Skip to main content
Frontiers in Physiology logoLink to Frontiers in Physiology
. 2017 Dec 14;8:1055. doi: 10.3389/fphys.2017.01055

Oxidative Stress and Antioxidants in the Diagnosis and Therapy of Periodontitis

L'ubomíra Tóthová 1,2, Peter Celec 1,3,4,*
PMCID: PMC5735291  PMID: 29311982

Abstract

Oxidative stress has been implicated in the pathogenesis of numerous diseases. However, large interventional studies with antioxidants failed to show benefits in the prevention or treatment of cardiovascular diseases, cancer, or diabetes mellitus. Numerous clinical studies have confirmed the association of oxidative stress markers and periodontitis. Technical and biological variability is high for most of the analyzed markers and none of them seems to be optimal for routine clinical use. In a research setting, analysis of a palette of oxidative stress markers is needed to cover lipid peroxidation, protein oxidation, and the antioxidant status. The source of reactive oxygen species and their role in the pathogenesis of periodontitis remains unclear. Interventional experiments indicate that oxidative stress might be more than just a simple consequence of the inflammation. Small studies have confirmed that some antioxidants could have therapeutic value at least as an addition to the standard non-surgical treatment of periodontitis. A clear evidence for the efficiency of antioxidant treatment in large patient cohorts is lacking. Potentially, because lowering of oxidative stress markers might be a secondary effect of anti-inflammatory or antibacterial agents. As the field of research of oxidative stress in periodontitis gains attraction and the number of relevant published papers is increasing a systematic overview of the conducted observational and interventional studies is needed. This review summarizes the currently available literature linking oxidative stress and periodontitis and points toward the potential of adjuvant antioxidant treatment, especially in cases where standard treatment fails to improve the periodontal status.

Keywords: reactive oxygen species, free radicals, antioxidative therapy, systematic review, oral diseases

Introduction

Oxidative stress is both, a pathomechanism involved in numerous inflammatory diseases causing damage to lipids, nucleic acids and proteins—oxidative distress, as well as an important physiological process that enables the immune system to cope with microorganisms and intracellular cell signaling—oxidative eustress (Sies et al., 2017). Which edge of the sword is the dominant depends on the delicate balance between the production of reactive oxygen/nitrogen species and the antioxidant capacity of the tissue. The physiological functions of free radicals have been neglected for years and so, much more is known about the pathological role of oxidative stress. A variety of free radicals is produced and interacts with a variety of substrates. This leads to a palette of biomarkers that can be used for the assessment of oxidative stress-induced damage (Frijhoff et al., 2015).

Oxidative stress is usually defined as a disbalance of the production of free radicals and antioxidant mechanisms (Kopáni et al., 2006). However, free radicals are not a simple negative byproduct of oxygen metabolism. They are involved in immune responses, liver metabolism, but also in intracellular signaling pathways (Espinosa-Diez et al., 2015). The mechanisms involved in the physiological intracellular role of free radicals include the modulation of cysteine residues of redox-sensitive enzymes and other regulatory proteins (Finkel, 2011; Russell and Cotter, 2015). It has been hypothesized that under physiological conditions even high concentrations of any one primary reactive oxygen or nitrogen species does not lead to oxidative damage, as the cell has preventive and reparative tools to cope with the radical. The reactions of superoxide, nitric oxide, and other primary reactive species are reversible and are ideal for intracellular signaling (Weidinger and Kozlov, 2015). Thus, measuring oxidative stress using any one marker can lead to wrong interpretations. This might include the hope that administration of antioxidants will effectively treat diseases associated with oxidative stress. As recently hypothesized, one of the prototypic oxidative stress diseases—diabetes mellitus might be actually a consequence of deficiency of reactive oxygen species rather than of oxidative damage (Watson, 2014). It should not escape our notice that this proposed mechanism might suggest that antioxidants could increase the risk rather than prevent metabolic diseases.

Periodontitis is an inflammatory disease affecting supporting structures of the teeth leading at the end to loss of alveolar bone and teeth (Kinane et al., 2017). The main causative factor are microorganisms that colonize the subgingival dental plaque inducing an inflammatory host response. The inflammation affects, however, also the surrounding healthy tissue ultimately leading to the destruction of the periodontium (Kinane et al., 2011). Although lipopolysaccharide and proteolytic enzymes are essential in periodontitis, exaggerated inflammatory response, genetic predisposition, smoking, bad oral hygiene, and malnutrition are important in pathogenesis of periodontitis as well (Laine et al., 2012).

The role of oxidative stress in periodontitis has been postulated already decades ago (Shapira et al., 1991; Chapple, 1997). However, the suggested involvement was not clear. Some studies showed that leukocytes from patients with periodontitis are exhausted and have a low oxidation activity (Loesche et al., 1988), other studies pointed toward higher production of free radicals by leukocytes from periodontitis patients (Kimura et al., 1993). The contradictory findings from these studies might be related to the dynamics of the mechanisms during the pathogenesis of the disease, but they might also be explained by the different forms of periodontitis (Biasi et al., 1999).

The term oxidative stress is vague, similarly, antioxidants might affect many processes not directly related to free radical generation or action (Niki, 2016). This makes the interpretation of studies focusing on oxidative stress in periodontitis difficult. A systematic review of these clinical studies and animal experiments might, thus, be needed.

Biomarkers of oxidative stress and pathogenesis of periodontitis

Surprisingly, many observational studies analyzing oxidative stress in patients with periodontitis had relatively consistent results with higher oxidative stress markers in either saliva or blood and/or decreased antioxidant status in comparison to controls. The summary table of the identified studies can be found in Table 1. One of the largest observational studies has shown that the antioxidant status in blood, analyzed as vitamin C, bilirubin, and calculated total antioxidant capacity was inversely associated with mild and severe periodontitis (Chapple et al., 2007b). The more severe periodontitis, the clearer the association. Additionally, in a subgroup of never-smokers the antioxidants seemed to protect against development of periodontitis. Others smaller studies confirmed these results. Total antioxidant capacity was lower in plasma or serum of patients with chronic periodontitis (Chapple et al., 2002; Brock et al., 2004; Konopka et al., 2007). Similarly, superoxide dismutase activity (Huang et al., 2014) along with catalase and glutathione peroxidase activity (Tonguç et al., 2011) as important contributor to the total antioxidant capacity was found to be lower in periodontitis. In line with these results from blood, in the majority of studies, the antioxidant status was lower also locally—in saliva. The total antioxidant status/potential/capacity is in general the ability of a tissue to resist artificially induced oxidative stress, but studies differ in the analytical approaches. Nevertheless, the local antioxidant capacity was found to be lower in saliva from patients with an aggressive form of periodontitis when compared to chronic periodontitis (Acquier et al., 2017). Patients with chronic periodontitis have lower antioxidant capacity than control patients (Zhang et al., 2015; Ahmadi-Motamayel et al., 2017). Although the total antioxidant capacity is not a specific marker of antioxidant power, uric acid, glutathione peroxidase (Miricescu et al., 2014), and reduced glutathione (Gumus et al., 2009) as specific antioxidants were reported to be significantly lower in saliva of patients with chronic or aggressive periodontitis. On contrary, activities of the major antioxidant enzymes were found to be higher in chronic periodontitis patients in all investigated samples, i.e., plasma, erythrocytes, and in the gingival tissue (Panjamurthy et al., 2005). Similarly, SOD2 and GPX1 genes were overexpressed in the gingiva of chronic periodontitis patients (Duarte et al., 2012). Lactoferrin, myeloperoxidase and interleukin 1 beta were all positively correlated with the clinical markers of periodontal damage (Wei et al., 2004). However, whether such associations of higher antioxidant and pro-inflammatory response are a consequence or cause of severe periodontitis cannot be judged only from observations.

Table 1.

Observational studies analyzing oxidative stress in periodontitis.

Study design Sample size Note Sample type Outcome References
Cross-sectional study 20 AgP patients 20 ChP patients 20 controls Saliva ↑ ROS, TBARS with AgP; TRAP ↓ with AgP compared to ChP Acquier et al., 2017
Case-control study (cross-sectional) 55 ChP patients 55 healthy controls Serum Saliva ↓ TAC in serum and saliva; ↑ MDA increased in serum and saliva vs. controls Ahmadi-Motamayel et al., 2017
Prospective study 33 PW with ChP 18 PW with gingivitis 21 PW controls 27 non-PW ChP 25 non-PW controls Pregnancy Serum GCF ↓ TAC, SOD in PW vs. non-PW and in ChP group compared to controls; ↑ TAC, SOD in third trimester in PW with ChP vs. PW with ChP in the first trimester Akalin et al., 2009
Comparative study (cross-sectional) 20 T2DM patients with periodontitis 20 T2DM patients PH 20 SH patients with periodontitis T2DM Plasma ↓ plasma small molecule antioxidant capacity ↑ PC in T2DM with periodontitis vs. PH T2DM patients Allen et al., 2011
Cross-sectional study 33 ChP patients 16 patients with gingivitis 37 healthy controls Saliva ↑ TAC, 8-OHdG, MDA and activity of SOD, GPx in ChP vs. controls Almerich-Silla et al., 2015
Observational study (cross-sectional) 19 non-T2DM patients 24 T2DM patients with good metabolic control 27 T2DM patients with poor metabolic control T2DM T2DM Saliva ↓ GPx, glutathione reductase in poor metabolic control T2DM patients; ↑ GSSG/GSH ratio in poor metabolic control T2DM Arana et al., 2017
Case-control study 15 PH + normal weight 15 gingivitis + normal weight 15 ChP + normal weight 15 PH + obese 18 gingivitis + obese 15 ChP + obese Obesity GCF ↑ MDA, PC and ↓ TAC in ChP + obese group Atabay et al., 2017
Cross-sectional study 32 ChP post-menopausal women 31 ChP pre-menopausal 25 PH post-menopausal women 26 PH pre-menopausal women Menopause Serum GCF ↓ TAC, SOD activity in post-menopausal women with ChP in serum and GCF Baltacioglu et al., 2006
Cross-sectional study 35 AgP patients 33 ChP patients 30 PH controls Serum Saliva ↑ MDA, TOS, OSI in periodontitis groups; ↓ TAC in periodontitis groups vs. controls (except serum MDA) Baltacioglu et al., 2014
Cross-sectional study 23 ChP patients 19 PH controls Saliva ↑ TBARS in ChP (men) ↓ TAC in ChP (women); trend toward ↓ DNA integrity in ChP Banasová et al., 2015
Cross-sectional study 9 patients with periodontitis 9 healthy controls Gingival tissue ↑ activities of MPO, GPx, glutathione S-transferase in periodontitis group; ↑ TBARS and GSSG levels increased vs. controls Borges et al., 2007
Cross-sectional study 20 ChP patients 20 healthy controls Serum Saliva GCF ↓ TAC in GCF and plasma in ChP vs. control; ↓ TAC in saliva in ChP vs. control Brock et al., 2004
Cross-sectional study 32 ChP patients 32 PH control Saliva Gingival tissue ↑ 8-OHdG and mtDNA deletions in ChP group vs. control Canakci et al., 2009
Cross-sectional study 30 ChP patients 30 PH control Blood Gingival tissue ↑ mtDNA deletion in ChP group vs. control Canakci et al., 2006
Cross-sectional study 10 preeclampsia ChP 10 preeclampsia PH 10 normotensive ChP 10 normotensive PH Preeclampsia Serum Saliva GCF ↓ TAC in ChP women with preeclampsia (GCF, serum, saliva); ↓ SOD and GPx activities decreased in ChP women with preeclampsia (GCF and serum); ↑ MDA in ChP preeclamptic women (GCF, serum) Canakci et al., 2007
Cross-sectional study 31 pre-menopausal 31 peri-menopausal 31 post-menopausal ChP women Menopause GCF Gingival tissue ↑ 8-OHdG in ChP post-menopausal women in GCF and gingival tissue Chandra et al., 2017
Cross-sectional study 10 ChP patients 10 PH control Plasma GCF ↓ TAC in GCF and plasma in ChP vs. control; ↓ GSH and GSSG in GCF in ChP Chapple et al., 2002
Observational correlational study 11,480 participants; 1,567 with mild periodontitis 609 with severe periodontitis Serum ↓ vitamin C, bilirubin and TAC (calculated) in mild or severe periodontitis Chapple et al., 2007b
Cross-sectional study 17 periodontitis patients 20 healthy controls Saliva ↓ TAC in periodontitis Diab-Ladki et al., 2003
Cross-sectional study 12 SH and PH controls 15 SH and ChP patients 8 well-controlled T2DM patients with ChP 14 poor-controlled T2DM patients with ChP T2DM Gingival tissue Peroxiredoxin 1 and GPX1 overexpressed in ChP; Peroxiredoxin 2 and SOD2 up-regulated especially in poor—controlled T2DM with ChP Duarte et al., 2012
Cross-sectional study 20 ChP patients with RA 20 PH patients with RA 20 ChP patients SH 20 SH and PH controls Rheumatoid arthritis Serum GCF ↑ TOS in GCF of ChP and RA ChP groups (no difference for TOS and OSI in serum) Esen et al., 2012
Cross-sectional study 18 ChP patients with hyperlipidemia 18 PH patients with hyperlipidemia 19 ChP patients SH 19 PH SH controls Hyperlipidemia Serum ↑ MDA and 8-OHdG in patients with ChP and hyperlipidemia Fentoglu Ö et al., 2015
Cross-sectional study 24 ChP with depression 23 ChP without depression Depression Plasma ↑ nitric oxide metabolites, lipid peroxides, AOPP and TRAP in ChP with depression Gomes et al., 2017
Cross-sectional study 16 T1DM with periodontitis 25 T2DM with periodontitis 24 SH with periodontitis T1DM T2DM Saliva ↓ GSH and ↓GSSG in the patients with T1DM Gumus et al., 2009
Case-control study 115 P women (6 month postpartum follow-up) 72 non-P women Pregnancy Saliva ↑ 8-OHdG in PW; ↓ GPx decreased in PW; ↓ TBARS postpartum vs. non-pregnant women Gümüş et al., 2015
Prospective study 218 P women 459 P women with mild periodontitis 114 P women with moderate-severe periodontitis Pregnancy Serum ↑ 8-isoprostane in PW with moderate-severe periodontitis Hickman et al., 2011
Prospective study 50 ChP patients 50 healthy controls Serum Saliva ↓ SOD in saliva and serum; ↑ prostaglandin E2, D2, prostaglandin F2α and TXB2, 5- hydroxyeicosatetraenoic acid, F2-isoprostanes; ↑ prostacyclin I2 and 13- hydroxyoctadecadienoic acid, 9- hydroxyoctadecadienoic acid Huang et al., 2014
Cross-sectional Study 55 patients with DS 74 patients with mental retardation 88 healthy controls Down Syndrome Mental retardation Whole blood ↑ oxidative burst activity of blood (monocytes and granulocytes) in DS patients with decreased periodontal health Khocht et al., 2014
Cross-sectional study 26 AgP patients 30 ChP patients 25 healthy controls Gingival blood Peripheral blood ↑ 8-OHdG in gingival blood of ChP and AgP patients; ↓ TAC in gingival blood of ChP patients; ↓ TAC in peripheral blood of both groups Konopka et al., 2007
Cross-sectional study 1,258 old men Old age Serum ↓ beta-cryptoxanthin and beta-carotene with decreased periodontal health of old men Linden et al., 2009
Cross-sectional study 356 periodontitis patients 207 PH controls Plasma ↑ reactive oxygen metabolites and shorter leukocyte telomere length in ChP Masi et al., 2011
Cross-sectional Correlational study 20 ChP patients 20 PH controls Saliva ↑ 8-OHdG, MDA in ChP; ↓ uric acid, GPx activities and TAC in ChP (correlation with bone loss markers) Miricescu et al., 2014
Cross-sectional study 10 T2DM patients PH 8 SH controls T2DM Periodontal tissue ↑ MDA, ↓ GSH in periodontal tissue of T2DM Monea et al., 2014
Cross-sectional study 24 ChP patients with ACS 24 patients PH with ACS 24 ChP patients without ACS 24 controls PH without ACS Acute coronary syndrome Saliva ↑ 8-OHdG, MDA, and PC in patients (correlation with periodontal and cardiovascular markers) Nguyen et al., 2016
Cross-sectional study 25 ChP patients 25 healthy controls Plasma Gingival tissue Erythrocytes ↑ TBARS in ChP; ↑ SOD, CAT, GPx activities in ChP; ↓ vitamins E, C and GSH in ChP Panjamurthy et al., 2005
Cross-sectional study 29 ChP patients 20 healthy controls Saliva ↑ 8-OHdG in ChP (correlation with P. gingivalis) Sawamoto et al., 2005
Cross-sectional cohort study 46 severe periodontitis patients 37 moderate periodontitis patients 46 mild periodontitis and healthy Saliva ↑ PC increased in severe periodontitis; ↓ urate and FRAP in severe periodontitis Sculley and Langley-Evans, 2003
Cross-sectional study 20 ChP patients with RA 20 PH patients with RA 20 ChP patients without RA 20 PH SH controls Rheumatoid arthritis Serum ↑ OSI and prolidase in ChP patients with RA Sezer et al., 2013
Cross-sectional study 20 ChP patients with PS 20 PH patients with PS 20 ChP patients with PsA 20 PH patients with PsA 20 ChP SH patients 20 PH SH controls Psoriasis Psoriatic arthritis Serum ↑ OSI (irrespective of periodontitis) in patients groups; (PS and PsA showed no effect on clinical parameters in ChP patients) Sezer et al., 2016
Cross-sectional study 4,717 participants Diabetes mellitus Hypertension Serum Periodontitis with highest 8-isoprostane quartile associated with ↑ CRP Singer et al., 2015
Cross-sectional correlational study 29 severe periodontitis 77 moderate periodontitis 96 mild periodontitis and healthy Diabetes mellitus Hypertension Serum ↑ ROM in patients with worst periodontal status Tamaki et al., 2014b
Cross-sectional study 25 severe periodontitis 43 moderate periodontitis 92 mild periodontitis and healthy Diabetes mellitus Hypertension Saliva Superoxide and hydroxyl radical scavenging activities associated with periodontitis Tamaki et al., 2015
Cross-sectional study 39 patients with periodontitis Saliva ↓ antioxidant concentrations related to periodontal status Tartaglia et al., 2017
Cross-sectional study 23 smokers with ChP 23 former smokers with ChP 19 non-smokers with ChP 20 PH non-smokers controls Smoking Serum Gingival tissue ↑ MDA in serum and gingival tissue in smoking ChP patients groups; ↓ SOD, CAT and GPx in ChP groups Tonguç et al., 2011
Cross-sectional study 82 children Saliva TBARS in correlation to periodontal status; TAC related to periodontal status and oral hygiene; AOPP related to caries in children Tothova et al., 2013
Cross-sectional study 30 ChP patients with T2DM 30 ChP SH patients 30 PH patients with T2DM 30 PH SH controls T2DM Plasma Saliva Red blood cell lysate ↑ MDA in ChP patients irrespective of T2DM Trivedi et al., 2014
Cross-sectional study 100 ChP patients 50 healthy controls Saliva ↑ 8-OHdG and human neutrophil elastase/alpha1-proteinase inhibitor in ChP Villa-Correa et al., 2015
Cross-sectional study 19 patients with periodontitis 8 healthy controls - GCF ↑ GPx, lactoferrin, myeloperoxidase and IL-1beta in periodontal tissues (correlation with clinical periodontal markers) Wei et al., 2004
Cross-sectional study 31 smokers 90 non-smokers Smoking Saliva ↑ 8-epi-PGF(2alpha) with periodontal status and smoking; Smoking ↑ TXB(2) and PGF(2alphas) and ↓ 6-oxo-PGF(1alpha) Wolfram et al., 2006
Cross-sectional study 58 ChP patients 42 AgP patients 60 healthy controls Saliva Buccal mucosa ↑ micronuclei and nuclear abnormalities, as well as 8-OHdG in both periodontitis groups Zamora-Perez et al., 2015
Cross-sectional study 45 severe periodontitis patients 37 healthy controls Saliva ↓ TAC in periodontitis (TOS no difference) Zhang et al., 2015

ChP, chronic periodontitis; AgP, aggressive periodontitis; SH, systematically healthy; PH, periodontal healthy; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; PS, psoriasis; PsA, psoriatic arthritis; DS, Down syndrome; PW, pregnant woman; RA, Rheumatoid arthritis; ACS, Acute coronary syndrome; GCF, gingival crevicular fluid; TAC, total antioxidant capacity; TBARS, thiobarbituric acid reacting substances; 8-HOdG, 8-hydroxydeoxyguanosine; GPx, glutathione peroxidase; GSH, reduced glutathione; GSSG, oxidized glutathione; PC, protein carbonyls; MDA, malondialdehyde; TOS, total oxidant status; SOD, superoxid dismutase; OSI, oxidative stress index; IL-1beta, interleukin 1beta; ROM, reactive oxygen metabolites; ROS, reactive oxygen species; FRAP, ferric reducing antioxidant power; 4-HNE, 4-hydroxy-2-nonenal; CAT, catalase; CRP, C-reactive protein; AOPP, advanced oxidation protein products; TRAP, total radical-trapping antioxidant potential; MPO, myeloperoxidase; mtDNA, mitochondrial DNA.

Regarding the analyzed markers of oxidative stress, a comparison of the published studies is complicated, if not impossible due to the huge variability of measured markers. Malondialdehyde, 8-hydroxydeoxyguanosine (Konopka et al., 2007; Almerich-Silla et al., 2015), protein carbonyls (Nguyen et al., 2016; Atabay et al., 2017), thiobarbituric acid reacting substances (Borges et al., 2007), nitric oxide, advanced oxidation protein products, lipid peroxidation products (Gomes et al., 2017), 8-isprostanes (Hickman et al., 2011) were all higher in patients with periodontitis. The most commonly measured markers of oxidative stress seem to be malondialdehyde and thiobarbituric acid reacting substances pointing toward oxidative damage of lipids, especially, lipid membranes. Lipid peroxidation was higher in saliva (Tothova et al., 2013), serum (Tonguç et al., 2011), and in the gingival tissue (Panjamurthy et al., 2005) of patients with chronic or aggressive periodontitis. Correlational studies confirmed a positive association of these markers with periodontal status scores (Chapple et al., 2007b; Tamaki et al., 2014b). The less commonly measured markers related to oxidative damage as mitochondrial DNA (Canakci et al., 2006), micronuclei and nuclear abnormalities (Zamora-Perez et al., 2015), as well as a leukocyte telomere length shortening (Masi et al., 2011) were all higher in periodontitis as well.

Oxidative stress was found to be involved in the pathogenesis of many diseases besides periodontitis. Virtually, almost all inflammatory diseases lead to increased oxidative stress. This in turn can trigger more damage to the tissues, not excluding the gingival tissue and, thus, worsening periodontitis. There are several studies describing oxidative stress in systemic diseases with regard to the periodontal status. Nguyen et al. (2016) investigated patients with the acute coronary syndrome with or without chronic periodontitis, patients with periodontitis and healthy controls. Lipid, protein, and DNA oxidation markers were higher in periodontitis than in the control group (Nguyen et al., 2016). Another study with rheumatoid arthritis and chronic periodontitis confirmed higher oxidative stress markers in plasma and lower antioxidant capacity in both groups when compared to healthy control. However, if both comorbidities were present, there was no further enhancement of oxidative stress (Sezer et al., 2013).

Taken together, observational cross-sectional studies confirmed the association of oxidative stress and periodontitis. Higher oxidative stress and lower antioxidant status can be detected in plasma, saliva as well as in the gingival crevicular fluid of patients with various clinical forms of periodontitis. These findings support the use of body fluids, but especially the non-invasive diagnostic fluid saliva, as suitable sample types for diagnostics or monitoring the course of periodontitis. Current data do not support the use of a single oxidative stress marker. It is likely that a set of markers covering both, oxidative damage and antioxidative status, will be needed. The low specificity of oxidative stress markers calls for caution when interpreting the results even if more than one marker is used. The inter-individual and intra-individual variability of the analyzed markers is very high. This prevents their use at the level of individual diagnostics. The overview of the published observational studies shows the enormous heterogeneity of the patient populations as well as the used analytical tools. In a meta-analysis focusing on systematic oxidative stress, it was shown that higher malondialdehyde and nitric oxide as well as lower total antioxidant capacity of plasma/serum characterize patients with periodontitis in comparison to controls (Liu et al., 2014). Based on our overview, taken into account systemic and local oral biomarkers of oxidative stress, it is clear that there is a need for both, the use of a wider palette of markers to analyze oxidative stress and its causes in more detail, and the introduction of new biomarkers with a better sensitivity/specificity profile in specific subgroups of patients. Of special importance is the small sample size in most studies. A collaborative effort with a multi-center recruitment of patients and a standardized consensus protocol in the pre-analytical and analytical phase is highly needed.

Oxidative stress and treatment of periodontitis

The implication of oxidative stress in the pathogenesis of cardiovascular diseases and cancer as the major causes of death in the combination with the widespread availability of dietary antioxidants started a hype that was supported by the hypothesis that aging is caused by oxidative stress (Harman, 1956; Finkel and Holbrook, 2000). However, the hype was quickly over as large clinical studies revealed that antioxidants or at least antioxidant vitamins were not able to prevent any of the diseases of aging (Coulter et al., 2006; Sesso et al., 2008; Myung et al., 2013). Some studies even revealed a slight but increased risk in patients taking antioxidants in preeclampsia (Rumbold et al., 2006) and lung cancer (Alpha-Tocopherol, 1994). One meta-analysis showed that taking antioxidant may even increase the all-cause mortality by 5% (Bjelakovic et al., 2007). This might be related to the terminological and mechanistic confusion about antioxidants, which might have a very indirect effect on the production or effects of reactive oxygen species. In an era of evidence-based medicine the clear conclusion is that antioxidant dietary supplements should undergo clinical evaluation before marketing similarly to other medicinal drugs (Bjelakovic et al., 2012). Recent mechanistic experiments shed light on the details how antioxidants may stimulate tumor growth (Sayin et al., 2014) or increase the risk of metastasis (Piskounova et al., 2015). Antioxidants are highly variable in their mechanism and structure. Diseases and patients vary even more. So, negative results from oncology or cardiology should not be generalized to other diseases including periodontitis. It is very likely that the role of oxidative stress changes during disease progression and, thus, the potential antioxidant treatment affecting not only oxidative damage but also the inflammatory process might have different affects at various stages of the complex pathogenesis. The issue of antioxidants that mostly do not affect free radicals and their action in vivo, but rather interfere with variable cellular signaling pathways has been reviewed in a recently published paper (Azzi, 2017).

Many published studies have analyzed the effects of periodontitis treatment on oxidative stress (Table 2). Studies of special interest are those with covariates or comorbidities that were taken into account. A trial by Guentsch et al. (2008) examined healthy subjects and patients with periodontitis further divided into smokers and non-smokers. While smokers with periodontitis displayed highest malondialdehyde concentration and highest glutathione peroxidase activity along with lowest total antioxidant capacity in saliva. The non-surxgical treatment helped to normalize the values regardless of the smoking status (Guentsch et al., 2008). Several other trials confirmed the consistency of these findings (Chapple et al., 2007a; Abou Sulaiman and Shehadeh, 2010; Hendek et al., 2015). On the other hand, a severe systemic condition such as type 2 diabetes may lead to inefficiency of non-surgical therapies to improve the periodontal status or oxidative stress (Koromantzos et al., 2012). In general, most of the studies clearly show an improvement of oxidative damage after standard treatment of periodontitis. The studies, however, vary greatly regarding sample types, markers measured, and most cohorts were very small and highly variable. Thus, making relevant conclusions or recommendations for the clinical dentistry is difficult, if not impossible. Future research efforts should focus on the lack of uniformity and standardization as well as on the issue of the low informative value of small patient cohorts.

Table 2.

Interventional studies analyzing the effect of periodontitis on oxidative stress.

Treatment Treatment duration Sample size Comorbidity Sample type Results References
NST with vitamin C (2,000 mg/day) 1 month 30 ChP patients 30 healthy controls None Plasma ↓ TAC in ChP patients; NST lead to ↑ TAC in ChP patients; No effect of vitamin C Abou Sulaiman and Shehadeh, 2010
NST with lycopene (8 mg/day) 2 month 20 ChP patients None Serum ↓ MDA and clinical parameters after therapy Ambati et al., 2017
Oral hygiene education with insulin treatment for T1DM 3 months 32 T1DM patients at diagnosis 18 SH children with gingivitis 18 SH and PH children T1DM Serum Saliva GCF ↑ serum, salivary and GCF OSI in T1DM group; ↓ after treatment Aral et al., 2017
NST with lycopene (8 mg/day) 42 ChP patients None Plasma Saliva ↑ IL-1ss, UA and clinical parameters after treatment Arora et al., 2013
NST 14 days 13 ChP patients with FMF 15 ChP SH patients 14 PH patients with FMF 15 PH and SH controls Familial Mediterranean fever Serum GCF ↓ periodontal clinical markers after treatment; ↓ TOS in GCF in FMF patients with ChP; no difference in serum TOS and OSI Bostanci et al., 2014
ST with antioxidant gel (2mg lycopene) 1 week 31 ChP patients None GCF ↓ 8-OHdG after treatment, as well as periodontal clinical markers Chandra et al., 2013
NST 2 months 35 ChP patients 32 healthy controls None Plasma GCF ↓ TAC in GCF in ChP; ↑ after treatment; (plasma TAC no difference at baseline between groups, no change after treatment) Chapple et al., 2007a
NST 4 h 145 ChP patients (14 with therapy) 56 healthy controls None Serum Patients with severe periodontitis ↑ROM ↓ TAC; ↑ ROM after treatment D'Aiuto et al., 2010
NST with fluorescence-controlled Er:YAG laser radiation Laser therapy applied 1 day after NST 30 ChP patients None GCF no difference between with/without laser treatment in TAC and clinical parameters; IL-1beta and ↑ TNF-alpha after NST only; ↓ after NST with laser Dominguez et al., 2010
NST 30 ChP patients 30 healthy controls Smokers/non-smokers Serum Saliva ↑ MDA in smoking ChP patients; ↑ GPx in ChP groups; ↓ TAC in ChP groups; ↓ MDA and GPx after therapy Guentsch et al., 2008
NST 14 days 47 (24 / 23) ChP patients 46 (23/23) healthy controls Smokers/non-smokers Serum Saliva GCF ↑ salivary 8-OHdG and GPx in ChP; ↑ 4-HNE in GCF in ChP smokers; ↓ 8-OHdG after therapy in saliva and GCF Hendek et al., 2015
NST 7 ChP patients 7 healthy controls None Saliva ↑ TAC and ↓ SOD activity in ChP; ↓ TAC, SOD activity immediately after NST Kim et al., 2010
NST 60 moderate to severe periodontitis patients with T2DM T2DM Serum No effect of NST on d-8-iso, MMP-2, MMP-9 and hsCRP Koromantzos et al., 2012
NST; NST with antioxidants (6 mg/day; lycopene, zinc, and selenium) or antioxidants only 3 doses in 2 weeks 30 ChP patients 30 gingivitis patients 10 healthy controls None Saliva ↓ UA in ChP patients; antioxidant treatment ↑ UA Mathur et al., 2013
Tai Chi (5 days a week, 60 min) 6 months 71 sedentary patients with periodontitis Old age Saliva ↑ TAC, SOD after therapy Mendoza-Núñez et al., 2014
NST and surgical treatment Surgical treatment 6 weeks after NST 12 AgP patients None Serum Plasma Periodontitis severity associated with LDL concentrations; No changes in lipid profile after treatment; No difference in GSH and lipid hyperoxide after therapy Nibali et al., 2015
NST or oral hygiene instructions 2 visits within 7 days 42 ChP patients 21 healthy controls None Saliva NST lead to ↑ TAC, ALB, UA, GPx and ↓ SOD; (no effect of oral hygiene instructed therapy was found) Novakovic et al., 2014
NST 25 severe ChP patients 26 healthy controls None Serum Saliva Salivary 8-OHdG ↑ before treatment in ChP group; ↓ after NST ↑ salivary MDA and serum 4-HNE in ChP patients; NST had no effect Onder et al., 2017
NST with Coenzyme Q10; NST with tea tree oil gel or placebo 7 days 15 ChP patients (moderate to severe) None - Both antioxidant treatment procedures effective in ↓ clinical markers of ChP (PI, GI, PPD and CAL) Raut and Sethi, 2016
NST and oral hygiene instructions 2–4 months 29 ChP patients 20 healthy controls None Saliva ↑ 8-OHdG in ChP patients before therapy; ↓ 8-OHdG and P. gingivalis after treatment, as well as periodontal clinical markers Sawamoto et al., 2005
NST 8 ChP patients 8 healthy controls None Platelet suspension ↓ periodontal clinical parameters and CRP after therapy, as well as ↑ cGMP and SOD activity Siqueira et al., 2013
Surgical treatment with taurine (500 mg/day) 15 days 10 ChP patients None Plasma Gingival tissue ↓ TBARS, GPx in plasma and gingival tissue; ↑ GSH and ↓ periodontal clinical parameters after therapy Sree and Sethupathy, 2014
NST 78 ChP patients 17 healthy controls None Saliva ↑ 8-OHdG in ChP patients before therapy; ↓ 8-OHdG and periodontal clinical markers after therapy Takane et al., 2002
NST 22 ChP patients 22 healthy controls None Plasma ↑ oxidative index, oxidized LDL and CRP in ChP; ↓ of these parameters after treatment Tamaki et al., 2011
NST 25 ChP patients 25 patients with gingivitis 25 healthy controls None Serum ↓ serum TAC and CAT in both groups of patients; TAC ↑ in ChP patients after therapy (no treatment effect on CAT) Thomas et al., 2014
NST 4 visits within 14 days 30 (15/15) ChP patients 10 healthy controls Smokers/non-smokers GCF ↑ IL-1beta in ChP patients; ↓ IL-1beta after NST irrespective of smoking; (no difference in TAC and TOS before or after treatment between groups) Toker et al., 2012
NST 2 weeks 25 ChP patients with MS 25 ChP SH patients Metabolic syndrome Serum Saliva TOS and OSI showed no difference between groups in serum after therapy; ↑ TAC of MS ChP patients before treatment, but ↓ after therapy in serum; ↓ OSI and ↑ TAC in both groups after treatment in saliva Torumtay et al., 2016
NST Once per week/1 month 22 ChP patients None Saliva ↑ SOD in patients with low dental visits after NST; ↑ TAC in patients with regular dental visits after therapy Yang et al., 2014
NST with dietary intervention 3 visits/6 months 37 ChP patients (19 without intervention; 18 with intervention) None Plasma Saliva ↑ TAC after dietary intervention in plasma; no differences in periodontal clinical parameters after dietary intervention Zare Javid et al., 2014

ChP, chronic periodontitis; SH, systematically healthy; PH, periodontal healthy; NST, non-surgical treatment; ST, surgical treatment; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; FMF, familial Mediterranean fever; MS, metabolic syndrome; GCF, gingival crevicular fluid; TAC, total antioxidant capacity; TBARS, thiobarbituric acid reacting substances; 8-HOdG, 8-hydroxydeoxyguanosine; GPx, glutathione peroxidase; GSH, reduced glutathione; MDA, malondialdehyde; TOS, total oxidant status; SOD, superoxid dismutase; OSI, oxidative stress index; IL-1ss, salivary interleukin 1beta; ROM, reactive oxygen metabolites; 4-HNE, 4-hydroxy-2-nonenal; hsCRP, high-sensitivity C-reactive protein; d-8-iso, d−8-iso prostaglandin F2a; MMP-2, matrix metalloproteinase 2; MMP-9, matrix metalloproteinase 9; LDL, low density lipoprotein; ALB, albumin; UA, uric acid; CAT, catalase; CRP, C-reactive protein; PI, plaque index; GI, gingival bleeding index; PPD, probing pocket depth; CAL, clinical attachment level; TNF-alpha, tumor necrosis factor alpha; cGMP, L-arginine-nitric oxide (NO)-cyclic guanosine monophosphate.

Although the causality of the association between oxidative stress and periodontitis is everything but clear, some clinical studies already tested antioxidants in periodontitis. One small, placebo controlled, randomized, and double-blind study showed, that single application of lycopene gel to periodontitis patients improved clinical attachment and decreased oxidative stress in gingival crevicular fluid (Chandra et al., 2013). Nevertheless, a published systematic review on antioxidant treatment of periodontitis revealed that a consistent effect in randomized clinical trials was found only for lipophile antioxidants such as lycopene and vitamin E, but not for hydrophile antioxidants such as vitamin C (Muniz et al., 2015). This might be related to the vulnerability of lipids to oxidative damage, but also to mitochondria as the site of effect of some antioxidants. Interestingly, antioxidants targeting directly mitochondria have been shown to be effective in decreasing inflammatory activity and organ damage in animal model of sepsis (Lowes et al., 2013).

Animal experiments

The high number of observational and interventional studies analyzing the association between oxidative stress and periodontitis indicates that there are many open questions that cannot be answered by more and more clinical studies. Many of the questions need controlled conditions in experiments. The number of animal experiments analyzing the role oxidative stress in periodontitis is small, but it increases. Periodontitis is mostly induced by ligature placement around the first molars of the animals or local injection of periodontal pathogens or their toxins (Genco et al., 1998; Fine, 2009; Oz and Puleo, 2011). In such a rat model, it was shown that periodontitis leads to an increased production of reactive oxygen species and markers of oxidative damage (Ekuni et al., 2010). In addition, oxidative stress induced by periodontitis seems to be associated with the dynamics and severity of the periodontal inflammation (Bosca et al., 2016).

Some animal experiments focus on the distant effects of periodontitis that seem to be mediated by oxidative stress. It was shown that mitochondrial DNA is oxidatively modified in the liver, kidney, heart, and brain of rats with induced periodontitis (Tomofuji et al., 2011). Another similar experiment by the same group has shown that periodontitis might worsen ethanol-induced liver damage (Tomofuji et al., 2008). The oxidative damage to the heart, but also endothelial dysfunction and resulting atherosclerosis induced by experimental periodontitis can be prevented by antioxidant treatment (Ekuni et al., 2009; Ozdem et al., 2017; Saito et al., 2017). A majority of the published experiments focus on the use of antioxidants such as vitamin C (Tomofuji et al., 2009b), N-acetylcysteine (Toker et al., 2009), or resveratrol (Tamaki et al., 2014a), but also drugs with an antioxidant activity beyond their main mechanism of action (de Araujo Junior et al., 2013; Culic et al., 2014; Oktay et al., 2015). Dietary interventions have also been investigated—high cholesterol diet seems to worsen periodontitis (Tomofuji et al., 2006). On contrary, the phytoestrogen genistein and cacao-enriched diet was shown to be protective against periodontal damage and oxidative stress induced by periodontitis in mice and rats, respectively (Tomofuji et al., 2009a; Bhattarai et al., 2017). The most promising candidate drug at least according to animal experiments is melatonin. This amphiphile molecule has an optimal distribution in the tissues and can, thus, reach the periodontal tissues even after systemic administration (Köse et al., 2017). Of special clinical relevance is the induction of periodontitis in diabetic rats since periodontitis is a common complication of diabetes in humans. Melatonin was able to prevent alveolar bone loss also in this experimental model (Kose et al., 2016). However, regarding the mechanism of action, it is not clear whether the protective effect of melatonin is due to its direct antioxidant characteristics or due to its immunomodulatory effects that might reduce oxidative stress indirectly (Kara et al., 2013). This uncertainty is not specific for melatonin. Any antioxidant might affect the immune response and, thus, have anti-inflammatory properties. Beyond established systemic antioxidants novel approaches with a local periodontal application are tested (Saita et al., 2016). Experimental tools such as genetically engineered mice that produce luciferase under the regulation of transcription factors related to oxidative stress and antioxidant response have been developed and might greatly improve our understanding of the role of oxidative stress in periodontitis (Kataoka et al., 2016). The new treatment options together with new and improved models could be very helpful in the fight against this widespread and serious disease.

Conclusion

The role of oxidative stress in periodontitis is not clear despite decades of research. Numerous studies have been published showing the potential of oxidative stress markers for screening, diagnosis or monitoring of the disease, but none is in routine clinical use. Similarly, animal experiments, as well as most of the interventional studies in patients, indicate that antioxidant treatment should be effective in the therapy of periodontitis, but no such treatment has been approved. The lack of translation could be either due to the lack of strong evidence for the clinical usefulness or due to obstacles in the application of the results including the low or absent commercial interest from major stakeholders. From a research perspective, an important issue is the lack of specificity—both, in diagnostics and treatment. Not even the source of free radical production is clear. While some studies point toward neutrophils (Katsuragi et al., 2003), others show that bacteria actively producing reactive oxygen species might contribute to oxidative stress in periodontitis (Huycke et al., 2002; Vlkova and Celec, 2009). It is of crucial importance that the number of conducted animal experiments in this field is increasing, especially of those focusing on the dynamics of oxidative stress during disease progression. The antioxidant treatment might be effective only in a subset of patients during a specific stage of periodontitis. The shift from pure observations to interventions and animal experiments that can be followed in the published literature in the recent years is highly positive and should bring this field of research closer to true clinical applications despite chronic lack of financial support and human resources.

Author contributions

L'T has analyzed the data from the literature, prepared tables, and drafted the manuscript; PC has designed the review, conducted the literature search, and drafted the manuscript.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Footnotes

Funding. The authors are supported from the European Operational Program funded by the ERDF (project ITMS: 26240120027).

References

  1. Abou Sulaiman A. E., Shehadeh R. M. (2010). Assessment of total antioxidant capacity and the use of vitamin C in the treatment of non-smokers with chronic periodontitis. J. Periodontol. 81, 1547–1554. 10.1902/jop.2010.100173 [DOI] [PubMed] [Google Scholar]
  2. Acquier A. B., De Couto Pita A. K., Busch L., Sánchez G. A. (2017). Parameters of oxidative stress in saliva from patients with aggressive and chronic periodontitis. Redox Rep. 22, 119–126. 10.1080/13510002.2016.1198104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ahmadi-Motamayel F., Goodarzi M. T., Jamshidi Z., Kebriaei R. (2017). Evaluation of salivary and serum antioxidant and oxidative stress statuses in patients with chronic periodontitis: a case-control study. Front. Physiol. 8:189. 10.3389/fphys.2017.00189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Akalin F. A., Baltacioglu E., Alver A., Karabulut E. (2009). Total antioxidant capacity and superoxide dismutase activity levels in serum and gingival crevicular fluid in pregnant women with chronic periodontitis. J. Periodontol. 80, 457–467. 10.1902/jop.2009.080218 [DOI] [PubMed] [Google Scholar]
  5. Allen E. M., Matthews J. B., O' Halloran D. J., Griffiths H. R., Chapple I. L. (2011). Oxidative and inflammatory status in Type 2 diabetes patients with periodontitis. J. Clin. Periodontol. 38, 894–901. 10.1111/j.1600-051X.2011.01764.x [DOI] [PubMed] [Google Scholar]
  6. Almerich-Silla J. M., Montiel-Company J. M., Pastor S., Serrano F., Puig-Silla M., Dasí F. (2015). Oxidative stress parameters in saliva and its association with periodontal disease and types of bacteria. Dis. Markers 2015:653537. 10.1155/2015/653537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. (1994). The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N. Engl. J. Med. 330, 1029–1035. 10.1056/NEJM199404143301501 [DOI] [PubMed] [Google Scholar]
  8. Ambati M., Rani K. R., Reddy P. V., Suryaprasanna J., Dasari R., Gireddy H. (2017). Evaluation of oxidative stress in chronic periodontitis patients following systemic antioxidant supplementation: a clinical and biochemical study. J. Nat. Sci. Biol. Med. 8, 99–103. 10.4103/0976-9668.198366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Aral C. A., Nalbantoglu Ö O., Nur B. G., Altunsoy M., Aral K. (2017). Metabolic control and periodontal treatment decreases elevated oxidative stress in the early phases of type 1 diabetes onset. Arch. Oral Biol. 82, 115–120. 10.1016/j.archoralbio.2017.06.009 [DOI] [PubMed] [Google Scholar]
  10. Arana C., Moreno-Fernández A. M., Gómez-Moreno G., Morales-Portillo C., Serrano-Olmedo I., de la Cuesta Mayor M. C., et al. (2017). Increased salivary oxidative stress parameters in patients with type 2 diabetes: relation with periodontal disease. Endocrinol. Diabetes Nutr. 64, 258–264. 10.1016/j.endinu.2017.03.005 [DOI] [PubMed] [Google Scholar]
  11. Arora N., Avula H., Avula J. K. (2013). The adjunctive use of systemic antioxidant therapy (lycopene) in nonsurgical treatment of chronic periodontitis: a short-term evaluation. Quintessence Int. 44, 395–405. 10.3290/j.qi.a29188 [DOI] [PubMed] [Google Scholar]
  12. Atabay V. E., Lutfioglu M., Avci B., Sakallioglu E. E., Aydogdu A. (2017). Obesity and oxidative stress in patients with different periodontal status: a case-control study. J. Periodont. Res. 52, 51–60. 10.1111/jre.12368 [DOI] [PubMed] [Google Scholar]
  13. Azzi A. (2017). Antioxidants: wonder drugs or quackery? Biofactors. [Epub ahead of print]. 10.1002/biof.1388 [DOI] [PubMed] [Google Scholar]
  14. Baltacioglu E., Akalin F. A., Alver A., Balaban F., Unsal M., Karabulut E. (2006). Total antioxidant capacity and superoxide dismutase activity levels in serum and gingival crevicular fluid in post-menopausal women with chronic periodontitis. J. Clin. Periodontol. 33, 385–392. 10.1111/j.1600-051X.2006.00923.x [DOI] [PubMed] [Google Scholar]
  15. Baltacioglu E., Yuva P., Aydin G., Alver A., Kahraman C., Karabulut E., et al. (2014). Lipid peroxidation levels and total oxidant/antioxidant status in serum and saliva from patients with chronic and aggressive periodontitis. oxidative stress index: a new biomarker for periodontal disease? J. Periodontol. 85, 1432–1441. 10.1902/jop.2014.130654 [DOI] [PubMed] [Google Scholar]
  16. Banasová L., Kamodyová N., Janšáková K., Tóthová L., Stanko P., Turna J., et al. (2015). Salivary DNA and markers of oxidative stress in patients with chronic periodontitis. Clin. Oral Investig. 19, 201–207. 10.1007/s00784-014-1236-z [DOI] [PubMed] [Google Scholar]
  17. Bhattarai G., Poudel S. B., Kook S. H., Lee J. C. (2017). Anti-inflammatory, anti-osteoclastic, and antioxidant activities of genistein protect against alveolar bone loss and periodontal tissue degradation in a mouse model of periodontitis. J. Biomed. Mater. Res. A 105, 2510–2521. 10.1002/jbm.a.36109 [DOI] [PubMed] [Google Scholar]
  18. Biasi D., Bambara L. M., Carletto A., Caramaschi P., Andrioli G., Urbani G., et al. (1999). Neutrophil migration, oxidative metabolism and adhesion in early onset periodontitis. J. Clin. Periodontol. 26, 563–568. 10.1034/j.1600-051X.1999.260901.x [DOI] [PubMed] [Google Scholar]
  19. Bjelakovic G., Nikolova D., Gluud L. L., Simonetti R. G., Gluud C. (2007). Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA 297, 842–857. 10.1001/jama.297.8.842 [DOI] [PubMed] [Google Scholar]
  20. Bjelakovic G., Nikolova D., Gluud L. L., Simonetti R. G., Gluud C. (2012). Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst. Rev. Cd007176 10.1002/14651858.CD007176.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Borges I., Jr., Moreira E. A., Filho D. W., de Oliveira T. B., da Silva M. B., Fröde T. S. (2007). Proinflammatory and oxidative stress markers in patients with periodontal disease. Mediators Inflamm. 2007:45794. 10.1155/2007/45794 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Boşca A. B., Miclăuş V., Ilea A., Câmpian R. S., Rus V., Ruxanda F., et al. (2016). Role of nitro-oxidative stress in the pathogenesis of experimental rat periodontitis. Clujul. Med. 89, 150–159. 10.15386/cjmed-529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Bostanci V., Toker H., Senel S., Ozdemir H., Aydin H. (2014). Effect of chronic periodontitis on serum and gingival crevicular fluid oxidant and antioxidant status in patients with familial mediterranean fever before and after periodontal treatment. J. Periodontol. 85, 706–712. 10.1902/jop.2013.130230 [DOI] [PubMed] [Google Scholar]
  24. Brock G. R., Butterworth C. J., Matthews J. B., Chapple I. L. (2004). Local and systemic total antioxidant capacity in periodontitis and health. J. Clin. Periodontol. 31, 515–521. 10.1111/j.1600-051X.2004.00509.x [DOI] [PubMed] [Google Scholar]
  25. Canakçi C. F., Canakçi V., Tatar A., Eltas A., Sezer U., Ciçek Y., et al. (2009). Increased salivary level of 8-hydroxydeoxyguanosine is a marker of premature oxidative mitochondrial DNA damage in gingival tissue of patients with periodontitis. Arch. Immunol. Ther. Exp. 57, 205–211. 10.1007/s00005-009-0026-9 [DOI] [PubMed] [Google Scholar]
  26. Canakçi C. F., Tatar A., Canakçi V., Cicek Y., Oztas S., Orbak R. (2006). New evidence of premature oxidative DNA damage: mitochondrial DNA deletion in gingival tissue of patients with periodontitis. J. Periodontol. 77, 1894–1900. 10.1902/jop.2006.060108 [DOI] [PubMed] [Google Scholar]
  27. Canakci V., Yildirim A., Canakci C. F., Eltas A., Cicek Y., Canakci H. (2007). Total antioxidant capacity and antioxidant enzymes in serum, saliva, and gingival crevicular fluid of preeclamptic women with and without periodontal disease. J. Periodontol. 78, 1602–1611. 10.1902/jop.2007.060469 [DOI] [PubMed] [Google Scholar]
  28. Chandra R. V., Sailaja S., Reddy A. A. (2017). Estimation of tissue and crevicular fluid oxidative stress marker in premenopausal, perimenopausal and postmenopausal women with chronic periodontitis. Gerodontology 34, 382–389. 10.1111/ger.12279 [DOI] [PubMed] [Google Scholar]
  29. Chandra R. V., Srinivas G., Reddy A. A., Reddy B. H., Reddy C., Nagarajan S., et al. (2013). Locally delivered antioxidant gel as an adjunct to nonsurgical therapy improves measures of oxidative stress and periodontal disease. J. Periodontal Implant Sci. 43, 121–129. 10.5051/jpis.2013.43.3.121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chapple I. L. (1997). Reactive oxygen species and antioxidants in inflammatory diseases. J. Clin. Periodontol. 24, 287–296. 10.1111/j.1600-051X.1997.tb00760.x [DOI] [PubMed] [Google Scholar]
  31. Chapple I. L., Brock G. R., Milward M. R., Ling N., Matthews J. B. (2007a). Compromised GCF total antioxidant capacity in periodontitis: cause or effect? J. Clin. Periodontol. 34, 103–110. 10.1111/j.1600-051X.2006.01029.x [DOI] [PubMed] [Google Scholar]
  32. Chapple I. L., Brock G., Eftimiadi C., Matthews J. B. (2002). Glutathione in gingival crevicular fluid and its relation to local antioxidant capacity in periodontal health and disease. Mol. Pathol. 55, 367–373. 10.1136/mp.55.6.367 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chapple I. L., Milward M. R., Dietrich T. (2007b). The prevalence of inflammatory periodontitis is negatively associated with serum antioxidant concentrations. J. Nutr. 137, 657–664. [DOI] [PubMed] [Google Scholar]
  34. Coulter I. D., Hardy M. L., Morton S. C., Hilton L. G., Tu W., Valentine D., et al. (2006). Antioxidants vitamin C and vitamin e for the prevention and treatment of cancer. J. Gen. Intern. Med. 21, 735–744. 10.1111/j.1525-1497.2006.00483.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Culic C., Parvu A. E., Alb S. F., Alb C., Pop A. (2014). Effect of Cimetidine on nitro-oxidative stress in a rat model of periodontitis. Clujul. Med. 87, 177–181. 10.15386/cjmed-273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. D'Aiuto F., Nibali L., Parkar M., Patel K., Suvan J., Donos N. (2010). Oxidative stress, systemic inflammation, and severe periodontitis. J. Dent. Res. 89, 1241–1246. 10.1177/0022034510375830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. de Araújo Júnior R. F., Souza T. O., de Moura L. M., Torres K. P., de Souza L. B., Alves Mdo S., et al. (2013). Atorvastatin decreases bone loss, inflammation and oxidative stress in experimental periodontitis. PLoS ONE 8:e75322. 10.1371/journal.pone.0075322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Diab-Ladki R., Pellat B., Chahine R. (2003). Decrease in the total antioxidant activity of saliva in patients with periodontal diseases. Clin. Oral Investig. 7, 103–107. 10.1007/s00784-003-0208-5 [DOI] [PubMed] [Google Scholar]
  39. Domínguez A., Gómez C., García-Kass A. I., García-Nuñez J. A. (2010). IL-1beta, TNF-alpha, total antioxidative status and microbiological findings in chronic periodontitis treated with fluorescence-controlled Er:YAG laser radiation. Lasers Surg. Med. 42, 24–31. 10.1002/lsm.20873 [DOI] [PubMed] [Google Scholar]
  40. Duarte P. M., Napimoga M. H., Fagnani E. C., Santos V. R., Bastos M. F., Ribeiro F. V., et al. (2012). The expression of antioxidant enzymes in the gingivae of type 2 diabetics with chronic periodontitis. Arch. Oral Biol. 57, 161–168. 10.1016/j.archoralbio.2011.08.007 [DOI] [PubMed] [Google Scholar]
  41. Ekuni D., Endo Y., Irie K., Azuma T., Tamaki N., Tomofuji T., et al. (2010). Imbalance of oxidative/anti-oxidative status induced by periodontitis is involved in apoptosis of rat submandibular glands. Arch. Oral Biol. 55, 170–176. 10.1016/j.archoralbio.2009.11.013 [DOI] [PubMed] [Google Scholar]
  42. Ekuni D., Tomofuji T., Sanbe T., Irie K., Azuma T., Maruyama T., et al. (2009). Vitamin C intake attenuates the degree of experimental atherosclerosis induced by periodontitis in the rat by decreasing oxidative stress. Arch. Oral Biol. 54, 495–502. 10.1016/j.archoralbio.2009.02.006 [DOI] [PubMed] [Google Scholar]
  43. Esen C., Alkan B. A., Kirnap M., Akgül O., Işikoglu S., Erel O. (2012). The effects of chronic periodontitis and rheumatoid arthritis on serum and gingival crevicular fluid total antioxidant/oxidant status and oxidative stress index. J. Periodontol. 83, 773–779. 10.1902/jop.2011.110420 [DOI] [PubMed] [Google Scholar]
  44. Espinosa-Diez C., Miguel V., Mennerich D., Kietzmann T., Sánchez-Pérez P., Cadenas S., et al. (2015). Antioxidant responses and cellular adjustments to oxidative stress. Redox Biol. 6, 183–197. 10.1016/j.redox.2015.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fentoglu Ö O., Kirzioglu F. Y., Bulut M. T., Kumbul Doguç D., Kulaç E., Önder C., et al. (2015). Evaluation of lipid peroxidation and oxidative DNA damage in patients with periodontitis and hyperlipidemia. J. Periodontol. 86, 682–688. 10.1902/jop.2015.140561 [DOI] [PubMed] [Google Scholar]
  46. Fine D. H. (2009). Of mice and men: animal models of human periodontal disease. J. Clin. Periodontol. 36, 913–914. 10.1111/j.1600-051X.2009.01456.x [DOI] [PubMed] [Google Scholar]
  47. Finkel T. (2011). Signal transduction by reactive oxygen species. J. Cell Biol. 194, 7–15. 10.1083/jcb.201102095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Finkel T., Holbrook N. J. (2000). Oxidants, oxidative stress and the biology of ageing. Nature 408, 239–247. 10.1038/35041687 [DOI] [PubMed] [Google Scholar]
  49. Frijhoff J., Winyard P. G., Zarkovic N., Davies S. S., Stocker R., Cheng D., et al. (2015). Clinical relevance of biomarkers of oxidative stress. Antioxid. Redox Signal. 23, 1144–1170. 10.1089/ars.2015.6317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Genco C. A., Van Dyke T., Amar S. (1998). Animal models for Porphyromonas gingivalis-mediated periodontal disease. Trends Microbiol. 6, 444–449. 10.1016/S0966-842X(98)01363-8 [DOI] [PubMed] [Google Scholar]
  51. Gomes C., Martinho F. C., Barbosa D. S., Antunes L. S., Póvoa H. C. C., Baltus T. H. L., et al. (2017). Increased root canal endotoxin levels are associated with chronic apical periodontitis, increased oxidative and nitrosative stress, major depression, severity of depression, and a lowered quality of life. Mol. Neurobiol. [Epub ahead of print]. 10.1007/s12035-017-0545-z [DOI] [PubMed] [Google Scholar]
  52. Guentsch A., Preshaw P., Bremer-Streck S., Klinger G., Glockmann E., Sigusch B. (2008). Lipid peroxidation and antioxidant activity in saliva of periodontitis patients: effect of smoking and periodontal treatment. Clin. Oral Investig. 12, 345–352. 10.1007/s00784-008-0202-z [DOI] [PubMed] [Google Scholar]
  53. Gümüş P., Buduneli N., Cetinkalp S., Hawkins S. I., Renaud D., Kinane D. F., et al. (2009). Salivary antioxidants in patients with type 1 or 2 diabetes and inflammatory periodontal disease: a case-control study. J. Periodontol. 80, 1440–1446. 10.1902/jop.2009.090159 [DOI] [PubMed] [Google Scholar]
  54. Gümüş P., Emingil G., Öztürk V. O., Belibasakis G. N., Bostanci N. (2015). Oxidative stress markers in saliva and periodontal disease status: modulation during pregnancy and postpartum. BMC Infect. Dis. 15:261. 10.1186/s12879-015-1003-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Harman D. (1956). Aging: a theory based on free radical and radiation chemistry. J. Gerontol. 11, 298–300. 10.1093/geronj/11.3.298 [DOI] [PubMed] [Google Scholar]
  56. Hendek M. K., Erdemir E. O., Kisa U., Ozcan G. (2015). Effect of initial periodontal therapy on oxidative stress markers in gingival crevicular fluid, saliva, and serum in smokers and non-smokers with chronic periodontitis. J. Periodontol. 86, 273–282. 10.1902/jop.2014.140338 [DOI] [PubMed] [Google Scholar]
  57. Hickman M. A., Boggess K. A., Moss K. L., Beck J. D., Offenbacher S. (2011). Maternal periodontal disease is associated with oxidative stress during pregnancy. Am. J. Perinatol. 28, 247–252. 10.1055/s-0030-1268706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Huang Y., Zhu M., Li Z., Sa R., Chu Q., Zhang Q., et al. (2014). Mass spectrometry-based metabolomic profiling identifies alterations in salivary redox status and fatty acid metabolism in response to inflammation and oxidative stress in periodontal disease. Free Radic. Biol. Med. 70, 223–232. 10.1016/j.freeradbiomed.2014.02.024 [DOI] [PubMed] [Google Scholar]
  59. Huycke M. M., Abrams V., Moore D. R. (2002). Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 23, 529–536. 10.1093/carcin/23.3.529 [DOI] [PubMed] [Google Scholar]
  60. Kara A., Akman S., Ozkanlar S., Tozoglu U., Kalkan Y., Canakci C. F., et al. (2013). Immune modulatory and antioxidant effects of melatonin in experimental periodontitis in rats. Free Radic. Biol. Med. 55, 21–26. 10.1016/j.freeradbiomed.2012.11.002 [DOI] [PubMed] [Google Scholar]
  61. Kataoka K., Ekuni D., Tomofuji T., Irie K., Kunitomo M., Uchida Y., et al. (2016). Visualization of oxidative stress induced by experimental periodontitis in keap1-dependent oxidative stress detector-luciferase mice. Int. J. Mol. Sci. 17:1907. 10.3390/ijms17111907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Katsuragi H., Ohtake M., Kurasawa I., Saito K. (2003). Intracellular production and extracellular release of oxygen radicals by PMNs and oxidative stress on PMNs during phagocytosis of periodontopathic bacteria. Odontology 91, 13–18. 10.1007/s10266-003-0022-1 [DOI] [PubMed] [Google Scholar]
  63. Khocht A., Russell B., Cannon J. G., Turner B., Janal M. (2014). Oxidative burst intensity of peripheral phagocytic cells and periodontitis in down syndrome. J. Periodont. Res. 49, 29–35. 10.1111/jre.12075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kim S. C., Kim O. S., Kim O. J., Kim Y. J., Chung H. J. (2010). Antioxidant profile of whole saliva after scaling and root planing in periodontal disease. J. Periodont. Implant Sci. 40, 164–171. 10.5051/jpis.2010.40.4.164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Kimura S., Yonemura T., Kaya H. (1993). Increased oxidative product formation by peripheral blood polymorphonuclear leukocytes in human periodontal diseases. J. Periodontal Res. 28, 197–203. 10.1111/j.1600-0765.1993.tb01069.x [DOI] [PubMed] [Google Scholar]
  66. Kinane D. F., Preshaw P. M., Loos B. G., Working Group 2 of Seventh European Workshop on Periodontology . (2011). Host-response: understanding the cellular and molecular mechanisms of host-microbial interactions–consensus of the seventh European workshop on periodontology. J. Clin. Periodontol. 38(Suppl. 11), 44–48. 10.1111/j.1600-051X.2010.01682.x [DOI] [PubMed] [Google Scholar]
  67. Kinane D. F., Stathopoulou P. G., Papapanou P. N. (2017). Periodontal diseases. Nat. Rev. Dis. Primers 3:17038 10.1038/nrdp.2017.38 [DOI] [PubMed] [Google Scholar]
  68. Konopka T., Król K., Kopeć W., Gerber H. (2007). Total antioxidant status and 8-hydroxy-2'-deoxyguanosine levels in gingival and peripheral blood of periodontitis patients. Arch. Immunol. Ther. Exp. 55, 417–422. 10.1007/s00005-007-0047-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kopáni M., Celec P., Danisovic L., Michalka P., Biró C. (2006). Oxidative stress and electron spin resonance. Clin. Chim. Acta 364, 61–66. 10.1016/j.cca.2005.05.016 [DOI] [PubMed] [Google Scholar]
  70. Koromantzos P. A., Makrilakis K., Dereka X., Offenbacher S., Katsilambros N., Vrotsos I. A., et al. (2012). Effect of non-surgical periodontal therapy on C-reactive protein, oxidative stress, and matrix metalloproteinase (MMP)-9 and MMP-2 levels in patients with type 2 diabetes: a randomized controlled study. J. Periodontol. 83, 3–10. 10.1902/jop.2011.110148 [DOI] [PubMed] [Google Scholar]
  71. Kose O., Arabaci T., Kara A., Yemenoglu H., Kermen E., Kizildag A., et al. (2016). Effects of melatonin on oxidative stress index and alveolar bone loss in diabetic rats with periodontitis. J. Periodontol. 87, e82–e90. 10.1902/jop.2016.150541 [DOI] [PubMed] [Google Scholar]
  72. Köse O., Arabaci T., Kizildag A., Erdemci B., Özkal Eminoglu D., Gedikli S., et al. (2017). Melatonin prevents radiation-induced oxidative stress and periodontal tissue breakdown in irradiated rats with experimental periodontitis. J. Periodont. Res. 52, 438–446. 10.1111/jre.12409 [DOI] [PubMed] [Google Scholar]
  73. Laine M. L., Crielaard W., Loos B. G. (2012). Genetic susceptibility to periodontitis. Periodontol 2000 58, 37–68. 10.1111/j.1600-0757.2011.00415.x [DOI] [PubMed] [Google Scholar]
  74. Linden G. J., McClean K. M., Woodside J. V., Patterson C. C., Evans A., Young I. S., et al. (2009). Antioxidants and periodontitis in 60-70-year-old men. J. Clin. Periodontol. 36, 843–849. 10.1111/j.1600-051X.2009.01468.x [DOI] [PubMed] [Google Scholar]
  75. Liu Z., Liu Y., Song Y., Zhang X., Wang S., Wang Z. (2014). Systemic oxidative stress biomarkers in chronic periodontitis: a meta-analysis. Dis. Markers 2014:931083. 10.1155/2014/931083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Loesche W. J., Robinson J. P., Flynn M., Hudson J. L., Duque R. E. (1988). Reduced oxidative function in gingival crevicular neutrophils in periodontal disease. Infect. Immun. 56, 156–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Lowes D. A., Webster N. R., Murphy M. P., Galley H. F. (2013). Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis. Br. J. Anaesth. 110, 472–480. 10.1093/bja/aes577 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Masi S., Salpea K. D., Li K., Parkar M., Nibali L., Donos N., et al. (2011). Oxidative stress, chronic inflammation, and telomere length in patients with periodontitis. Free Radic. Biol. Med. 50, 730–735. 10.1016/j.freeradbiomed.2010.12.031 [DOI] [PubMed] [Google Scholar]
  79. Mathur A., Mathur L., Manohar B., Mathur H., Shankarapillai R., Shetty N., et al. (2013). Antioxidant therapy as monotherapy or as an adjunct to treatment of periodontal diseases. J. Indian Soc. Periodontol. 17, 21–24. 10.4103/0972-124X.107469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Mendoza-Núñez V. M., Hernández-Monjaraz B., Santiago-Osorio E., Betancourt-Rule J. M., Ruiz-Ramos M. (2014). Tai Chi exercise increases SOD activity and total antioxidant status in saliva and is linked to an improvement of periodontal disease in the elderly. Oxid. Med. Cell. Longev. 2014:603853. 10.1155/2014/603853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Miricescu D., Totan A., Calenic B., Mocanu B., Didilescu A., Mohora M., et al. (2014). Salivary biomarkers: relationship between oxidative stress and alveolar bone loss in chronic periodontitis. Acta Odontol. Scand. 72, 42–47. 10.3109/00016357.2013.795659 [DOI] [PubMed] [Google Scholar]
  82. Monea A., Mezei T., Popsor S., Monea M. (2014). Oxidative stress: a link between diabetes mellitus and periodontal disease. Int. J. Endocrinol. 2014:917631. 10.1155/2014/917631 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Muniz F. W., Nogueira S. B., Mendes F. L., Rosing C. K., Moreira M. M., de Andrade G. M., et al. (2015). The impact of antioxidant agents complimentary to periodontal therapy on oxidative stress and periodontal outcomes: a systematic review. Arch. Oral Biol. 60, 1203–1214. 10.1016/j.archoralbio.2015.05.007 [DOI] [PubMed] [Google Scholar]
  84. Myung S. K., Ju W., Cho B., Oh S. W., Park S. M., Koo B. K., et al. (2013). Efficacy of vitamin and antioxidant supplements in prevention of cardiovascular disease: systematic review and meta-analysis of randomised controlled trials. BMJ 346:f10. 10.1136/bmj.f10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Nguyen T. T., Ngo L. Q., Promsudthi A., Surarit R. (2016). Salivary oxidative stress biomarkers in chronic periodontitis and acute coronary syndrome. Clin. Oral Investig. 21, 2345–2353. 10.1007/s00784-016-2029-3 [DOI] [PubMed] [Google Scholar]
  86. Nibali L., Rizzo M., Li Volti G., D'Aiuto F., Giglio R. V., Barbagallo I., et al. (2015). Lipid subclasses profiles and oxidative stress in aggressive periodontitis before and after treatment. J. Periodont. Res. 50, 890–896. 10.1111/jre.12283 [DOI] [PubMed] [Google Scholar]
  87. Niki E. (2016). Oxidative stress and antioxidants: distress or eustress? Arch. Biochem. Biophys. 595, 19–24. 10.1016/j.abb.2015.11.017 [DOI] [PubMed] [Google Scholar]
  88. Novakovic N., Todorovic T., Rakic M., Milinkovic I., Dozic I., Jankovic S., et al. (2014). Salivary antioxidants as periodontal biomarkers in evaluation of tissue status and treatment outcome. J. Periodont. Res. 49, 129–136. 10.1111/jre.12088 [DOI] [PubMed] [Google Scholar]
  89. Oktay S., Chukkapalli S. S., Rivera-Kweh M. F., Velsko I. M., Holliday L. S., Kesavalu L. (2015). Periodontitis in rats induces systemic oxidative stress that is controlled by bone-targeted antiresorptives. J. Periodontol. 86, 137–145. 10.1902/jop.2014.140302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Önder C., Kurgan S., Altingöz S. M., Bagiş N., Uyanik M., Serdar M. A., et al. (2017). Impact of non-surgical periodontal therapy on saliva and serum levels of markers of oxidative stress. Clin. Oral Investig. 21, 1961–1969. 10.1007/s00784-016-1984-z [DOI] [PubMed] [Google Scholar]
  91. Oz H. S., Puleo D. A. (2011). Animal models for periodontal disease. J. Biomed. Biotechnol. 2011:754857. 10.1155/2011/754857 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Özdem M., Kirzioglu F. Y., Yilmaz H. R., Vural H., Fentoglu Ö., Uz E., et al. (2017). Antioxidant effects of melatonin in heart tissue after induction of experimental periodontitis in rats. J. Oral Sci. 59, 23–29. 10.2334/josnusd.16-0034 [DOI] [PubMed] [Google Scholar]
  93. Panjamurthy K., Manoharan S., Ramachandran C. R. (2005). Lipid peroxidation and antioxidant status in patients with periodontitis. Cell. Mol. Biol. Lett. 10, 255–264. [PubMed] [Google Scholar]
  94. Piskounova E., Agathocleous M., Murphy M. M., Hu Z., Huddlestun S. E., Zhao Z., et al. (2015). Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191. 10.1038/nature15726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Raut C. P., Sethi K. S. (2016). Comparative evaluation of co-enzyme Q10 and Melaleuca alternifolia as antioxidant gels in treatment of chronic periodontitis: a clinical study. Contemp. Clin. Dent. 7, 377–381. 10.4103/0976-237X.188572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Rumbold A. R., Crowther C. A., Haslam R. R., Dekker G. A., Robinson J. S. (2006). Vitamins C and E and the risks of preeclampsia and perinatal complications. N. Engl. J. Med. 354, 1796–1806. 10.1056/NEJMoa054186 [DOI] [PubMed] [Google Scholar]
  97. Russell E. G., Cotter T. G. (2015). New insight into the role of reactive oxygen species (ROS) in cellular signal-transduction processes. Int. Rev. Cell Mol. Biol. 319, 221–254. 10.1016/bs.ircmb.2015.07.004 [DOI] [PubMed] [Google Scholar]
  98. Saita M., Kaneko J., Sato T., Takahashi S. S., Wada-Takahashi S., Kawamata R., et al. (2016). Novel antioxidative nanotherapeutics in a rat periodontitis model: Reactive oxygen species scavenging by redox injectable gel suppresses alveolar bone resorption. Biomaterials 76, 292–301. 10.1016/j.biomaterials.2015.10.077 [DOI] [PubMed] [Google Scholar]
  99. Saito T., Yamamoto Y., Feng G. G., Kazaoka Y., Fujiwara Y., Kinoshita H. (2017). Lidocaine prevents oxidative stress-induced endothelial dysfunction of the systemic artery in rats with intermittent periodontal inflammation. Anesth. Analg. 124, 2054–2062. 10.1213/ANE.0000000000002102 [DOI] [PubMed] [Google Scholar]
  100. Sawamoto Y., Sugano N., Tanaka H., Ito K. (2005). Detection of periodontopathic bacteria and an oxidative stress marker in saliva from periodontitis patients. Oral Microbiol. Immunol. 20, 216–220. 10.1111/j.1399-302X.2005.00215.x [DOI] [PubMed] [Google Scholar]
  101. Sayin V. I., Ibrahim M. X., Larsson E., Nilsson J. A., Lindahl P., Bergo M. O. (2014). Antioxidants accelerate lung cancer progression in mice. Sci. Transl. Med. 6, 221ra215. 10.1126/scitranslmed.3007653 [DOI] [PubMed] [Google Scholar]
  102. Sculley D. V., Langley-Evans S. C. (2003). Periodontal disease is associated with lower antioxidant capacity in whole saliva and evidence of increased protein oxidation. Clin. Sci. 105, 167–172. 10.1042/CS20030031 [DOI] [PubMed] [Google Scholar]
  103. Sesso H. D., Buring J. E., Christen W. G., Kurth T., Belanger C., MacFadyen J., et al. (2008). Vitamins E and C in the prevention of cardiovascular disease in men: the physicians' health study II randomized controlled trial. JAMA 300, 2123–2133. 10.1001/jama.2008.600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Sezer U., Erciyas K., Ustün K., Pehlivan Y., Senyurt S. Z., Aksoy N., et al. (2013). Effect of chronic periodontitis on oxidative status in patients with rheumatoid arthritis. J. Periodontol. 84, 785–792. 10.1902/jop.2012.120179 [DOI] [PubMed] [Google Scholar]
  105. Sezer U., Senyurt S. Z., Gündogar H., Erciyas K., Ustun K., Kimyon G., et al. (2016). Effect of chronic periodontitis on oxidative status in patients with psoriasis and psoriatic arthritis. J. Periodontol. 87, 557–565. 10.1902/jop.2015.150337 [DOI] [PubMed] [Google Scholar]
  106. Shapira L., Borinski R., Sela M. N., Soskolne A. (1991). Superoxide formation and chemiluminescence of peripheral polymorphonuclear leukocytes in rapidly progressive periodontitis patients. J. Clin. Periodontol. 18, 44–48. 10.1111/j.1600-051X.1991.tb01118.x [DOI] [PubMed] [Google Scholar]
  107. Sies H., Berndt C., Jones D. P. (2017). Oxidative stress. Annu. Rev. Biochem. 86, 715–748. 10.1146/annurev-biochem-061516-045037 [DOI] [PubMed] [Google Scholar]
  108. Singer R. E., Moss K., Kim S. J., Beck J. D., Offenbacher S. (2015). Oxidative stress and IgG antibody modify periodontitis-CRP association. J. Dent. Res. 94, 1698–1705. 10.1177/0022034515602693 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Siqueira M. A., Fischer R. G., Pereira N. R., Martins M. A., Moss M. B., Mendes-Ribeiro A. C., et al. (2013). Effects of non-surgical periodontal treatment on the L-arginine-nitric oxide pathway and oxidative status in platelets. Exp. Biol. Med. 238, 713–722. 10.1177/1535370213480690 [DOI] [PubMed] [Google Scholar]
  110. Sree S. L., Sethupathy S. (2014). Evaluation of the efficacy of taurine as an antioxidant in the management of patients with chronic periodontitis. Dent. Res. J. 11, 228–233. [PMC free article] [PubMed] [Google Scholar]
  111. Takane M., Sugano N., Iwasaki H., Iwano Y., Shimizu N., Ito K. (2002). New biomarker evidence of oxidative DNA damage in whole saliva from clinically healthy and periodontally diseased individuals. J. Periodontol. 73, 551–554. 10.1902/jop.2002.73.5.551 [DOI] [PubMed] [Google Scholar]
  112. Tamaki N., Cristina Orihuela-Campos R., Inagaki Y., Fukui M., Nagata T., Ito H. O. (2014a). Resveratrol improves oxidative stress and prevents the progression of periodontitis via the activation of the Sirt1/AMPK and the Nrf2/antioxidant defense pathways in a rat periodontitis model. Free Radic. Biol. Med. 75, 222–229. 10.1016/j.freeradbiomed.2014.07.034 [DOI] [PubMed] [Google Scholar]
  113. Tamaki N., Hayashida H., Fukui M., Kitamura M., Kawasaki K., Nakazato M., et al. (2014b). Oxidative stress and antibody levels to periodontal bacteria in adults: the Nagasaki Islands study. Oral Dis. 20, e49–e56. 10.1111/odi.12127 [DOI] [PubMed] [Google Scholar]
  114. Tamaki N., Tomofuji T., Ekuni D., Yamanaka R., Morita M. (2011). Periodontal treatment decreases plasma oxidized LDL level and oxidative stress. Clin. Oral Investig. 15, 953–958. 10.1007/s00784-010-0458-y [DOI] [PubMed] [Google Scholar]
  115. Tamaki N., Yoshino F., Fukui M., Hayashida H., Yoshida A., Kitamura M., et al. (2015). Relationship among salivary antioxidant activity, cytokines, and periodontitis: the Nagasaki Island study. J. Clin. Periodontol. 42, 711–718. 10.1111/jcpe.12438 [DOI] [PubMed] [Google Scholar]
  116. Tartaglia G. M., Gagliano N., Zarbin L., Tolomeo G., Sforza C. (2017). Antioxidant capacity of human saliva and periodontal screening assessment in healthy adults. Arch. Oral Biol. 78, 34–38. 10.1016/j.archoralbio.2017.02.003 [DOI] [PubMed] [Google Scholar]
  117. Thomas B., Madani S. M., Prasad B. R., Kumari S. (2014). Comparative evaluation of serum antioxidant levels in periodontally diseased patients: an interventional study. Contemp. Clin. Dent. 5, 340–344. 10.4103/0976-237X.137938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Toker H., Akpinar A., Aydin H., Poyraz O. (2012). Influence of smoking on interleukin-1beta level, oxidant status and antioxidant status in gingival crevicular fluid from chronic periodontitis patients before and after periodontal treatment. J. Periodont. Res. 47, 572–577. 10.1111/j.1600-0765.2012.01468.x [DOI] [PubMed] [Google Scholar]
  119. Toker H., Ozdemir H., Eren K., Ozer H., Sahin G. (2009). N-acetylcysteine, a thiol antioxidant, decreases alveolar bone loss in experimental periodontitis in rats. J. Periodontol. 80, 672–678. 10.1902/jop.2009.080509 [DOI] [PubMed] [Google Scholar]
  120. Tomofuji T., Azuma T., Kusano H., Sanbe T., Ekuni D., Tamaki N., et al. (2006). Oxidative damage of periodontal tissue in the rat periodontitis model: effects of a high-cholesterol diet. FEBS Lett. 580, 3601–3604. 10.1016/j.febslet.2006.05.041 [DOI] [PubMed] [Google Scholar]
  121. Tomofuji T., Ekuni D., Irie K., Azuma T., Endo Y., Tamaki N., et al. (2009a). Preventive effects of a cocoa-enriched diet on gingival oxidative stress in experimental periodontitis. J. Periodontol. 80, 1799–1808. 10.1902/jop.2009.090270 [DOI] [PubMed] [Google Scholar]
  122. Tomofuji T., Ekuni D., Irie K., Azuma T., Tamaki N., Maruyama T., et al. (2011). Relationships between periodontal inflammation, lipid peroxide and oxidative damage of multiple organs in rats. Biomed. Res. 32, 343–349. 10.2220/biomedres.32.343 [DOI] [PubMed] [Google Scholar]
  123. Tomofuji T., Ekuni D., Sanbe T., Irie K., Azuma T., Maruyama T., et al. (2009b). Effects of vitamin C intake on gingival oxidative stress in rat periodontitis. Free Radic. Biol. Med. 46, 163–168. 10.1016/j.freeradbiomed.2008.09.040 [DOI] [PubMed] [Google Scholar]
  124. Tomofuji T., Sanbe T., Ekuni D., Azuma T., Irie K., Maruyama T., et al. (2008). Oxidative damage of rat liver induced by ligature-induced periodontitis and chronic ethanol consumption. Arch. Oral Biol. 53, 1113–1118. 10.1016/j.archoralbio.2008.05.015 [DOI] [PubMed] [Google Scholar]
  125. Tonguç M. O., Öztürk O., Sutcu R., Ceyhan B. M., Kilinc G., Sonmez Y., et al. (2011). The impact of smoking status on antioxidant enzyme activity and malondialdehyde levels in chronic periodontitis. J. Periodontol. 82, 1320–1328. 10.1902/jop.2011.100618 [DOI] [PubMed] [Google Scholar]
  126. Torumtay G., Kirzioglu F. Y., Ozturk Tonguc M., Kale B., Calapoglu M., Orhan H. (2016). Effects of periodontal treatment on inflammation and oxidative stress markers in patients with metabolic syndrome. J. Periodont. Res. 51, 489–498. 10.1111/jre.12328 [DOI] [PubMed] [Google Scholar]
  127. Tothova L., Celecova V., Celec P. (2013). Salivary markers of oxidative stress and their relation to periodontal and dental status in children. Dis. Markers 34, 9–15. 10.1155/2013/591765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Trivedi S., Lal N., Mahdi A. A., Mittal M., Singh B., Pandey S. (2014). Evaluation of antioxidant enzymes activity and malondialdehyde levels in patients with chronic periodontitis and diabetes mellitus. J. Periodontol. 85, 713–720. 10.1902/jop.2013.130066 [DOI] [PubMed] [Google Scholar]
  129. Villa-Correa Y. A., Isaza-Guzman D. M., Tobon-Arroyave S. I. (2015). Prognostic value of 8-Hydroxy-2'-Deoxyguanosine and human neutrophil elastase/alpha1-proteinase inhibitor complex as salivary biomarkers of oxidative stress in chronic periodontitis. J. Periodontol. 86, 1260–1267. 10.1902/jop.2015.150293 [DOI] [PubMed] [Google Scholar]
  130. Vlkova B., Celec P. (2009). Does Enterococcus faecalis contribute to salivary thiobarbituric acid-reacting substances? In Vivo 23, 343–345. [PubMed] [Google Scholar]
  131. Watson J. D. (2014). Type 2 diabetes as a redox disease. Lancet 383, 841–843. 10.1016/S0140-6736(13)62365-X [DOI] [PubMed] [Google Scholar]
  132. Wei P. F., Ho K. Y., Ho Y. P., Wu Y. M., Yang Y. H., Tsai C. C. (2004). The investigation of glutathione peroxidase, lactoferrin, myeloperoxidase and interleukin-1beta in gingival crevicular fluid: implications for oxidative stress in human periodontal diseases. J. Periodont. Res. 39, 287–293. 10.1111/j.1600-0765.2004.00744.x [DOI] [PubMed] [Google Scholar]
  133. Weidinger A., Kozlov A. V. (2015). Biological activities of reactive oxygen and nitrogen species: oxidative stress vs. signal transduction. Biomolecules 5, 472–484. 10.3390/biom5020472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Wolfram R. M., Budinsky A. C., Eder A., Presenhuber C., Nell A., Sperr W., et al. (2006). Salivary isoprostanes indicate increased oxidation injury in periodontitis with additional tobacco abuse. Biofactors 28, 21–31. 10.1002/biof.5520280103 [DOI] [PubMed] [Google Scholar]
  135. Yang P. S., Huang W. C., Chen S. Y., Chen C. H., Lee C. Y., Lin C. T., et al. (2014). Scaling-stimulated salivary antioxidant changes and oral-health behavior in an evaluation of periodontal treatment outcomes. Sci. World J. 2014:814671. 10.1155/2014/814671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Zamora-Perez A. L., Ortiz-Garcia Y. M., Lazalde-Ramos B. P., Guerrero-Velazquez C., Gomez-Meda B. C., Ramirez-Aguilar M. A., et al. (2015). Increased micronuclei and nuclear abnormalities in buccal mucosa and oxidative damage in saliva from patients with chronic and aggressive periodontal diseases. J. Periodont. Res. 50, 28–36. 10.1111/jre.12175 [DOI] [PubMed] [Google Scholar]
  137. Zare Javid A., Seal C. J., Heasman P., Moynihan P. J. (2014). Impact of a customised dietary intervention on antioxidant status, dietary intakes and periodontal indices in patients with adult periodontitis. J. Hum. Nutr. Diet. 27, 523–532. 10.1111/jhn.12184 [DOI] [PubMed] [Google Scholar]
  138. Zhang T., Andrukhov O., Haririan H., Muller-Kern M., Liu S., Liu Z., et al. (2015). Total antioxidant capacity and total oxidant status in saliva of periodontitis patients in relation to bacterial load. Front. Cell. Infect. Microbiol. 5:97. 10.3389/fcimb.2015.00097 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Frontiers in Physiology are provided here courtesy of Frontiers Media SA

RESOURCES