Next Article in Journal
Amazonian Native Palm Fruits as Sources of Antioxidant Bioactive Compounds
Next Article in Special Issue
Hydrogen Sulfide, Oxidative Stress and Periodontal Diseases: A Concise Review
Previous Article in Journal
Determination of Phenolic Content in Different Barley Varieties and Corresponding Malts by Liquid Chromatography-diode Array Detection-Electrospray Ionization Tandem Mass Spectrometry
Previous Article in Special Issue
Drinking Hydrogen-Rich Water Has Additive Effects on Non-Surgical Periodontal Treatment of Improving Periodontitis: A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Periodontal Disease-Induced Atherosclerosis and Oxidative Stress

1
Department of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
2
Stomatology Hospital, Tongji University, Shanghai 200072, China
3
Department of Periodontology, Peking University School and Hospital of Stomatology, 22 Zhongguancun Avenue South, Haidian District, Beijing 100081, China
4
Department of Oral and Maxillofacial Surgery, School of Medicine, Tokyo Women's Medical University, Tokyo 162-8666, Japan
*
Author to whom correspondence should be addressed.
Antioxidants 2015, 4(3), 577-590; https://doi.org/10.3390/antiox4030577
Submission received: 9 July 2015 / Revised: 11 August 2015 / Accepted: 18 August 2015 / Published: 2 September 2015
(This article belongs to the Special Issue Antioxidants and Periodontal Diseases)

Abstract

:
Periodontal disease is a highly prevalent disorder affecting up to 80% of the global population. Recent epidemiological studies have shown an association between periodontal disease and cardiovascular disease, as oxidative stress plays an important role in chronic inflammatory diseases such as periodontal disease and cardiovascular disease. In this review, we focus on the mechanisms by which periodontopathic bacteria cause chronic inflammation through the enhancement of oxidative stress and accelerate cardiovascular disease. Furthermore, we comment on the antioxidative activity of catechin in atherosclerosis accelerated by periodontitis.

1. Introduction

Periodontal disease, a chronic, destructive, and inflammatory condition affecting a large portion of the adult population, is a major cause of tooth loss and is characterized by a chronic infection associated with gram-negative anaerobic bacteria in the dental biofilm. It leads to the irreversible destruction of tissues supporting the teeth and is clinically detectable as periodontal pockets and alveolar bone loss [1,2]. Because periodontitis often causes bacteremia, a relationship between periodontitis and systemic diseases via periodontal pathogens has been explored [3]. Two or more periodontopathic bacteria have been detected in the cardiac valve [4,5] and in aortic aneurysm cases [6]; therefore, periodontal infection may also affect the progression of cardiovascular disease (CVD). However, many questions regarding this causal relationship and its pathological mechanism remain unanswered. This review discusses the involvement of oxidative stress caused by periodontopathic bacteria in the development of atherosclerosis. Furthermore, we mention the possibility of preventing atherosclerosis by the intake of catechin, which has both antioxidative and anti-inflammatory effects.

2. Increase in Oxidative Stress and Lipid Oxidation Caused by Infection

Periodontitis is a multifactorial disease, and the genetic background of patients as well as the presence of pathogenic bacteria and the immune mechanisms are thus important elements. The occurrence and progress of inflammatory responses in the periodontium are a result of the interaction between the bacteria that exists in the subgingival space and the immune cells in periodontal tissue that respond to them.
It is said that periodontopathogens injure tissue directly through harmful toxic products that induce cell death and tissue necrosis and that they do so indirectly through the activation of inflammatory cells that produce inflammatory cytokines [7]. However, many microbial products have little or no direct toxic effect on the host. Instead, they possess the potential to activate inflammatory reactions that cause tissue damage. It is now well accepted that it is the host’s response to the plaque bacteria, rather than microbial virulence per se, that directly causes the tissue damage. During the course of inflammation in response to dental plaque bacterial species, the gingival fibroblasts produce interleukin-1 (IL-1), IL-6, IL-8, tumor necrosis factor-a (TNF-α), and transforming growth factor beta (TGF-β) [8,9,10]. These cytokines and chemokines are an important mediator of the inflammatory responses, and some of them recruit polymorphonuclear leukocytes to the site of infection [11], which then produce reactive oxygen species (ROS) through NADPH oxidase catalyzation [12]. Oxidative DNA damage in fibroblasts and increased ROS production in polymorphonuclear leukocytes have been reported in experimental periodontitis in animal studies [13,14]. Patients with periodontitis also have increased levels of lipid peroxidation in plasma, saliva, and gingival crevicular fluid [15], and these levels have been correlated with the severity of periodontal disease [15,16]. Furthermore, the inflammatory response stimulates increased osteoclastic bone resorption, which degrades the alveolar bone supporting the teeth in the infected area. Overwhelming evidence indicates that oxidative stress plays a central role in the periodontal tissue and alveolar bone destruction [17,18]. Indeed, a recent clinical study indicated that an increase in plasma reactive oxygen metabolites (ROM) levels was associated with that in the clinical attachment level in periodontal patients whose plasma ROM level was low [19].
Mitochondria are the major producer of ROS in cells, and disruption of the structure and function of mitochondria are now thought to trigger numerous diseases such as atherosclerotic cardiovascular disease, obesity and type2 diabetes [20]. Chronic over-production of mitochondrial reactive oxygen species leads to increased oxidation of low-density lipoprotein (LDL) in atherosclerosis [21]. Therefore, lipid peroxidation also plays an important role in cardiovascular diseases [22]. In particular, LDL oxidation may be a key step in the development of atherosclerosis [23]. Oxidation of LDL is essential for its accumulation within macrophages and the formation of foam cells, which can upregulate proatherogenic chemokines and adhesion molecules [24] and induce IL-6, TNF-α, and C-reactive protein secretion [25]. Aggregatibacter actinomycetemcomitans infection of apolipoprotein E-deficient spontaneously hyperlipidemic (ApoEshl) mice promoted LDL oxidation, as indicated by marked upregulation of 4-hydroxynoneal, oxidated LDL (ox-LDL), and phospholipase A2 in the aorta; ox-LDL, 8-oxo-2′-deoxyguanosine, and myeloperoxidase serum levels; and aortic expression of nicotinamide adenine dinucleotide phosphate oxidase, caveolin-1, and the receptor for advanced glycation endproducts (RAGE) [26]. Porphyromonas gingivalis also increased oxidative modification of LDL [27,28] and rupture of atherosclerotic plaque through induction of matrix metalloproteinase (MMP) [29]. Co-incubation of a murine macrophage cell line with P. gingivalis in the presence of LDL resulted in the formation of foam cells in a dose-dependent manner [30].
These results suggest that periodontopathic bacteria contribute to lipid oxidation and further inflammatory induction in remote structures as well as the oral cavity.

3. Activation of the Inflammasome in Periodontal Disease and Atherosclerosis

Although the secretion of cytokines at inflammatory sites is initially protective in the elimination of infectious bacteria, excessive or sustained production of proinflammatory cytokines is related to periodontal destruction, with periodontal attachment loss and alveolar bone resorption. The cytokine concentration in gingival crevicular fluid (GCF) and saliva is higher in patients with aggressive periodontitis than in healthy subjects and decreases after periodontal treatment. [31,32]. Macrophages represent an important source of pro-inflammatory cytokines, including IL-1β and TNF-α; under dysregulation, these contribute to the destruction of host tissue. Thus, understanding the mechanisms of periodontal pathogen-induced immune cell signaling could provide useful information for the prevention and treatment of periodontitis.
P. gingivalis is a gram-negative asaccharolytic bacterium that has long been implicated in human periodontitis [33]. Recent evidence suggests that this bacterium contributes to periodontitis by functioning as a keystone pathogen [34,35]. Virulence factors of P. gingivalis, including lipopolysaccharide (LPS), hemagglutinins, gingipains, and fimbriae, are important in the induction of immune responses, including the production of inflammatory cytokines, such as IL-1, and the activation of inflammation-related signaling pathways [36]. IL-1 also mediates periodontal tissue destruction by stimulating alveolar bone resorption.
Inflammatory and immune mechanisms activated by infectious agents are also important in the development of atherosclerosis [37]. Several epidemiological studies have revealed that host immune reactions against persistent infectious pathogens, including P. gingivalis, may promote the development of atherosclerosis [38,39]. In this respect, the Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome may play an important role in IL-1β production in response to several bacterial ligands such as LPS, peptidoglycan, and bacterial RNA [40,41,42,43]. Peripheral blood mononuclear cell-derived macrophages reportedly express NLRP3 mRNA [44] to release proinflammatory IL-1β, which is highly induced by bacterial LPS [45]. In contrast, NLRP3-deficient macrophages do not produce active IL-1β in response to bacterial infection [40,41].
NLRP3 proteins were also upregulated in gingival tissues from periodontitis patients [46] and in the aorta of coronary atherosclerosis patients [47]. The oral challenge of ApoEshl mice with P. gingivalis showed significantly increased gingival and aortic NLRP3; pro-IL-1β, pro-IL-18, and pro-caspase-1 mRNA levels; and IL-1β, IL-18, and TNF-α peritoneal protein levels compared with gingipain- or fimbriae-deficient mutants, suggesting that gingipains and fimbriae are important virulence factors in NLRP3-inflammasome activation followed by inflammatory cytokine production [48]. Indeed, major fimbriae from P. gingivalis induced inflammatory cytokine production by murine peritoneal macrophages and monocytes [49]. Furthermore, gingipains from P. gingivalis induced proinflammatory cytokine production in human macrophages [50]. On the other hand, a previous report indicated that NLRP3-related proteins were not secreted into the culture medium due to degradation of the proteins by P. gingivalis protease, even if their expression was elevated [51]. However, sufficient levels of IL-1β and TNF-α were produced in cultured murine macrophages by repeated infection with P. gingivalis in vivo [48]. Because the in vitro culture of macrophages is performed using a closed system and an extended culture period, the direct effect of proteases may not reflect in vivo competence. In fact, elevated levels of IL-1β can be found in diseased periodontal tissue and gingival crevicular fluid as well as in saliva [46,52,53].

4. Activation of the Inflammasome by Oxidative Stress and oxLDL

Host cells produce ROS as a mechanism to combat intracellular microorganisms and as a signal mediator in the cells for various cellular functions [54,55,56]. However, persistent production of ROS causes induction of oxidative stress and may trigger cell death, inflammatory responses, and perturbation of tissue homeostasis in sequence [57]. Recent studies on chronic infections revealed that cytosolic ROS and oxidative-stress pathways play a role in the host metabolic pathway by modulating immune response and persistent infections in specialized immune cells [58,59,60,61].
Signaling by ROS also drives inflammasome activation. ROS activates the inflammasome through mitogen-activated protein kinases and extracellular signal-regulated protein kinases 1 and 2. Dysregulation of the inflammasome plays a significant role in various pathological processes. Recent animal studies show that ROS act as second messengers whose signaling drives NLRP3 inflammasome activation [62,63]. Furthermore, ox-LDL also induces secretion of IL-1b by human macrophages via ROS-dependent NLRP3 inflammasome activation [64]. Therefore, it is possible that the NLRP3 inflammasome is activated through ROS and ox-LDL produced due to infection, as well as through direct inflammasome activation due to infection by periodontal bacteria.

5. Persistence of Inflammation by Inflammasome and T helper 17 (Th17) Cell Activation

Activation of the inflammasome is critical for IL-1-driven inflammation and plays a central role in the pathology of autoinflammatory diseases. Recent studies have also shown that IL-1 or IL-18 can promote IL-17-prduction from Th17 cells and γδT cells. In general, IL-1-driven IL-17 production plays a critical role in host protective immunity to infection by fungi, bacteria, and certain viruses. However, in some settings, the sustained production of IL-17 from Th17 cells and γδT cells activated by inflammasome-derived IL-1 or IL-18 have major pathogenic roles in many autoimmune and chronic inflammatory diseases [65].
The presence of IL-17 has been demonstrated in human and experimental atherosclerotic tissues and plasma, and IL-17 might play a role in the pathogenesis of atherosclerosis [66,67]. For example, several studies have reported that IL-17 inhibition via the neutralization of antibodies or target-gene knockdown reduced atherosclerotic plaques in mice [66,68]. When P. gingivalis-challenged mice showed notable accumulation of atherosclerotic plaques by Oil Red O staining, the intracellular cytokine staining of spleen cells revealed significantly elevated CD4+ IL-17A+ T cells and slightly increased CD4+ Foxp3+ T cells, but the ratio of CD4+ interferon gamma (IFN-γ)+ T cells hardly changed [69]. IL-17 and IL-1β production and retinoic acid related orphan receptor γt (RORγt) expression were also significantly increased in the splenic cells of P. gingivalis-challenged mice. Furthermore, the expression of Th17-related genes, such as IL-6, TGF-β, RORγt, signal transducer and activator of transcription 3 (STAT3), and IL-23, were elevated in the splenic cells and heart tissue of P. gingivalis-challenged mice. These results suggest the possibility that ROS or ox-LDL formed due to infection by P. gingivalis activates Th17 cells through the NLRP3 inflammasome. Indeed, Lim et al. [70] reported that ox-LDL contributes to the development of Th17 in murine arteriosclerosis.
Recent studies have also demonstrated that P. gingivalis promotes Th17-inducing pathways in chronic periodontitis [71,72]. Persistence of the Th17 population has been shown to support chronic inflammation and to directly mediate tissue destruction through the activation of resident matrix cells such as fibroblasts and osteoclasts in vitro [73,74]. Therefore, uncovering the mechanisms through which P. gingivalis may contribute to the generation of chronic inflammatory tissue-destructive mechanisms may be relevant not only for periodontitis but also for several systemic inflammatory diseases such as atherosclerosis.
Serotype b Aa challenge also increased the murine splenic IL-17+CD4+ T cell population, Th-17-related serum cytokine levels (IL-17, IL-6, TGF-β, and IL-1β), Th17-related gene expression (IL-1β, IL-6, IL-17RA, STAT3, IL-21, and TGF-β), and atherosclerotic plaque formation in the aorta [75]. Furthermore, the Aa challenge increased the expression of Th17-related microRNA (miR-155 and miR146b) and the inflammasome-related molecules absent in melanoma 2 (AIM-2), Mincle, and NLRP3 in the aorta. These results suggest the possibility that the increase in Th17 cells observed in Aa-challenged mice is related to exacerbation of atherosclerosis. Serotype b Aa strains have been shown to have a greater capacity to trigger Th17-type cytokine production in vitro [76]. Serotype b strains are more frequently detected in patients with chronic and aggressive periodontitis than in healthy subjects [77,78]. Therefore, the association between the presence of serotype b Aa strains and periodontitis suggests the increased inflammatory potential of this species.
Because Aa challenge significantly increased AIM-2, Mincle, and NLRP3 mRNA expression in ApoEshl mice, recognition of innate and inflammatory signals by dendritic cells via pattern-recognition receptors may activate the intracellular pathways involved in CD4+ T cell differentiation into Th17 cells.
It was recently shown that miR-155 was expressed in mouse and human atherosclerosis lesion [79]. Further, miR-155 deficiency in Apolipoprotein E knockout (ApoE-KO) mice attenuated atherosclerosis by reducing macrophage inflammation and Th17 cell numbers. Indeed, miR-155 promotes the development of inflammatory Th17/Th1 cell subsets [80]. MicroRNA (MiR)-146b was also shown to be involved in the pathogenesis of murine viral myocarditis and Clostridium difficile-related colitis by regulating Th17 differentiation [81,82]. As Aa challenge significantly increased miR-155 and miR-146b microRNA expression in ApoEshl mice, Aa may regulate Th17 differentiation by enhancing miR-155 and miR-146b expression [75].

6. Prevention of Periodontal Disease and Atherosclerosis Using Antioxidants

A positive correlation between green tea consumption and cardiovascular health has been established in cohort or cross-sectional study [83]. Catechins are the main polyphenolic compounds in green tea. They exert vascular protective effects through multiple mechanisms, including anti-inflammatory, antioxidant, antihypertensive, anti-thrombogenic, and lipid lowering effects, that are effective in vascular protection [84]. Catechins perform antioxidant activity by scavenging free radicals, inhibiting redox active transcription factors, inhibiting pro-oxidant enzymes, and inducing antioxidant enzymes. They are effective in attenuating lipid biosynthesis via modulation of relevant enzymes and regulation of intestinal lipid absorption, thereby improving serum lipid profiles. These effects are critical in influencing the progression of atherosclerotic lesions and amelioration of cardiovascular diseases with improved endothelial function [85]. Atherosclerotic lesion areas of the aortic sinus caused by P. gingivalis infection decreased in epigallocatechin-3-gallate (EGCG)-treated ApoE-KO mice, wherein EGCG reduced the production of C-reactive protein, MCP-1, and oxidized LDL and slightly lowered LDL/very low-density lipoprotein (VLDL) cholesterol in P. gingivalis-challenged mice serum [86]. Furthermore, the increases in inflammatory- and oxidative stress-related mediators, such as chemokine (C-C motif) ligand 2 (CCL2), MMP-9, intercellular adhesion molecule-1(ICAM-1), heat shock protein 60 (HSP60), CD44, lectin-like receptor-1 for ox-LDL (LOX-1), NADPH oxidase 4 (NOX-4), p22phox, and inducible nitric oxide synthase (iNOS) gene expression levels in the aorta of P. gingivalis-challenged mice were reduced by EGCG-treatment. However, HO-1 mRNA levels were elevated by EGCG treatment, suggesting that EGCG inhibits P. gingivalis-induced atherosclerosis through anti-inflammatory and anti-oxidative effects [86]. Therefore, previous catechin consumption may be useful for the prevention of pathogen-accelerated atherosclerosis.
Positive correlation is also obtained application of antioxidant to periodontitis. Topical application of a dentifrice containing green tea catechin in an induced periodontitis rat model also reduced inflammatory cell infiltration in the periodontal lesions to a greater extent than did the control dentifrice [87]. Gingival expression of a lipid peroxidation marker hexanoyl-lysine, nitrotyrosine, a marker of oxidative protein damage, and TNF-α was reduced in response to the dentifrice with green tea catechin compared with the control dentifrice [87]. Therefore, addition of green tea catechin to a dentifrice may prove useful for the improvement of periodontal inflammation by reducing oxidative stress and the expression of pro-inflammatory cytokines. Recently, it was shown that catechin suppressed LPS-induced inflammatory bone resorption, and protected against alveolar bone loss in mice [88]. Catechin may be effective in preventing gingival and periodontal inflammation and bone loss. Several proteolytic enzymes, including gingipains, collagenases, and dipeptidyl aminopeptidase, produced by P. gingivalis also play important roles in the pathogenesis of periodontitis by hydrolyzing serum and tissue proteins, leading to tissue destruction [89]. Therefore, inhibitors of these enzymes may play important roles as valuable therapeutic agents. Polyphenols isolated from cranberry and green tea, which are natural plant-derived inhibitors of P. gingivalis proteases, are also effective in the inhibition of several proteases produced by P. gingivalis [89]. Furthermore, the epidemiological relationship between the intake of green tea and periodontal disease also demonstrated a significant reduction in periodontal pocket probing depths [90] and tooth loss [91]. These results indicate an association of catechin consumption with reduced odds for probing depths and tooth loss. A recent clinical trial also indicated that local application of green tea catechin gel in chronic periodontitis reduced periodontal pockets and inflammation [92]. Furthermore, locally delivered lycopene gel as an antioxidant in the treatment of chronic periodontitis was effective in increasing clinical attachment and reducing gingival inflammation, probing depth, and oxidative injury [93].

7. Conclusions

That oxidative stress and ox-LDL levels were increased in the blood and local sites of periodontal or atherosclerotic patients and in the mouse model suggests that lipid peroxidation activates the NLRP3 inflammasome and contributes to the activation of IL-17-producing Th17. Furthermore, it suggests the possibility that infection and sustained production of inflammatory cytokines, including IL-1β, IL-18, and IL-17, through lipid peroxidation results in chronic inflammation. On the other hand, treatment of catechin, a polyphenol with antioxidant, anti-inflammatory, and antithrombotic activities, protected mice from infection caused by periodontopathic bacteria and suppressed gingival inflammation, bone resorption, and arteriosclerotic progress. These results confirm the role of lipid peroxidation, as a correlation between periodontal disease and atherosclerosis, and can be a starting point for further research on the efficiency of different antioxidant agents in the prevention and treatment of periodontal disease and atherosclerosis. However, there are several limitations to the present study. The level of oxidative stress is a result of the balance between ROS production and anti-oxidant defense [94]. Although ascorbic acid and α-tocopherol are low-molecular-mass anti-oxidants that protect the cells from ROS and are a part of anti-oxidant defense system [95], their low concentration is associated with a higher risk of several chronic inflammatory diseases that are related to periodontal disease [96], diabetes [97], and coronary heart disease [98,99]. Increased plasma glutathione peroxidase concentration in GCF, which is an antioxidant in the human defense against oxidative stress, can also be considered as an indicator of a local increase in oxidative stress [100]. Therefore, the increase in ROS generation may have led to the occurrence of oxidative stress, which in turn caused an increased need for antioxidant production to establish the ROS-anti-oxidants balance to protect the tissues. Further study with larger samples and more complete periodontal health and disease information would be necessary to substantiate these findings.

Acknowledgments

This study was supported by Research funds by Grants-in-Aid for Scientific Research (26463145) from the Japan Society for the Promotion of Science and Nihon University Multidisciplinary Research Grant for (2014–2015).

Author Contributions

Tomoko Kurita-Ochiai and Masafumi Yamamoto conceived and designed the experiments; Yu Cai, Ru Jia and Yohei Yamaguchi performed the experiments and analyzed the data. Tomoko Kurita-Ochiai wrote the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Loesche, W.J.; Grossman, N.S. Periodontal disease as a specific, albeit chronic, infection: Diagnosis and treatment. Clin. Microbial. Rev. 2001, 14, 727–752. [Google Scholar] [CrossRef] [PubMed]
  2. Burt, B. Position paper: Epidemiology of periodontal diseases. J. Periodontal 2005, 76, 1406–1419. [Google Scholar] [CrossRef]
  3. Olsen, I. Update on bacteraemia related to dental procedures. Transfu. Apheresis Sci. 2008, 39, 173–178. [Google Scholar] [CrossRef] [PubMed]
  4. Nakano, K.; Inaba, H.; Nomura, R.; Nemoto, H.; Takeuchi, H.; Yoshioka, H.; Toda, K.; Taniguchi, K.; Amano, A.; Ooshima, T. Distribution of Porphyromonas gingivalis fimA genotypes in cardiovascular specimens from Japanese patients. Oral Microbial. Immunol. 2008, 23, 170–172. [Google Scholar] [CrossRef] [PubMed]
  5. Nakano, K.; Nemoto, H.; Nomura, R.; Inaba, H.; Yoshioka, H.; Taniguchi, K.; Amano, A.; Ooshima, T. Detection of oral bacteria in cardiovascular specimens. Oral Microbial. Immunol. 2009, 24, 64–68. [Google Scholar] [CrossRef] [PubMed]
  6. Kurihara, N.; Inoue, Y.; Iwai, T.; Umeda, M.; Huang, Y.; Ishikawa, I. Detection and localization of periodontopathic bacteria in abdominal aortic aneurysms. Eur. J. Vasc. Endovasc. Surg. 2004, 28, 553–558. [Google Scholar] [CrossRef] [PubMed]
  7. Page, R.C.; Kornman, K.S. The pathogenesis of human periodontitis: an introduction. Periodontol. 2000 1997, 14, 9–11. [Google Scholar] [CrossRef] [PubMed]
  8. Imatani, T.; Kato, T.; Okuda, K. Production of inflammatory cytokines by human gingival fibroblasts stimulated by cell-surface preparations of Porphyromonas gingivalis. Oral Microbial. Immunol. 2001, 16, 65–72. [Google Scholar] [CrossRef]
  9. Morandini, A.C.; Sipert, C.R.; Ramos-Junior, E.S.; Brozoski, D.T.; Santos, C.F. Periodontal ligament and gingival fibroblasts participate in the production of TGF-β, interleukin (IL)-8 and IL-10. Braz. Oral Res. 2011, 25, 157–162. [Google Scholar] [PubMed]
  10. Yucel-Lindberg, T.; Brunius, G. Epidermal growth factor synergistically enhances interleukin-8 production in human gingival fibroblasts stimulated with interleukin-1beta. Arch. Oral Boil. 2006, 51, 892–898. [Google Scholar] [CrossRef] [PubMed]
  11. Kobayashi, Y. The role of chemokines in neutrophil biology. Front. Biosci. 2008, 13, 2400–2407. [Google Scholar] [CrossRef] [PubMed]
  12. Giannopoulou, C.; Krause, K.H.; Muller, F. The NADPH oxidase NOX2 plays a role in periodontal pathologies. Semin. Immunopathol. 2008, 30, 273–278. [Google Scholar] [CrossRef] [PubMed]
  13. Tomofuji, T.; Azuma, T.; Kusano, H.; Sanbe, T.; Ekuni, D.; Tamaki, N.; Yamamoto, T.; Watanabe, T. Oxidative damage of periodontal tissue in the rat periodontitis model: Effects of a high-cholesterol diet. FEBS Lett. 2006, 580, 3601–3604. [Google Scholar] [CrossRef] [PubMed]
  14. Ekuni, D.; Tomofuji, T.; Tamaki, N.; Sanbe, T.; Azuma, T.; Yamanaka, R.; Yamamoto, T.; Watanabe, T. Mechanical stimulation of gingiva reduces plasma 8-OHdG level in rat periodontitis. Arch. Oral Boil. 2008, 53, 324–329. [Google Scholar] [CrossRef] [PubMed]
  15. Akalin, F.A.; Baltacioglu, E.; Alver, A.; Karabulut, E. Lipid peroxidation levels and total oxidant status in serum, saliva and gingival crevicular fluid in patients with chronic periodontitis. J. Clin. Periodontal 2007, 34, 558–565. [Google Scholar] [CrossRef] [PubMed]
  16. Bastos, A.S.; Graves, D.T.; Loureiro, A.P.D.M.; Rossa Junior, C.; Abdalla, D.S.P.; Faulin, T.E.S.; Olsen Camara, N.; Andriankaja, O.M.; Orrico, S.R. Lipid peroxidation is associated with the severity of periodontal disease and local inflammatory markers in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 2012, 97, E1353–E1362. [Google Scholar] [CrossRef] [PubMed]
  17. Katsuragi, H.; Ohtake, M.; Kurasawa, I.; Saito, K. Intracellular production and extracellular release of oxygen radicals by PMNs and oxidative stress on PMNs during phagocytosis of periodontopathic bacteria. Odontology 2003, 91, 13–18. [Google Scholar] [CrossRef] [PubMed]
  18. Sculley, D.V.; Langley-Evans, S.C. Salivary antioxidants and periodontal disease status. Proc. Nutr. Soc. 2002, 61, 137–143. [Google Scholar] [CrossRef] [PubMed]
  19. Machida, T.; Tomofuji, T.; Ekuni, D.; Yamane, M.; Yoneda, T.; Kawabata, Y.; Kataoka, K.; Tamaki, N.; Morita, M. Longitudinal relationship between plasma reactive oxygen metabolites and periodontal condition in the maintenance phase of periodontal treatment. Dis. Marker. 2014. [Google Scholar] [CrossRef] [PubMed]
  20. Bullon, P.; Newman, H.N.; Battino, M. Obesity, diabetes mellitus, atherosclerosis and chronic periodontitis: a shared pathology via oxidative stress and mitochondrial dysfunction? Periodontol. 2000 2014, 64, 139–153. [Google Scholar] [CrossRef] [PubMed]
  21. Madamanchi, N.R.; Runge, M.S. Mitochondrial dysfunction in atherosclerosis. Circ. Res. 2007, 100, 460–473. [Google Scholar] [CrossRef] [PubMed]
  22. Logani, M.K.; Davies, R.E. Lipid oxidation: biologic effects and antioxidants—A review. Lipids 1980, 15, 485–495. [Google Scholar] [CrossRef] [PubMed]
  23. Stocker, R.; Keaney, J.F., Jr. Role of oxidative modifications in atherosclerosis. Physiol. Rev. 2004, 84, 1381–1478. [Google Scholar] [CrossRef] [PubMed]
  24. Li, D.; Liu, L.; Chen, H.; Sawamura, T.; Mehta, J.L. LOX-1, an oxidized LDL endothelial receptor, induces CD40/CD40L signaling in human coronary artery endothelial cells. Arterioscler. Thromb. Vascu. Boil. 2003, 23, 816–821. [Google Scholar] [CrossRef] [PubMed]
  25. Hulthe, J.; Fagerberg, B. Circulating oxidized LDL is associated with increased levels of cell-adhesion molecules in clinically healthy 58-year old men (AIR study). Med. Sci. Monit. 2002, 8, CR148–CR152. [Google Scholar] [PubMed]
  26. Jia, R.; Kurita-Ochiai, T.; Oguchi, S.; Yamamoto, M. Periodontal pathogen accelerates lipid peroxidation and atherosclerosis. J. Dent. Res. 2013, 92, 247–252. [Google Scholar] [CrossRef] [PubMed]
  27. Hashimoto, M.; Kadowaki, T.; Tsukuba, T.; Yamamoto, K. Selective proteolysis of apolipoprotein B-100 by Arg-gingipain mediates atherosclerosis progression accelerated by bacterial exposure. J. Biochem. 2006, 140, 713–723. [Google Scholar] [CrossRef] [PubMed]
  28. Bengtsson, T.; Karlsson, H.; Gunnarsson, P.; Skoglund, C.; Elison, C.; Leanderson, P.; Lindahl, M. The periodontal pathogen Porphyromonas gingivalis cleaves apoB-100 and increases the expression of apoM in LDL in whole blood leading to cell proliferation. J. Intern. Med. 2008, 263, 558–571. [Google Scholar] [CrossRef] [PubMed]
  29. Ding, Y.; Uitto, V.J.; Firth, J.; Salo, T.; Haapasalo, M.; Konttinen, Y.T.; Sorsa, T. Modulation of host matrix metalloproteinases by bacterial virulence factors relevant in human periodontal diseases. Oral Dis. 1995, 1, 279–286. [Google Scholar] [CrossRef] [PubMed]
  30. Miyakawa, H.; Honma, K.; Qi, M.; Kuramitsu, H.K. Interaction of Porphyromonas gingivalis with low-density lipoproteins: Implications for a role for periodontitis in atherosclerosis. J. Periodontal Res. 2004, 39, 1–9. [Google Scholar] [CrossRef] [PubMed]
  31. Hernandez, M.; Dutzan, N.; Garcia-Sesnich, J.; Abusleme, L.; Dezerega, A.; Silva, N.; Gonzalez, F.E.; Vernal, R.; Sorsa, T.; Gamonal, J. Host-pathogen interactions in progressive chronic periodontitis. J. Dent. Res. 2011, 90, 1164–1170. [Google Scholar] [CrossRef] [PubMed]
  32. Yue, Y.; Liu, Q.; Xu, C.; Loo, W.T.; Wang, M.; Wen, G.; Cheung, M.N.; Bai, L.J.; Dou, Y.D.; Chow, L.W.; et al. Comparative evaluation of cytokines in gingival crevicular fluid and saliva of patients with aggressive periodontitis. Int. J. Boil. Marker. 2013, 28, 108–112. [Google Scholar] [CrossRef] [PubMed]
  33. Socransky, S.S.; Haffajee, A.D.; Cugini, M.A.; Smith, C.; Kent, R.L., Jr. Microbial complexes in subgingival plaque. J. Clin. Periodontal 1998, 25, 134–144. [Google Scholar] [CrossRef]
  34. 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]
  35. Darveau, R.P.; Hajishengallis, G.; Curtis, M.A. Porphyromonas gingivalis as a potential community activist for disease. J. Dent. Res. 2012, 91, 816–820. [Google Scholar] [CrossRef] [PubMed]
  36. Bostanci, N.; Belibasakis, G.N. Porphyromonas gingivalis: An invasive and evasive opportunistic oral pathogen. FEMS Microbial. Lett. 2012, 333, 1–9. [Google Scholar] [CrossRef] [PubMed]
  37. Epstein, S.E.; Zhou, Y.F.; Zhu, J. Infection and atherosclerosis: Emerging mechanistic paradigms. Circulation 1999, 100, e20–e28. [Google Scholar] [CrossRef] [PubMed]
  38. Taniguchi, A.; Nishimura, F.; Murayama, Y.; Nagasaka, S.; Fukushima, M.; Sakai, M.; Yoshii, S.; Kuroe, A.; Suzuki, H.; Iwamoto, Y.; et al. Porphyromonas gingivalis infection is associated with carotid atherosclerosis in non-obese Japanese type 2 diabetic patients. Metab. Clin. Exp. 2003, 52, 142–145. [Google Scholar] [CrossRef] [PubMed]
  39. Wada, K.; Kamisaki, Y. Roles of oral bacteria in cardiovascular diseases—From molecular mechanisms to clinical cases: Involvement of Porphyromonas gingivalis in the development of human aortic aneurysm. J. Pharmacol. Sci. 2010, 113, 115–119. [Google Scholar] [CrossRef] [PubMed]
  40. Kanneganti, T.D.; Ozoren, N.; Body-Malapel, M.; Amer, A.; Park, J.H.; Franchi, L.; Whitfield, J.; Barchet, W.; Colonna, M.; Vandenabeele, P.; et al. Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature 2006, 440, 233–236. [Google Scholar] [CrossRef] [PubMed]
  41. Mariathasan, S.; Weiss, D.S.; Newton, K.; McBride, J.; O’Rourke, K.; Roose-Girma, M.; Lee, W.P.; Weinrauch, Y.; Monack, D.M.; Dixit, V.M. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 2006, 440, 228–232. [Google Scholar] [CrossRef] [PubMed]
  42. Martinon, F.; Agostini, L.; Meylan, E.; Tschopp, J. Identification of bacterial muramyl dipeptide as activator of the NALP3/cryopyrin inflammasome. Curr. Boil. 2004, 14, 1929–1934. [Google Scholar] [CrossRef] [PubMed]
  43. Becker, C.E.; O’Neill, L.A. Inflammasomes in inflammatory disorders: The role of TLRs and their interactions with NLRs. Semin. Immunopathol. 2007, 29, 239–248. [Google Scholar] [CrossRef] [PubMed]
  44. Kummer, J.A.; Broekhuizen, R.; Everett, H.; Agostini, L.; Kuijk, L.; Martinon, F.; van Bruggen, R.; Tschopp, J. Inflammasome components NALP 1 and 3 show distinct but separate expression profiles in human tissues suggesting a site-specific role in the inflammatory response. J. Histochem. Cytochem. 2007, 55, 443–452. [Google Scholar] [CrossRef] [PubMed]
  45. Pontillo, A.; Girardelli, M.; Agostinis, C.; Masat, E.; Bulla, R.; Crovella, S. Bacterial LPS differently modulates inflammasome gene expression and IL-1beta secretion in trophoblast cells, decidual stromal cells, and decidual endothelial cells. Reprod. Sci. 2013, 20, 563–566. [Google Scholar] [CrossRef] [PubMed]
  46. Park, E.; Na, H.S.; Song, Y.R.; Shin, S.Y.; Kim, Y.M.; Chung, J. Activation of NLRP3 and AIM2 inflammasomes by Porphyromonas gingivalis infection. Infect. Immune. 2014, 82, 112–123. [Google Scholar] [CrossRef] [PubMed]
  47. Zheng, F.; Xing, S.; Gong, Z.; Xing, Q. NLRP3 inflammasomes show high expression in aorta of patients with atherosclerosis. Heart Lung Circ. 2013, 22, 746–750. [Google Scholar] [CrossRef] [PubMed]
  48. Yamaguchi, Y.; Kurita-Ochiai, T.; Kobayashi, R.; Suzuki, T.; Ando, T. Activation of the NLRP3 inflammasome in Porphyromonas gingivalis-accelerated atherosclerosis. Pathog. Dis. 2015, 73. [Google Scholar] [CrossRef] [PubMed]
  49. Hajishengallis, G.; Sojar, H.; Genco, R.J.; DeNardin, E. Intracellular signaling and cytokine induction upon interactions of Porphyromonas gingivalis fimbriae with pattern-recognition receptors. Immunol. Investig. 2004, 33, 157–172. [Google Scholar] [CrossRef]
  50. Fitzpatrick, R.E.; Aprico, A.; Wijeyewickrema, L.C.; Pagel, C.N.; Wong, D.M.; Potempa, J.; Mackie, E.J.; Pike, R.N. High molecular weight gingipains from Porphyromonas gingivalis induce cytokine responses from human macrophage-like cells via a nonproteolytic mechanism. J. Innate Immun. 2009, 1, 109–117. [Google Scholar] [CrossRef] [PubMed]
  51. Huck, O.; Elkaim, R.; Davideau, J.L.; Tenenbaum, H. Porphyromonas gingivalis-impaired innate immune response via NLRP3 proteolysis in endothelial cells. Innate Immun. 2015, 21, 65–72. [Google Scholar] [CrossRef] [PubMed]
  52. Kaushik, R.; Yeltiwar, R.K.; Pushpanshu, K. Salivary interleukin-1beta levels in patients with chronic periodontitis before and after periodontal phase I therapy and healthy controls: A case-control study. J. Periodontal 2011, 82, 1353–1359. [Google Scholar] [CrossRef] [PubMed]
  53. Konopka, L.; Pietrzak, A.; Brzezinska-Blaszczyk, E. Effect of scaling and root planing on interleukin-1beta, interleukin-8 and MMP-8 levels in gingival crevicular fluid from chronic periodontitis patients. J. Periodontal Res. 2012, 47, 681–688. [Google Scholar] [CrossRef] [PubMed]
  54. Spooner, R.; Yilmaz, O. The role of reactive-oxygen-species in microbial persistence and inflammation. Int. J. Mol. Sci. 2011, 12, 334–352. [Google Scholar] [CrossRef] [PubMed]
  55. Said-Sadier, N.; Padilla, E.; Langsley, G.; Ojcius, D.M. Aspergillus fumigatus stimulates the NLRP3 inflammasome through a pathway requiring ROS production and the Syk Tyrosine kinase. PloS ONE 2010, 5. [Google Scholar] [CrossRef] [PubMed]
  56. Abdul-Sater, A.A.; Said-Sadier, N.; Lam, V.M.; Singh, B.; Pettengill, M.A.; Soares, F.; Tattoli, I.; Lipinski, S.; Girardin, S.E.; Rosenstiel, P.; et al. Enhancement of reactive oxygen species production and chlamydial infection by the mitochondrial Nod-like family member NLRX1. J. Boil. Chem. 2010, 285, 41637–41645. [Google Scholar] [CrossRef] [PubMed]
  57. Pourova, J.; Kottova, M.; Voprsalova, M.; Pour, M. Reactive oxygen and nitrogen species in normal physiological processes. Acta Physiol. 2010, 198, 15–35. [Google Scholar] [CrossRef] [PubMed]
  58. Cruz, C.M.; Rinna, A.; Forman, H.J.; Ventura, A.L.; Persechini, P.M.; Ojcius, D.M. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J. Biol. Chem. 2007, 282, 2871–2879. [Google Scholar] [CrossRef] [PubMed]
  59. Forman, H.J.; Maiorino, M.; Ursini, F. Signaling functions of reactive oxygen species. Biochemistry 2010, 49, 835–842. [Google Scholar] [CrossRef] [PubMed]
  60. Hasan, N.; Yusuf, N.; Toossi, Z.; Islam, N. Suppression of Mycobacterium tuberculosis induced reactive oxygen species (ROS) and TNF-α mRNA expression in human monocytes by allicin. FEBS Lett. 2006, 580, 2517–2522. [Google Scholar] [CrossRef] [PubMed]
  61. Yang, C.S.; Lee, H.M.; Lee, J.Y.; Kim, J.A.; Lee, S.J.; Shin, D.M.; Lee, Y.H.; Lee, D.S.; El-Benna, J.; Jo, E.K. Reactive oxygen species and p47phox activation are essential for the Mycobacterium tuberculosis-induced pro-inflammatory response in murine microglia. J. Neuroinflamm. 2007, 4, 27. [Google Scholar] [CrossRef] [PubMed]
  62. Fukumoto, J.; Fukumoto, I.; Parthasarathy, P.T.; Cox, R.; Huynh, B.; Ramanathan, G.K.; Venugopal, R.B.; Allen-Gipson, D.S.; Lockey, R.F.; Kolliputi, N. NLRP3 deletion protects from hyperoxia-induced acute lung injury. Cell Physiol. 2013, 305, C182–C189. [Google Scholar] [CrossRef] [PubMed]
  63. Heid, M.E.; Keyel, P.A.; Kamga, C.; Shiva, S.; Watkins, S.C.; Salter, R.D. Mitochondrial reactive oxygen species induces NLRP3-dependent lysosomal damage and inflammasome activation. J. Immun. 2013, 191, 5230–5238. [Google Scholar] [CrossRef] [PubMed]
  64. Jiang, Y.; Wang, M.; Huang, K.; Zhang, Z.; Shao, N.; Zhang, Y.; Wang, W.; Wang, S. Oxidized low-density lipoprotein induces secretion of interleukin-1beta by macrophages via reactive oxygen species-dependent NLRP3 inflammasome activation. Biochem. Biophys. Res. Commun. 2012, 425, 121–126. [Google Scholar] [CrossRef] [PubMed]
  65. Mills, K.H.; Dungan, L.S.; Jones, S.A.; Harris, J. The role of inflammasome-derived IL-1 in driving IL-17 responses. J. Leukoc. Biol. 2013, 93, 489–497. [Google Scholar] [CrossRef] [PubMed]
  66. Smith, E.; Prasad, K.M.; Butcher, M.; Dobrian, A.; Kolls, J.K.; Ley, K.; Galkina, E. Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E-deficient mice. Circulation 2010, 121, 1746–1755. [Google Scholar] [CrossRef] [PubMed]
  67. Gao, Q.; Jiang, Y.; Ma, T.; Zhu, F.; Gao, F.; Zhang, P.; Guo, C.; Wang, Q.; Wang, X.; Ma, C.; et al. A critical function of Th17 proinflammatory cells in the development of atherosclerotic plaque in mice. J. Immun. 2010, 185, 5820–5827. [Google Scholar] [CrossRef] [PubMed]
  68. Usui, F.; Kimura, H.; Ohshiro, T.; Tatsumi, K.; Kawashima, A.; Nishiyama, A.; Iwakura, Y.; Ishibashi, S.; Takahashi, M. Interleukin-17 deficiency reduced vascular inflammation and development of atherosclerosis in Western diet-induced ApoE-deficient mice. Biochem. Biophys. Res. Commun. 2012, 420, 72–77. [Google Scholar] [CrossRef] [PubMed]
  69. Cai, Y.; Kobayashi, R.; Hashizume-Takizawa, T.; Kurita-Ochiai, T. Porphyromonas gingivalis infection enhances Th17 responses for development of atherosclerosis. Arch. Oral Biol. 2014, 59, 1183–1191. [Google Scholar] [CrossRef] [PubMed]
  70. Lim, H.; Kim, Y.U.; Sun, H.; Lee, J.H.; Reynolds, J.M.; Hanabuchi, S.; Wu, H.; Teng, B.B.; Chung, Y. Proatherogenic conditions promote autoimmune T helper 17 cell responses in vivo. Immunity 2014, 40, 153–165. [Google Scholar] [CrossRef] [PubMed]
  71. Moutsopoulos, N.M.; Kling, H.M.; Angelov, N.; Jin, W.; Palmer, R.J.; Nares, S.; Osorio, M.; Wahl, S.M. Porphyromonas gingivalis promotes Th17 inducing pathways in chronic periodontitis. J. Autoimmun. 2012, 39, 294–303. [Google Scholar] [CrossRef] [PubMed]
  72. Adibrad, M.; Deyhimi, P.; Ganjalikhani Hakemi, M.; Behfarnia, P.; Shahabuei, M.; Rafiee, L. Signs of the presence of Th17 cells in chronic periodontal disease. J. Periodontal Res. 2012, 47, 525–531. [Google Scholar] [CrossRef] [PubMed]
  73. Takayanagi, H. Osteoimmunology and the effects of the immune system on bone. Nat. Rev. Rheumatol. 2009, 5, 667–676. [Google Scholar] [CrossRef] [PubMed]
  74. Sato, K.; Suematsu, A.; Okamoto, K.; Yamaguchi, A.; Morishita, Y.; Kadono, Y.; Tanaka, S.; Kodama, T.; Akira, S.; Iwakura, Y.; et al. Th17 functions as an osteoclastogenic helper T cell subset that links T cell activation and bone destruction. J. Exp. Med. 2006, 203, 2673–2682. [Google Scholar] [CrossRef] [PubMed]
  75. Jia, R.; Hashizume-Takizawa, T.; Du, Y.; Yamamoto, M.; Kurita-Ochiai, T. Aggregatibacter actinomycetemcomitans induces Th17 cells in atherosclerotic lesions. Pathog. Dis. 2015, 73. [Google Scholar] [CrossRef] [PubMed]
  76. Diaz-Zuniga, J.; Yanez, J.P.; Alvarez, C.; Melgar-Rodriguez, S.; Hernandez, M.; Sanz, M.; Vernal, R. Serotype-dependent response of human dendritic cells stimulated with Aggregatibacter actinomycetemcomitans. J. Clin. Periodontal 2014, 41, 242–251. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, C.; Wang, T.; Chen, W. Occurrence of Aggregatibacter actinomycetemcomitans serotypes in subgingival plaque from United States subjects. Mol. Oral Microbial. 2010, 25, 207–214. [Google Scholar] [CrossRef] [PubMed]
  78. Cortelli, J.R.; Aquino, D.R.; Cortelli, S.C.; Roman-Torres, C.V.; Franco, G.C.; Gomez, R.S.; Batista, L.H.; Costa, F.O. Aggregatibacter actinomycetemcomitans serotypes infections and periodontal conditions: A two-way assessment. Eur. J. Clin. Microbial. Infect. Dis. 2012, 31, 1311–1318. [Google Scholar] [CrossRef] [PubMed]
  79. Du, F.; Yu, F.; Wang, Y.; Hui, Y.; Carnevale, K.; Fu, M.; Lu, H.; Fan, D. MicroRNA-155 deficiency results in decreased macrophage inflammation and attenuated atherogenesis in apolipoprotein E-deficient mice. Arterioscler. Thromb. Vascu. Biol. 2014, 34, 759–767. [Google Scholar] [CrossRef] [PubMed]
  80. Zhang, J.; Cheng, Y.; Cui, W.; Li, M.; Li, B.; Guo, L. MicroRNA-155 modulates Th1 and Th17 cell differentiation and is associated with multiple sclerosis and experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2014, 266, 56–63. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, Y.L.; Wu, W.; Xue, Y.; Gao, M.; Yan, Y.; Kong, Q.; Pang, Y.; Yang, F. MicroRNA-21 and -146b are involved in the pathogenesis of murine viral myocarditis by regulating TH-17 differentiation. Arch. Virol. 2013, 158, 1953–1963. [Google Scholar] [CrossRef] [PubMed]
  82. Viladomiu, M.; Hontecillas, R.; Pedragosa, M.; Carbo, A.; Hoops, S.; Michalak, P.; Michalak, K.; Guerrant, R.L.; Roche, J.K.; Warren, C.A.; et al. Modeling the role of peroxisome proliferator-activated receptor gamma and microRNA-146 in mucosal immune responses to Clostridium difficile. PloS ONE 2012, 7. [Google Scholar] [CrossRef] [PubMed]
  83. Kuriyama, S. The relation between green tea consumption and cardiovascular disease as evidenced by epidemiological studies. J. Nutr. 2008, 138, 1548S–1553S. [Google Scholar] [PubMed]
  84. Babu, P.V.; Liu, D. Green tea catechins and cardiovascular health: an update. Curr. Med. Chem. 2008, 15, 1840–1850. [Google Scholar] [CrossRef] [PubMed]
  85. Soory, M. Inflammatory mechanisms and redox status in periodontal and cardiometabolic diseases: Effects of adjunctive nutritional antioxidants and statins. Infect. Disord. Drug Targets 2012, 12, 301–315. [Google Scholar] [CrossRef] [PubMed]
  86. Cai, Y.; Kurita-Ochiai, T.; Hashizume, T.; Yamamoto, M. Green tea epigallocatechin-3-gallate attenuates Porphyromonas gingivalis-induced atherosclerosis. Pathog. Dis. 2013, 67, 76–83. [Google Scholar] [CrossRef] [PubMed]
  87. Maruyama, T.; Tomofuji, T.; Endo, Y.; Irie, K.; Azuma, T.; Ekuni, D.; Tamaki, N.; Yamamoto, T.; Morita, M. Supplementation of green tea catechins in dentifrices suppresses gingival oxidative stress and periodontal inflammation. Arch. Oral Biol. 2011, 56, 48–53. [Google Scholar] [CrossRef] [PubMed]
  88. Tominari, T.; Matsumoto, C.; Watanabe, K.; Hirata, M.; Grundler, F.M.; Miyaura, C.; Inada, M. Epigallocatechin gallate (EGCG) suppresses lipopolysaccharide-induced inflammatory bone resorption, and protects against alveolar bone loss in mice. FEBS Open Bio. 2015, 5, 522–527. [Google Scholar] [CrossRef] [PubMed]
  89. Grenier, D.; La, V.D. Proteases of Porphyromonas gingivalis as important virulence factors in periodontal disease and potential targets for plant-derived compounds: a review article. Curr. Drug Targets 2011, 12, 322–331. [Google Scholar] [CrossRef] [PubMed]
  90. Kushiyama, M.; Shimazaki, Y.; Murakami, M.; Yamashita, Y. Relationship between intake of green tea and periodontal disease. J. Periodontal 2009, 80, 372–377. [Google Scholar] [CrossRef] [PubMed]
  91. Koyama, Y.; Kuriyama, S.; Aida, J.; Sone, T.; Nakaya, N.; Ohmori-Matsuda, K.; Hozawa, A.; Tsuji, I. Association between green tea consumption and tooth loss: Cross-sectional results from the Ohsaki Cohort 2006 Study. Prev. Med. 2010, 50, 173–179. [Google Scholar] [CrossRef] [PubMed]
  92. Chava, V.K.; Vedula, B.D. Thermo-reversible green tea catechin gel for local application in chronic periodontitis: A 4-week clinical trial. J. Periodontal 2013, 84, 1290–1296. [Google Scholar] [CrossRef] [PubMed]
  93. Chandra, R.V.; Sandhya, Y.P.; Nagarajan, S.; Reddy, B.H.; Naveen, A.; Murthy, K.R. Efficacy of lycopene as a locally delivered gel in the treatment of chronic periodontitis: Smokers vs. nonsmokers. Quintessence Intern. 2012, 43, 401–411. [Google Scholar]
  94. Sies, H. Oxidative stress: Introduction. In Oxidative Stress: Oxidants and Antioxidants; Sies, H., Ed.; Academic Press: London, UK, 1991; pp. 15–21. [Google Scholar]
  95. Halliwell, B. Free radicals and antioxidants: A personal view. Nutr. Rev. 1994, 52, 253–265. [Google Scholar] [CrossRef] [PubMed]
  96. Iwasaki, M.; Manz, M.C.; Taylor, G.W.; Yoshihara, A.; Miyazaki, H. Relations of serum ascorbic acid and alpha-tocopherol to periodontal disease. J. Dent. Res. 2012, 91, 167–172. [Google Scholar] [CrossRef] [PubMed]
  97. Skrha, J.; Prazny, M.; Hilgertova, J.; Weiserova, H. Serum alpha-tocopherol and ascorbic acid concentrations in type 1 and type 2 diabetic patients with and without angiopathy. Clin. Chimi. Acta 2003, 329, 103–108. [Google Scholar] [CrossRef]
  98. Vita, J.A.; Keaney, J.F., Jr.; Raby, K.E.; Morrow, J.D.; Freedman, J.E.; Lynch, S.; Koulouris, S.N.; Hankin, B.R.; Frei, B. Low plasma ascorbic acid independently predicts the presence of an unstable coronary syndrome. J. Am. Coll. Cardiol. 1998, 31, 980–986. [Google Scholar] [CrossRef]
  99. Spencer, A.P.; Carson, D.S.; Crouch, M.A. Vitamin E and coronary artery disease. Arch. Inter. Med. 1999, 159, 1313–1320. [Google Scholar] [CrossRef]
  100. Patel, S.P.; Rao, N.S.; Pradeep, A.R. Effect of nonsurgical periodontal therapy on crevicular fluid and serum glutathione peroxidase levels. Dis. Marker. 2012, 32, 1–7. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Kurita-Ochiai, T.; Jia, R.; Cai, Y.; Yamaguchi, Y.; Yamamoto, M. Periodontal Disease-Induced Atherosclerosis and Oxidative Stress. Antioxidants 2015, 4, 577-590. https://doi.org/10.3390/antiox4030577

AMA Style

Kurita-Ochiai T, Jia R, Cai Y, Yamaguchi Y, Yamamoto M. Periodontal Disease-Induced Atherosclerosis and Oxidative Stress. Antioxidants. 2015; 4(3):577-590. https://doi.org/10.3390/antiox4030577

Chicago/Turabian Style

Kurita-Ochiai, Tomoko, Ru Jia, Yu Cai, Yohei Yamaguchi, and Masafumi Yamamoto. 2015. "Periodontal Disease-Induced Atherosclerosis and Oxidative Stress" Antioxidants 4, no. 3: 577-590. https://doi.org/10.3390/antiox4030577

Article Metrics

Back to TopTop