factors associated with bacterial endocarditis. Infect Immun. 1990;58:515–522. doi: 10.1128/iai.58.2.515-522.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Herzberg MC, Meyer MW. Dental plaque, platelets and cardiovascular diseases. Ann Periodontol. 1998;3:151–160. doi: 10.1902/annals.1998.3.1.151. [DOI] [PubMed] [Google Scholar]
  • 59.Erickson PR, Herzberg MC. A collagen like immunodeterminant on the surface of Streptococcus sanguis platelet aggregation. J Immunol. 1987;138:3360–3366. [PubMed] [Google Scholar]
  • 60.Erickson PR, Herzberg MC. The Streptococcus sanguis plateletaggregation- associated protein. Identification and characterization of the minimal platelet-interactive domain. J Biol Chem. 1993;268:1646–1649. [PubMed] [Google Scholar]
  • 61.Erickson PR, Herzberg MC, Tierney G. Cross-reactive immunodeterminats on Streptococcus sanguis and collagen.Predicting the structure of the platelet-interactive domains. J Biol Chem. 1992;267:10018–10023. [PubMed] [Google Scholar]
  • 62.Herzberg MC, MacFarlane GD, Gong K, et al. Platelet-interactive phenotype of Steptococcus sanguis influences the course of experimental endocarditis. Infect Immun. 1992;60:4809–4818. doi: 10.1128/iai.60.11.4809-4818.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Meyer MW, Witt AR, Krishnan LK, et al. Therapeutic advantage of recombinant human plasminogen activator in endocarditis:evidence from experiments in rabbits. Thromb Haemost. 1995;73:680–682. [PubMed] [Google Scholar]
  • 64.Steiner M, Villablanca J, Kersey J, et al. Viridans streptococcal shock in bone marrow transplantation patients. Am J Hematol. 1993;42:354–358. doi: 10.1002/ajh.2830420405. [DOI] [PubMed] [Google Scholar]
  • 65.Rossetti F, Cesaro S, Putti MC, Zanesco L. High-dose cytosine arabinoside and viridans streptococcus sepsis in children with leukemia. Pediatr Hematol Oncol. 1995;12:387–392. doi: 10.3109/08880019509029589. [DOI] [PubMed] [Google Scholar]
  • 66.Herzberg MC, Gong K, MacFarlane GD, et al. Phenotypic characterization of streptococcous sanguis: virulence factors associated with bacterial endocarditis. Infect Immun. 1990;58:515–522. doi: 10.1128/iai.58.2.515-522.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Herzberg MC, Meyer MW. Effects of oral flora on platelets: possible consequences in cardiovascular disease. J Periodontol. 1996;67(Suppl10):1138–1142. doi: 10.1902/jop.1996.67.10s.1138. [DOI] [PubMed] [Google Scholar]
  • 68.Valtonen VV. Infection as a risk factor for infection and atherosclerosis. Ann Med. 1991;23:539–543. doi: 10.3109/07853899109150515. [DOI] [PubMed] [Google Scholar]
  • 69.Pearce WH, Sweis I, Yao JS, McCarthy WJ, Koch AE. Interleukin-1 beta and tumor necrosis factor-alpha release in normal and diseased human infrarenal aortas. J Vasc Surg. 1992;16:784–789. [PubMed] [Google Scholar]
  • 70.Funk JL, Feingold KR, Moser AH, Grunfeld C. Lipopolysaccharide stimulation of RAW 264.7 macrophages induces lipid accumulation and foam cell formation. Atherosclerosis. 1993;98:67–82. doi: 10.1016/0021-9150(93)90224-i. [DOI] [PubMed] [Google Scholar]
  • 71.Clinton SK, Fleet JC, Loppnow H, et al. Interleukin 1 gene expression in rabbit vascular tissue in vivo. Am J Pathol. 1991;138:1005–1014. [PMC free article] [PubMed] [Google Scholar]
  • 72.Offenbacher S. Periodontal disease: pathogenesis. Ann Peridontol. 1996;1:821–878. doi: 10.1902/annals.1996.1.1.821. [DOI] [PubMed] [Google Scholar]
  • 73.Hernichel-Gorbach E, Kornman KS, Holt SC, et al. Host responses in patients with generalized refractory periodontitis. J Periodontol. 1994;65:8–16. doi: 10.1902/jop.1994.65.1.8. [DOI] [PubMed] [Google Scholar]
  • 74.Takada H, Kotani S. Structure-function relationships of lipid A. In: Morison D, Ryan J, editors. Molecular biochemistry and cellular biology. Bacterial endotoxic lipopolysaccharides: vol I. New York: CRC Press; 1992. pp. 107–134. [Google Scholar]
  • 75.Fisher W, Behr T, Hartmann R, Peter-Katalinic J, Egge H. Teichoic acid and lipoteichoic acid of Streptococcus pneumoniae possess identical chain structures. A reinvestigation of teichoid acid (C polysaccharide) Eur J Biochem. 1993;215:851–857. doi: 10.1111/j.1432-1033.1993.tb18102.x. [DOI] [PubMed] [Google Scholar]
  • 76.Schenkein HA, Gunsolley JC, Best AM, et al. Antiphosphorylcholine antibody levels are elevated in humans with periodontal disease. Infect Immun. 1999;67:4814–4818. doi: 10.1128/iai.67.9.4814-4818.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Gmur R, Thurnheer T, Guggenheim B. Dominant cross-reactive antibodies generated during the response to a variety of oral bacterial species detect phosphorylcholine. J Dent Res. 1999;78:77–85. doi: 10.1177/00220345990780011201. [DOI] [PubMed] [Google Scholar]
  • 78.Schenkein HA, Barbour SE, Berry CR, Kipps B, Tew JG. Invasion of human vascular endothelial cells by Actinobacillus actineomycetemcomitans via the receptor for platelet-activating factor. Infect Immun. 2000;68:5416–5419. doi: 10.1128/iai.68.9.5416-5419.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cundell DR, Tuomanen EI. Receptor specificity of adherence of Streptococcus pneumoniae to human type-II pneumocytes and vascular endothelial cells in vitro. Microb Pathog. 1994;17:361–374. doi: 10.1006/mpat.1994.1082. [DOI] [PubMed] [Google Scholar]
  • 80.Cundell DR, Gerard NP, Gerard C, Idanpaan-Heikkila I, Tuomanen EI. Streptococcus pneumoniae anchor to activated human cells by the receptor for platelet-activating factor. Nature. 1995;377:435–438. doi: 10.1038/377435a0. [DOI] [PubMed] [Google Scholar]
  • 81.Cundell DR, Gerard C, Idanpaan-Heikkila I, Gerard NP. PA receptor anchors Streptococcus pneumoniae to activated human endothelial cells. Adv Exp Med Biol. 1996;416:89–94. doi: 10.1007/978-1-4899-0179-8_16. [DOI] [PubMed] [Google Scholar]
  • 82.Beckmann E, Levitt D. In vitro plaque-forming cell response induced by Streptococcus pneumoniae in humans. Scand J Immunol. 1984;19:1–10. doi: 10.1111/j.1365-3083.1984.tb00894.x. [DOI] [PubMed] [Google Scholar]
  • 83.Harnett W, Harnett MM. Inhibition of murine B cell proliferation and down-regulation of protein kinase C levels by a phosphorylcholine-containing filarial excretory-secretory product. J Immunol. 1984;151:4829–4837. [PubMed] [Google Scholar]
  • 84.Purkall D, Tew JG, Schenkein HA. Opsonization of Actinobacillus actinomycetemcominans by immunoglobulin G antibody reactive with phosphorylcholine. Infect Immun. 2002;70:6485–6488. doi: 10.1128/IAI.70.11.6485-6488.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Schenkein HA, Berry CR, Purkall D, Burnmeister JA, Brooks CN, Tew JG. Phosphorylcholine-dependent cross-reactivity between dental plaque bacteria and oxidized low-density lipoproteins. Infec Immun. 2001;69:6612–6617. doi: 10.1128/IAI.69.11.6612-6617.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Shaw PX, Goodyears CS, Chang MK, Witztum JL, Silverman GJ. The autoreactivity of anti-phosphorylcholine antibodies for atherosclerosis-associated neo-antigens and apoptotic cells. J Immunol. 2003;170:6151–6157. doi: 10.4049/jimmunol.170.12.6151. [DOI] [PubMed] [Google Scholar]
  • 87.McMillin JB, Dowhan W. Cardiolipin and apoptosis. Biochim Biophys Acta. 2002;1585:97–107. doi: 10.1016/s1388-1981(02)00329-3. [DOI] [PubMed] [Google Scholar]
  • 88.Levine JS, Branch DW, Rauch J. The antiphospholipid syndrome. N Engl J Med. 2002;346:752–763. doi: 10.1056/NEJMra002974. [DOI] [PubMed] [Google Scholar]
  • 89.Kandiah DA, Krilis SA. Beta 2-glycoprotein I. Lupus. 1994;3:207–212. doi: 10.1177/096120339400300401. [DOI] [PubMed] [Google Scholar]
  • 90.Galli M, Comfurius P, Maassen C, et al. Anticardiolipin antibodies (ACA) directed not to cardiolipin but to a plasma protein cofactor. Lancet. 1990;335:1544–1547. doi: 10.1016/0140-6736(90)91374-j. [DOI] [PubMed] [Google Scholar]
  • 91.McNally T, Purdy G, Mackie IJ, Machin SJ, Isenberg DA. The use of an anti-beta 2-glycoprotein-I assay for discrimination between anticardiolipin antibodies associated with infection and increased risk for thrombosis. Br J Haematol. 1995;91:471–473. doi: 10.1111/j.1365-2141.1995.tb05324.x. [DOI] [PubMed] [Google Scholar]
  • 92.Hojnik M, Gilburd B, Ziporen I, et al. Anticardiolipin antibodies in infections are heterogenous in their dependency on beta 2-glycoprotein I: analysis of anticardiolipin antibodies in leprosy. Lupus. 1994;3:515–521. doi: 10.1177/096120339400300615. [DOI] [PubMed] [Google Scholar]
  • 93.Blank M, Krause I, Fridkin M, et al. Bacterial induction of autoantibodies to beta2-glycoprotein-I accountis for the infectious etiology of antiphospholipid syndrome. J Clin Invest. 2002;109:797–804. doi: 10.1172/JCI12337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Blank M, Shoenfeld Y. Beta-2-glycoprotein-I, infections, antiphospholipid syndrome and therapeutic considerations. Clin Immunol. 2004;112:190–199. doi: 10.1016/j.clim.2004.02.018. [DOI] [PubMed] [Google Scholar]
  • 95.Gharavi AE, Pierangeli SS, Espinola RG, Liu X, Colden-Standfield M, Harris EN. Antiphospholipid antibodies induced in mice by immunization with a cytomegalovirus-derived peptide cause thrombosis and activation of endothelial cells in vivo. Athritis Rheum. 2002;46:545–552. doi: 10.1002/art.10130. [DOI] [PubMed] [Google Scholar]
  • 96.Schenkein HA, Berry CR, Burnmeister JA, et al. Anti-cardiolipin antibodies in sera from patients with periodontitis. J Dent Res. 2003;82:919–922. doi: 10.1177/154405910308201114. [DOI] [PubMed] [Google Scholar]
  • 97.Schenkein HA, Best AM, Brooks CN, et al. Anti-cardiolipin and increased serum adhesion molecule levels in patients with aggressive periodontitis. J Periodontol. 2007;78:459–466. doi: 10.1902/jop.2007.060305. [DOI] [PubMed] [Google Scholar]
  • 98.Armitage GC. Periodontal infections and cardiovascular diseasehow strong is the association? Oral Dis. 2000;6:335–350. doi: 10.1111/j.1601-0825.2000.tb00126.x. [DOI] [PubMed] [Google Scholar]

Articles from Hippokratia are provided here courtesy of Hippokratio General Hospital of Thessaloniki

RESOURCES