Next Article in Journal
Efficacy and Tolerability of 6-Month Treatment with Tamsulosin Plus the Hexanic Extract of Serenoa repens versus Tamsulosin Plus 5-Alpha-Reductase Inhibitors for Moderate-to-Severe LUTS-BPH Patients: Results of a Paired Matched Clinical Study
Previous Article in Journal
The Role of Full-Thickness Skin Grafts in Patient’s Rehabilitation after Maxillectomy and Midface Defects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Therapeutic Applications of Natural Compounds in Diabetes-Associated Periodontitis

1
School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan
2
Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
3
Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
4
Department of Medical Research and Education, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yi-lan, Luodong 265501, Taiwan
5
Department of Anatomy, School of Medicine, China Medical University, Taichung 404333, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(13), 3614; https://doi.org/10.3390/jcm11133614
Submission received: 3 May 2022 / Revised: 10 June 2022 / Accepted: 15 June 2022 / Published: 22 June 2022
(This article belongs to the Section Dentistry, Oral Surgery and Oral Medicine)

Abstract

:
Diabetes mellitus (DM) is a major worldwide health burden. DM is a metabolic disease characterized by chronic hyperglycemia, and if left untreated, can lead to various complications. Individuals with uncontrolled DM are more susceptible to periodontitis due to both a hyper-inflammatory host response and an impaired immune response. Periodontitis, on the other hand, may exacerbate DM by increasing both local and systemic inflammatory components of DM-related complications. The current standard for periodontal treatment in diabetes-associated periodontitis (DP) focuses mostly on reducing bacterial load and less on controlling the excessive host response, and hence, may not be able to resolve DP completely. Over the past decade, natural compounds have emerged as an adjunct approach for modulating the host immune response with the hope of curing DP. The anti-oxidant, anti-inflammatory, and anti-diabetic characteristics of natural substances are well-known, and they can be found in regularly consumed foods and drinks, as well as plants. The pathophysiology of DP and the treatment benefits of various bioactive extracts for DP will be covered in this review.

1. Introduction

About 537 million people worldwide have diabetes mellitus (DM) and this number is expected to rise drastically to 643 million by 2030 and 783 million by 2045 [1]. According to WHO, DM is a chronic, metabolic disease characterized by elevated levels of blood sugar, which over time, leads to serious damage to the heart, blood vessels, eyes, kidneys, and nerves. Untreated DM also poses a major risk for periodontitis, a multifaceted local inflammatory condition of the tooth-supporting structures [2]. Periodontitis normally develops as a result of the host’s reaction to local microbial stimuli and excessive blood sugar levels contribute to the prevalence and severity of this local disease by amplifying the host response [3]. Periodontitis is mainly characterized by periodontal pocket formation, loss of connective tissue attachment, alveolar bone resorption, and if left untreated, can lead to tooth loss [4]. Some studies have revealed that an altered host immune response in DM results in excessive inflammation and increases the severity of periodontal tissue destruction in diabetes-associated periodontitis (DP) [5,6]. In fact, patients with DP exhibit greater alveolar bone loss and a poorer prognosis after standard treatments compared with patients who do not have DM [5].
At times, conventional methods, such as mechanical scaling and root planing (SRP) and antiseptic mouthwash, may not be able to resolve DP completely. SRP involves mechanical removal of the bacterial biofilm that has adhered to the tooth or root surfaces, although it may not fully eradicate bacteria, especially in difficult-to-reach spots, such as the furcation areas. Antiseptic mouthwash, such as chlorhexidine (CHX), is often used as an adjunct to periodontal therapy since it is effective against various oral microbes. However, a few studies have revealed that prolonged use of CHX can cause several adverse effects, including tooth discoloration, parotid gland swelling, and taste disturbances [7]. Aside from that, SRP is occasionally supplemented with systemic antibiotics in cases of aggressive periodontitis. Even so, repeated and frequent use of antibiotics promotes the development of bacterial resistance, which is a grave public health concern [8]. The mentioned standard periodontal treatments principally focus on reducing bacterial burden and less on controlling the excessive host response in DP. Thus, it is imperative to focus on alternative products that can regulate the host’s hyper-inflammatory state to curb DP. Over the past decade, natural compounds have emerged as an adjunct approach for modulating the host immune response in an attempt to cure DP. Natural compounds in commonly consumed foods and beverages, as well as herbs, are well-known for their anti-oxidant [9], anti-inflammatory [10], and anti-diabetic properties [11]. Although recent reviews have assessed the impact of natural compounds on diabetes [12,13,14] and periodontitis [15,16], reviews on the effect of natural product on DP are lacking. To this end, this review includes all animal and clinical studies that have reported outcomes for natural compounds in DP. To date, the natural compounds that can modulate DP are resveratrol [17,18,19,20,21], curcumin [22,23,24,25,26,27], melatonin [28,29,30,31,32], propolis [33,34,35], vitamin C [36,37,38], green tea [39,40,41], ginger [42], aloe vera [43], rutin [26], and vitamin E [44]. Thus, this review will cover the pathogenesis of DP and the therapeutic effects of the above mentioned extracts on DP.

1.1. Protocol and Search Strategy

The protocol for this scoping review was performed according to the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR). The purpose of this study was to determine the impact of different natural products on DP as stated in the present literature. PubMed, Embase, and WoS databases were searched for the keywords (“natural products” OR natural compounds OR herbs OR natural medicine OR anti-oxidant) AND (“periodontitis”) AND (“diabetes”) from 2012 to the end of April 2022. The included articles were supplemented by a manual search of the bibliographies of all listed papers.
The inclusion criteria required studies of natural products and their effect on DP in vivo. No restrictions were placed on the language. Exclusion criteria included commentary, reviews, systematic reviews, meta-analyses, and books/book chapters. As for study selection and data extraction, the selection process involved three stages (Figure 1): (1) databases were searched, duplicate papers were manually eliminated, and the remainder screened for eligibility; (2) papers retrieved in stage 1 were screened based on titles and abstracts using the inclusion and exclusion criteria; (3) complete texts were analyzed using the same criteria. Figure 1 depicts the flow of the study selection process.

1.2. Pathogenesis of Diabetes-Associated Periodontitis

In diabetic patients, hyperglycemia induces production of advanced glycation end-products (AGEs) [45]. AGEs play a crucial role in the progression of DM-related complications [46], for instance, periodontitis. Periodontitis is a local inflammatory condition of the tooth and surrounding structures, such as connective tissue and alveolar bone [4]. This localized inflammatory condition is mainly triggered by the host response to pathogenic bacteria in the oral cavity, which causes the structures to break down. It was evidenced that the prevalence and severity of periodontitis is greater among diabetic subjects compared to non-diabetic patients [47,48]. In DM, the build-up of AGEs leads to increased susceptibility to periodontitis through several mechanisms [49,50]. It is thought that AGEs can alter immune cells, stimulate production of reactive oxygen species (ROS) leading to increased oxidative stress [51], and enhance the release of proinflammatory mediators [52].
The interaction between AGE and its receptor (RAGE) yields long-term effects on neutrophils, macrophages, and monocytes, which are the key cellular targets of diabetes-associated periodontitis (DP). People with DM often have enhanced neutrophils activation [52] and a shift in monocyte/macrophage phenotype [49]. Activated neutrophils trigger the production of a variety of chemokines and cytokines necessary for inflammatory and immune responses [53]. These include the promotion of cellular oxidant stress, activation of the transcription factor nuclear factor kappa B (NF-κB), and production of pro-inflammatory cytokines, such as IL-1β and TNF-α [49,54,55]. With exaggerated activation of neutrophils, proteases are released into the surrounding periodontal tissues that can cause excessive damage [56,57].
Among the downstream effects of inflammatory cytokines, the up-regulation of matrix metalloproteinases (MMPs) is controlled by activation of the transcription factor NF-κB [58,59,60]. It is well recognized that MMPs, such as the collagenases MMP-1, MMP-8, and MMP-13, and the gelatinases MMP-2 and MMP-9, are responsible for the destruction of various constituents of the extracellular matrix, including collagen [61,62]. Moreover, these MMPs produced locally by inflamed oral tissues can travel through the circulation and degrade insulin receptors. This indirectly contributes to insulin resistance, promotes hyperglycemia, and exacerbates the diabetic condition [63,64].
Oxidative stress is well recognized for its involvement in degradation of the extracellular matrix and stimulation of bone resorption by osteoclasts in periodontal tissue [65,66]. The possible sources of oxidative stress in DM include auto-oxidation of glucose, shifts in redox balances, decreased tissue concentrations of low molecular weight anti-oxidants, such as reduced glutathione (GSH), and impaired activities of anti-oxidant defense enzymes, such as superoxide dismutase (SOD) and catalase (CAT) [51]. In DM patients, the activity of NADPH oxidase (NOX) is elevated in activated neutrophils [53,67]. NOX is considered the major source of radical oxygen species (ROS) [67]. The generation of ROS by NOX, also known as the oxidative burst, results in the eradication of microbial invasion by macrophages and neutrophils, and hence functions as an inflammatory mediator [68]. Moreover, the impaired expression of anti-oxidants, which are pre-dominantly mediated by the NrF2 transcription factor, further contributes to ROS-associated damage [69].
Despite the fact that diabetes is an independent risk factor for periodontitis [70], periodontitis has a deleterious impact on the glycemic control of diabetes [71]. Diabetic patients with periodontitis may present with greater systemic inflammatory, as seen in elevated serum levels of IL-6, TNF-α, and CRP. A raised inflammatory state, on the other hand, can worsen both insulin resistance and glycemic control [72]. In a nutshell, individuals with DM appear to be more susceptible to periodontitis for two reasons: first, because of hyper-inflammatory components that lead to a predisposition for tissue destruction, and second, because of an impaired immune response, which may delay the host’s natural healing/regenerative abilities during the disease course. Periodontitis, on the other hand, might increase the diabetes-related host inflammatory component both locally and systemically, exacerbating DM. A summary of the pathogenesis of DP is illustrated in Figure 2. In the following sections, we will discuss the role of different natural compounds in modulating the host response in DP. Table 1 summarizes the study types of each natural compound and their clinical evaluation.

2. Therapeutic Effect of Natural Compounds in Diabetes-Associated Periodontitis

2.1. Resveratrol

Resveratrol (RSV) is a common stilbene present in various foods, such as grapes, peanuts, pistachio, and cranberry [73]. RSV is known for its anti-oxidant, anti-inflammatory, and even anti-cancer qualities [74,75]. The therapeutic potential of RSV for DP has been explored in three animal studies and two clinical trials, as tabulated in Table 2. All investigations agreed on the anti-inflammatory effects of RSV [18,20,21,76], and three confirmed its anti-diabetic [18,20,21] capabilities for managing DP. The results of these studies revealed a remarkable attenuation of bone loss, reduced inflammatory cytokines, and improved fasting blood glucose levels, so it can be considered an effective approach for reducing bone resorption and improving metabolic management.
Zhen et al. demonstrated the anti-inflammatory and anti-diabetic properties of RSV in experimental DP mice and murine gingival epithelial cells in vitro [21]. When RSV was administered to DP mice for 4 weeks, there was significant improvement in fasting blood glucose (FBG) and reduced alveolar bone loss compared to mice without RSV treatment. Moreover, supplementation repressed the elevated levels of TLR4 induced by DM/DP [77,78,79] as well as down-regulating inflammatory cytokine levels in both DP mice and gingival epithelial cells [76]. These findings strongly imply that RSV suppressed DP by improving hyperglycemia and the anti-inflammatory capability of the host by inhibiting both TLR4 expression and activation of its downstream signaling [80].
When Fabiano et al. compared the effect of RSV alone and RSV plus insulin (INS) on DP mice, the efficacy of RSV was comparable to insulin for reducing blood glucose levels [81]. RSV, in addition, had a synergistic effect with insulin on lowering blood glucose levels and inflammatory cytokines, such as IL-1β, IL-7, IL-6, and attenuating bone loss [81]. Furthermore, quantification of oxidative stress in biopsied gingiva from intervened DP mice revealed enhanced SIRT 1 and SOD levels, and reduced NADPH oxidase activity, similar to earlier studies [21,82]. Silent information regulator 1 (SIRT 1) [83] and superoxide dismutase 1 (SOD) are both anti-oxidative enzymes that regulate oxidative stress and protect cells from ROS toxicity [84,85].
In contrast, NADPH oxidase is an oxidative enzyme that catalyzes superoxide and peroxide generation, and hence, is responsible for the up-regulation of ROS production [86]. Since RSV has been shown to reduce the expression of NADPH oxidase in current and prior studies [87], this indicates that RSV can reduce the impact of oxidative stress mediated by ROS by quenching ROS molecules and reducing their production as well [88]. Thus, the anti-oxidant capacity of RSV is linked to its influence on SIRT 1, SOD 1, and NADPH oxidase levels, which helps to reduce the development and/or impact of oxidative stress.
Giménez-Siurana et al. employed a new drug delivery approach that incorporated RSV with silk fibroin nanoparticles (SFN) [19]. Silk fibroin (SF) has been shown to have an outstanding capacity to store a medication and serve as a vehicle for it in the form of nanoparticles [89,90,91]. This is particularly useful for overcoming the low bioavailability of RSV and its relatively rapid metabolism and elimination. Thus, in a DP rat model, Giménez-Siurana et al. compared the anti-inflammatory efficacy of liquid SF, RSV-SFN, and combination of both (SF + RSV-SFN). Treatment with RSV-SFN demonstrated the least inflammatory impact, whereas pairing it with liquid SF resulted in greatest reduction in inflammatory cytokines (IL-1β and IL-6) and thinnest epithelium [89,92]. This indicates that pairing the two might have a synergistic effect on inflammation in DP. Encapsulating polyphenols in SF nanoparticles has been shown to boost their capacity, improve their stability in the body, and prevent them from passing through unwanted metabolic processes [89].
Zare Javid et al. (2017) examined the anti-hyperglycemic and anti-inflammatory potential of RSV in clinical patients with DP [18]. Subjects with DP were randomized into two groups: intervention and control that received either RSV or placebo capsules, respectively, for four weeks along with non-surgical scaling and root planning (SRP). Clinically, supplementation resulted in significantly reduced periodontal pockets (PD) and better insulin levels and insulin resistance compared to controls, which has been described in previous studies [88,93]. Akin to earlier studies [93,94], RSV resulted in improved fasting blood glucose levels, however the difference was not significant when compared to the controls.
After 1 month of RSV supplementation, the same group reported a substantial reduction in inflammation, as measured by lower IL-6 levels, compared to the baseline [17]. Although this study demonstrated an insignificant improvement in total anti-oxidant capacity (TAC) in the test group, another study revealed otherwise [95]. A considerable reduction was observed in normocholesterolemic (NCs) individuals [95] and healthy subjects [96]. These discoveries suggest that RSV may improve TAC in healthy humans, but not in patients. In terms of periodontal status, CAL was examined and considerable improvements were observed in both groups; however, RSV supplementation alone did not significantly enhance the periodontal condition. CAL is a more precise metric since it uses a fixed reference point, typically the cemento-enamel junction (CEJ).
The investigations conducted on cell cultures and animal models show promising results for the effects of RSV in treating DP. The anti-diabetic, anti-inflammatory, and anti-oxidative potential of RSV seems to be enhanced when used in conjunction with diabetes medication (e.g., insulin) or delivered via nanoparticles (e.g., silk fibroin). Although the pre-clinical studies demonstrated remarkable outcomes, the limited clinical trials did not provide promising results. RSV in adjunct to SRP significantly resolved inflammation and improved PD, however it did not significantly alter fasting blood glucose, TAC, or CAL, which is a more accurate measure for periodontal loss around a tooth. This could be due to low bioavailability of RSV following oral administration and its quick metabolism and elimination. Thus, future research is crucial to improve the formulation and delivery system for RSV.

2.2. Curcumin

Curcumin (CUR) has been extensively studied these recent years for its therapeutic potential to modulate the immune system in DP [97]. It is a diferuloylmethane derived from the yellow spice turmeric, which is derived from the plant Curcuma longa. It has been reported as an efficient anti-oxidant, protecting numerous organs from oxidative stress-induced pathophysiology, including DP [23,24,98]. One clinical study and five pre-clinical studies were included in this review, as represented in Table 3.
This review identified three animal studies that evaluated the effect of a chemically modified curcumin (CMC2.24) on DP [22,23,24]. CMC2.24 is a phenylamino carbonyl curcumin, which is tri-ketonic in contrast to di-ketonic natural curcumin compounds. The advantages of this compound over the natural CUR includes greater efficacy as an MMP inhibitor and improved bioavailability. There was a consensus among the three studies, confirming the impact of CMC2.24 on bone loss reduction and inflammatory markers, where the activity of MMPs and the levels of pro-inflammatory cytokines were inhibited [22,23,24]. The outcomes of the mentioned studies confirmed that administration of CMC2.24 has an anti-inflammatory effect, and suggests it as an efficacious treatment for reducing bone resorption.
In 2016, Elburki et al. demonstrated the efficacy of CMC2.24 on DP rats, where they found a substantial reduction in alveolar bone loss and remarkable suppression of the pathological release of MMP-2, MMP-8, MMP-9, IL-6, and IL-1β [24]. It is well recognized that MMPs are responsible for the destruction of various constituents of the extracellular matrix, including collagen. In the following year (2017), the inhibitory effect of CMC2.24 on bone loss was shown to be mediated via the suppression of p38 MAPK and NF-κB signaling. Interestingly, CMC2.24 has been shown to be effective in DP in the presence of continuing severe hyperglycemia [99,100,101,102].
Likewise, Deng et al. demonstrated that CMC2.24 exerted no influence on glycemic control, however it was still able to "normalize" inflammatory and other cells. The compound reduced abnormal macrophage accumulation, improved defective chemotactic activity, and significantly enhanced resolvin (RvD1) activity [22]. The increased activity of RvD1 following CMC2.24 intervention was also reported in an earlier study [103]. RvD1 belongs to a group of molecules derived from omega-3 fatty acids that can orchestrate the resolution of acute inflammation [104]. These results suggest that CMC2.24 has no effect on blood glucose levels (at least in the short term), but it can target the complications of uncontrolled hyperglycemia involving an inflammatory/immune response that is exacerbated by host-microbial interactions.
Unlike CMC2.24, natural CUR possesses the ability to improve blood glucose levels. When Pimentel et al. compared the effect of natural CUR and insulin on combating DP, both had a similar impact on blood glucose [27]. When CUR and insulin were used in synergy, the improvements were superior in terms of alveolar bone level, glycemic control, and pro-inflammatory cytokine levels (IL-6 and IL-1β). Natural CUR was also found to significantly up-regulate RUNX2, a gene involved in the development and maintenance of the teeth, bones, and cartilage. Furthermore, CUR encouraged the up-regulation of SIRT [105], a protein that is persistently found reduced in DM and is associated with the regulation of mitochondrial anti-oxidant defense [106]. Moreover, when CUR was combined with insulin, the ratio of receptor activator by NF-κB ligand (RANKL)/OPG was decreased. The anti-inflammatory/oxidative activity of CUR and its modulation of RANKL/RANK/OPG expression may play crucial roles in delaying osteoclastogenesis and decreasing alveolar bone loss [107].
The anti-oxidative ability of CUR was further described in an animal investigation by Iova et al. [26]. CUR was shown to reduce oxidative stress, both locally and systemically, and boosted anti-oxidant enzymes, such as glutathione (GSH), oxidized glutathione (GSSG), GSH/GSSG, and catalase (CAT). These anti-oxidant enzymes are renowned for their ability to reduce oxidizing species, such as superoxide, peroxyl, and hydroxyl radicals [108,109].
In a clinical trial carried out by Ivanaga et al. [25], the clinical efficacy of CUR solution as a photo-sensitizer was evaluated for treating residual pockets in DP patients. After 6 months, it was found that LED irradiation alone or with CUR solution as an adjunct therapy to SR, provided a short-term (3 months) clinical benefit in terms of CAL gain, however there were no significant differences between the groups.
CMC2.24 and natural CUR each combat alveolar bone loss in DP in a slightly different way. CMC2.24 targets the hyper-immune/inflammatory response via suppression of MMPs, pro-inflammatory cytokines, and restoration of impaired immune cells while exerting no effect on blood glucose levels (at least in the short term) [22,23,24]. On the other hand, natural CUR repressed uncontrolled hyperglycemia, increased anti-oxidant capacity, and up-regulated anti-inflammation [25,26,27]. Caution must be taken when taking CUR together with anti-diabetic drugs to prevent hypoglycemia. Despite the hopeful results, toxicity and weight loss, as well as other negative consequences, were not observed in any of the trials considered, and the advantages of higher/lower dosages were not established. Different animal models are being used to conduct pharmacokinetic and safety research, which are required before this chemical can be tested in human clinical trials.

2.3. Melatonin

Melatonin‘s primary role is to regulate the sleep cycle in a circadian manner [110]. It is a common prescription sleep medication to regulate sleep. Melatonin (MLT) can be sourced from many plants, including Feverfew (Tanacetumparthenium) and St John’s wort (Hypericumperforatum) [111]. Interestingly, few studies have associated poor sleep quality with an increased risk of diabetes or a worsening of the condition [112,113], so it is worth looking into whether the effect of MLT in DP is attributable to better sleep quality. During the last decades, MLT was discovered as a noteworthy free radical scavenger [114,115,116] and has anti-inflammatory and glucose-regulatory properties [117,118]. As listed in Table 4, this review identified five clinical studies, two of which utilized topical application of MLT in cross-sectional studies, whereas the others utilized oral intake of the compound in clinical trials.
In the cross-sectional studies by Cutando et al. [31,32], DP patients were given topical MLT (1% orobase cream formula) in both upper and lower arches on the attached gingival surface. After 20 days of application, the results were measured in terms of clinical and biological parameters. Topical MLT significantly decreased the gingival index and pocket depth as well as serum IL-6 and CRP levels. Apart from its remarkable anti-inflammatory potential, topical MLT also down-regulated salivary RANKL and elevated salivary OPG levels, which is consistent with earlier studies [119,120,121]. The present data implies that RANKL is a key cytokine in bone destruction during periodontitis, and that OPG, as a RANKL decoy receptor, protects against destructive bone disease [122,123]. In light of this, MLT may have a favorable impact in slowing osteoclastogenesis, improving alveolar bone quality, and preventing the development of periodontal disease.
On the other hand, oral intake of MLT alongside SRP in investigations by Anton et al. [28] and Bazyar et al. [30] demonstrated an anti-inflammatory phenotype characterized by significantly improved PD and CAL, and repression of pro-inflammatory cytokines, such as IL-6, hs-CRP, and IL-1β. Moreover, MLT appears to have beneficial glucose-metabolic effects, since patients exhibited lower HbA1c after taking 3 mg of MLT (lower dosage) daily for 8 weeks. MLT consumption has been linked to improved glycemic management [124], and its use during the daytime has been linked to lower insulin sensitivity in other investigations [125,126,127].
Zare Javid et al. showed that ingestion of a higher dose of MLT (6 mg) daily for 8 weeks resulted in superior improvement for three anti-oxidant enzymes (SOD, CAT and GPX) and a lipid peroxidation marker (MDA) compared to the placebo group [29]. Previous studies demonstrated doses of 2 mg and 5 mg of MLT daily did not up-regulate these anti-oxidant enzymes, but both dosages significantly decreased MDA [128]. Since MDA is a marker of oxidative stress [129], this suggests that lower doses of MLT can exert anti-oxidant properties regardless of their effect on anti-oxidant enzymes. More research is needed to determine if the effect of MLT-induced overexpression of all three anti-oxidant enzymes on the anti-oxidant defense system is more or less effective as lower doses of MLT. If the higher doses are no better, then prescribing a lower dose of MLT is recommended.
From the clinical trials results, topical application of MLT for 20 days shows remarkable inflammation suppression, both clinically and biologically [31,32]. It is able to modulate local levels of pro-inflammatory cytokines and the RANKL/OPG ratio to slow osteoclastogenesis and deter the progression of DP. As for systemic intake of the supplement, MLT serves as a potent anti-oxidant [130] that is protective against oxidative stress [131] in periodontal tissue with clinical effects. Although MLT has been reported to modulate glucose-metabolic effects [132], it is still crucial that DM patients consult a physician beforehand to prevent excessive hypoglycemia when paired with anti-diabetic medication. Currently, there is no official recommended maximum dose for MLT in adults, but a range of 1 mg to 6 mg appears to be safe and effective [133]. Therefore, a low dose of MLT (under 6 mg) provides a therapeutic benefit as an adjunct to periodontal treatment for DP.

2.4. Propolis

Propolis (PP) is an organic resinous structure made by honeybees from a range of substances, such as plant parts, buds, and sap. The content of PP varies based on the plant source, which differs depending on the geographic zone, such as Brazil, Peru, Taiwan, and Europe. The biological and pharmacological capabilities of PP, which include anti-inflammatory, anti-bacterial [134], anti-oxidant [134,135], immunomodulatory actions [136], and anti-diabetic and anti-hyperglycemic properties [137], have sparked considerable interest. Recently, caffeic acid phenethyl ester (CAPE) has been discovered to be an important active molecule in PP and is responsible for most of its therapeutic properties [138].
To date, there have been two pre-clinical and one clinical study on the use of PP in DP, as summarized in Table 5. Aral et al. administered PP in DP mice models and showed that PP managed to reduce elevated blood glucose levels and repress alveolar bone loss, albeit with an insignificant decrease in pro-inflammatory cytokines [35]. In the study by Kızıldağ et al., the authors used caffeic acid phenethyl ester (CAPE) and investigated its effects in DP mice [34]. The exceptional anti-oxidative potential of CAPE was proven by up-regulated total anti-oxidant status as well as lowered total oxidant status and oxidative stress, comparable to a previous work [139]. Histochemistry analysis of the periodontal tissue demonstrated high scores for inflammatory reactions, ulcers, hyperemia, and the density of RANKL-stained cells, which were all alleviated after 15 days of CAPE intake. A high RANKL:OPG ratio has been implicated in the high rate of osteoclastogenesis and alveolar bone resorption during the course of periodontitis [140,141]. In line with previous evidence [142], it is plausible to suggest that CAPE exhibits the ability to inhibit ROS production, oxidative stress, and RANKL-induced osteoclastogenesis as it accelerates bone healing.
Vo et al. studied the influence of Taiwanese green propolis (TGP) on human gingival fibroblasts, and it was found to alleviate inflammation induced by high glucose levels [143]. This attenuation of inflammation occurred via inhibition of the NLRP3 inflammasome and its inflammatory cascade, which has been reported in other pathologies, such as gouty arthritis [144]. Activation of the NLRP3 inflammasome is known to be pro-inflammatory because it activates caspase-1, which governs pro-IL-1β maturation [145,146]. TGP targeted the upstream regulatory body involved in NLRP3 activation, including the NF-κB signaling pathway, Toll-like receptors (TLRs), and ROS production to protect gingival fibroblasts from inflammation.
Outcomes from the pre-clinical studies were comparable to those of the clinical study [33,143]. When DP patients were administered 400 mg PP daily for 6 months along with SRP treatment, glycemic control improved remarkably at both 3 and 6 months [33]. The clinical efficacy of PP was proven by significant improvements in PD and CAL in DP patients. Overall, the results for the long-term effectiveness of PP as an anti-diabetic, anti-inflammatory, and anti-oxidant for treating DP are promising. However, special precautions should be taken by patients who are allergic to bees or bee product, such as honey and conifers, since it may trigger allergic reactions.

2.5. Vitamin C

Vitamin C (Vit. C) has long been known as an essential micronutrient for humans that can be obtained from various fruits and vegetables, such as guavas, lemons, and broccoli. In subjects with DM, plasma Vit. C was significantly lower compared to healthy individuals [147]. Vit. C, which belongs to the group of free radical-scavenging anti-oxidants, promotes wound healing [148,149] and its supplementation in DP patients has been of interest lately.
This review included one animal study and two clinical trials, as demonstrated in Table 6. According to Toraman et al., local Vit. C treatment in DP rats significantly repressed alveolar bone loss (ABL) along with reduced MMP-8 and 8-OHdG in tissues, and serum CTX [36]. MMP-8 is known for its involvement in ECM breakdown, which can contribute to the severity of periodontitis [150], and 8-hydroxy2-deoxyguanosine (8-OHdG) is a critical biomarker of oxidative stress. Considering Vit. C has been shown to inhibit MMPs in other cells [151], it was proposed that Vit. C might help in suppressing oxidative stress and inflammation during DP, resulting in decreased bone resorption.
When Vit. C (450 mg) was given as a supplement to patients with DP for 2 weeks along with SRP, the bleeding index improved significantly compared to controls, despite unchanged PD and PI [38]. Similar outcomes have been described in non-smoker patients with periodontitis [136,152]. Although there were no changes in the PD and PI, it is nevertheless considered a slight clinical success since a decrease in bleeding tendency indicates the resolution of inflammation in the gingiva at least. When the dosage and period of Vit. C was increased to 500 mg for 2 months, no additional benefits were observed [37]. Plasma Vit. C was significantly increased in the intervention group, however there were no significant differences in periodontal parameters between the treatment and placebo groups, although they both improved from the baseline.
Vit. C did not alter FBS and HbA1c in DP patients, similar to another report [153]. It was reported that a higher dose of Vit. C (1000 mg) could elicit significantly decrease blood glucose [154]. Cellular uptake of vitamin C is promoted by insulin and inhibited by hyperglycemia [155]. This explains why higher doses of ascorbic acid might be necessary to overcome the effect of hyperglycemia to exert a beneficial effect.
In conclusion, supplementing initial periodontal treatment with 500 mg of Vit. C per day did not improve the periodontal state in periodontitis patients with uncontrolled type 2 diabetes. However, given the promising outcome for local Vit. C in the DP mouse model, and considering the challenge of cellular uptake of systemic Vit. C during hyperglycemia [36], it is worth investigating the effect of topical Vit. C in DP patients. Moreover, local injections of Vit. C have been shown to be an effective adjunctive treatment for reducing gingival inflammation in DP rats [156].

2.6. Green Tea

Green tea (GT) is known for its anti-oxidative [157], anti-bacterial [158], anti-inflammatory [158], and anti-diabetic [159] abilities. GT is a popular beverage across the world. It is often brewed from the dried leaves of Camellia sinensis, and the majority of its polyphenols are catechins, such as (-)-epicatechin (EC), (-)-epicatechingallate (ECG), (-)-epigallocatechin (EGC), and (-)-epigallocatechin 3-gallate (EGCG) [160,161].
Table 7 summarized the use of GT in DP. To date, there are one animal study and two clinical studies of GT in DP. In an early clinical trial (2013), Gadagi et al. used GT strips combined with supragingival scaling in periodontitis patients, both diabetic and non-diabetic, and evaluated its microbiological and clinical efficacy [39]. With hydroxypropyl methylcellulose (HPMC) as a vector, the GT was slowly released to the surrounding tissue, with full release occurring after 2 h. As a result, the clinical parameters, including GI, PD and CAL, improved in both groups (DM and non-DM) with GT intervention. At the same time, P. gingivalis was reduced at GT sites in non-DM patients (significantly) and DM patients (not significantly), according to its anti-microbial impact.
Apart from its anti-bacterial properties, with consistent clinical outcomes of the above study [39], some studies have observed that GT could return the levels of pro-inflammatory cytokines, TNF-α [162,163] and RANKL, [164] to normal while raising the expression of IL-10, RUNX-2, and OPG [41]. The down-regulation of pro-inflammatory cytokines coupled with up-regulation of IL-10 demonstrates that GT is a powerful anti-oxidant. This potential has been associated with suppression of the RANK system, synthesis of the anti-osteoclastogenic factor OPG, and a slowing down of resorption activity in the jawbone [165,166].
Catanzaro et al. demonstrated that giving DP mice GT improved their glycemic control, reduced plaque accumulation, and prevented periodontal disease progression [40]. Distinct from other studies [39,41], the authors investigated the effect of GT on the modulation of vascularization during the progression of DP. Chronic hyperglycemia is known to promote microvascular damage in the periodontium and this alteration has been associated with greater periodontal tissue loss in DM rats [167]. In individuals with microvascular complications, elevated VEGF expression has been found systemically [168,169]. The consumption of GT successfully inhibited microvascular alteration and returned the over-expression of VEGF to normal levels in DP mice.
In a nutshell, GT possesses anti-hyperglycemia, anti-inflammatory, anti-osteoclastogenic, and anti-oxidative effects in addition to inhibiting microvascular damage in periodontal tissue. One of the limitations of the animal studies is that the concentration of GT (which was brewed in water) in the animal models was not stated, since it may be challenging to regulate the volume of consumption by each rat. Be that as it may, the clinical trial showed promising results for locally applied GT in combating DP, and more studies should be carried out to enhance or extend the release of GT into periodontal tissue.

2.7. Ginger

Ginger is the rhizome of the plant Zingiberofficinale Roscoe and is one of the most widely used spice/herb worldwide [170]. Its major components, shogaol and gingerol, have demonstrated many beneficial pharmacological uses, including anti-diabetic, anti-oxidant, anti-inflammatory, anti-microbial, and anti-cancer [171]. Table 8 tabulated the research on the use of ginger in DP models.
When patients with mild to moderate DP were given 2 g of ginger daily for 8 weeks with SRP therapy, the patients demonstrated substantial clinical and biochemical inflammation resolution compared to the placebo group [42]. There were significant reductions in CAL and PD as well as decrements in inflammatory markers, such as IL-6, hs-CRP, and TNF-α. Likewise, Rehman et al. showed that zingerone therapy significantly decreased ROS levels, blocked nuclear factor-kappa B (NF-κB) activation, and reduced the levels of other inflammatory markers, including TNF-α, IL-6, and IL-1β in type 2 diabetic Wistar rat [172]. Apart from its anti-inflammatory effect, 6-shogaol has been reported to up-regulate the expression of anti-oxidants such as GPx and SPD in a clinical trial [42] as well as in animal models [173]. These findings indicate that ginger may protect DM patients from oxidative damage by decreasing oxidative stress and minimizing anti-oxidant depletion [174]. In a study of its molecular mechanism, 6-shogaol, the most abundant component in shogaol extract, demonstrated powerful anti-oxidative capacity by regulating oxidative and anti-oxidative activities in human gingival fibroblasts (HGF) [175]. In addition, 6-shogaol was found to inhibit ROS production [176], increase levels of HO-1 [177] and NQO1, and ameliorate the inflammatory cascade [178,179].
Overall, the anti-inflammatory and anti-oxidative abilities of ginger supplementation may be more effective for the control of systemic inflammation in DP patients compared to SRP alone, at least for a short period (2 months). It is recommended that future studies include post-intervention glycemic status to assess the anti-diabetic capacity of ginger and for any side effects, especially when paired with medications that lower blood glucose in DM patients.

2.8. Others: Aloe Vera, Rutin, Vitamin E

2.8.1. Aloe Vera

The research on the use of aloe vera (AV), rutin (RT), and vitamin E (Vit. E) in DP were listed in Table 9. Aloe vera (Aloe barbadensis Miller), also known as Ghritkumari/Gwarpatha, is a member of the Liliaceae family and has been utilized in traditional medicines for centuries. Aloe vera (AV) is well-known for its various therapeutic properties, for example, anti-inflammatory [180], anti-oxidant [181], anti-bacterial [182], and immune boosting properties. Apart from that, it has been shown in diabetic individuals to exhibit hypoglycemic and hypolipidemic effects [183]. Only one clinical study has assessed the effectiveness of AV in DP prevention. Pradeep et al. delivered AV in gel form to the periodontal pocket of DP subjects and measured its clinical effectiveness 3- and 6-months post-treatment [43]. When compared to the control group, the intervention group showed greater improvements in PI, mSBI at 3 months, and significant CAL gain at 6 months. This improvement might be explained by the antibacterial action of AV against periodontal infections, which has been observed in various investigations [182,184]. When used as an adjunct to SRP, local application of AV gel can benefit DP patients in terms of both immediate and long-term clinical efficacy, even in moderate to severe cases. To corroborate the findings of this investigation, long-term, double-blind, multicenter randomized clinical studies are required, and should include glycemic state assessment, histological bone gain evaluation, and microbiologic profiles.

2.8.2. Rutin

Rutin, a citrus flavonoid glycoside found in fruits such as orange, grapefruit, lemon, apples, and berries is a common element in multivitamin and herbal cures [185]. This complex has a wide variety of medicinal purposes, including anticancer, antibacterial, and anti-inflammatory effects [186,187]. Furthermore, rutin has been found to be effective in diabetes and malignancies [188]. Rutin, alone or in combination with curcumin, has been proven to reduce malondialdehyde (MDA), an oxidative stress biomarker, in gingiva tissue and blood when compared to rats without therapy. Moreover, rutin significantly enhanced anti-oxidant enzymes, including glutathione (GSH), oxidized glutathione (GSSG), GSH/GSSG, and catalase (CAT). These enzymes possess reducing characteristics for various oxidizing species, such as peroxyl, superoxide, and hydroxyl radicals [189]. Although it has been established that increased MDA levels and reduced GSH levels in diseased periodontal tissue correlate with more severe alveolar bone loss, future research is needed to evaluate whether rutin can transfer its promising effects to DP patients.

2.8.3. Vitamin E

Vitamin E is a fat-soluble vitamin, which consists of two main forms: α-tocopherols and γ tocopherols. The anti-oxidative properties of vitamin E have been well recognized along with its role in anti-inflammatory processes, and it has been implicated in preventing diseases such as cardiovascular diseases and cancer [190]. At present, vitamin E inhibited inducible nitric oxide synthase (iNOS) activity in a murine DP model. It was postulated that vitamin E represses gingival inflammation via lowering NO and hence oxidative stress, which agrees with a previous study on rats with periodontitis [191]. iNOS is a type of NO synthase present in macrophages [192,193] that releases nitric oxide (NO), a short-lived free radical [194]. iNOS expression has been shown to be higher in gingiva obtained from mice and patients with DP [195,196,197], however iNOS expression did not appear to have an additional deleterious effect on the clinical course of DP compared to periodontitis alone [197]. Due to the limited data concerning the pathogenic function of iNOS in the progression of DP, additional research is needed, particularly on vitamin E’s influence on periodontal and glycemic parameters in DP models to determine its clinical efficacy in combating DP.

3. Strengths, Limitations and Future Directions

By offering a broad review of pertinent research, this scoping study evaluated the existing knowledge of the issue of interest, noting trends, advancements, and comparability within and across each natural product. This review also enables us to identify gaps in the literature, such as the impact of different administration routes (topical vs. systemic) and doses on DP, as well as the uniformity of data collection methods. Additionally, our results may provide a platform for future research to contribute into the formulation of adjunct therapy using natural products in patients with DP. Nevertheless, this review has certain limitations, for example, to conclude a definitive effect of each natural bioactive on DP is nearly impossible due to the limited research data and the lack of homogeneity among study designs. Another factor is the small sample size in most randomized clinical trials (RCTs) with short follow-up period.
Considering the shortcomings of the present data mentioned in this review, future research might include the following suggestions. Clinical trials should include: (1) detailed descriptions of inclusion and exclusion criteria, case definition of DM, severity of periodontitis, etc.; (2) bigger sample sizes and repeated HbA1c to minimize inaccuracy; (3) longer follow-up to assess the sustainability of the treatment’s impact and any unwanted effects; (4) emphasis on designing a double-blind studies to reduce any bias risk. As for animal studies, they should include: (1) accurately described methods for the induction of either DM type 1 or 2; (2) measure accelerated periodontitis in animals (e.g., rats) that are extremely resistant to periodontal diseases under normal circumstances including ligature fixation, bacteria inoculation, etc. [198].

4. Conclusions

Usage of natural products has been practiced since ancient times and has continued in the current civilization for well-being and treatment for various disorders. Over the past decade, natural compounds have emerged as an adjunct approach for modulating the host immune response to resolve diabetes-associated periodontitis. Our paper has reviewed the data from various studies on the subject, focusing on the therapeutic benefits of natural compounds for diabetes-associated periodontitis. According to the findings, the mentioned natural compounds have a variety of positive pharmacological qualities, such as anti-oxidative, anti-inflammatory, anti-osteoclastogenesis, and anti-bacterial. Since these natural compounds possess medicinal potential for curbing the hyperinflammatory components of the host response, they might serve as novel adjunct remedies for controlling diabetes-associated periodontitis. However, more study is needed to maximize their therapeutic effect as well as determining the amount of natural extract in the final product, which can then be evaluated in well-designed clinical trials.

Author Contributions

Conceptualization, C.-C.Y.; writing—original draft preparation, M.Y.N., T.L. and S.-C.C.; writing—review and editing, P.-M.C. and C.-C.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Ministry of Science and Technology (MOST 108-2314-B-040-033-MY2) and China Medical University (CMU110-S-41) in Taiwan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. IDF Diabetes Atlas. 2021. Available online: https://diabetesatlas.org/atlas/tenth-edition/ (accessed on 28 March 2022).
  2. Wu, C.Z.; Yuan, Y.H.; Liu, H.H.; Li, S.S.; Zhang, B.W.; Chen, W.; An, Z.J.; Chen, S.Y.; Wu, Y.Z.; Han, B.; et al. Epidemiologic relationship between periodontitis and type 2 diabetes mellitus. BMC Oral Health 2020, 20, 204. [Google Scholar] [CrossRef] [PubMed]
  3. Ryan, M.E.; Carnu, O.; Kamer, A. The influence of diabetes on the periodontal tissues. J. Am. Dent. Assoc. 2003, 134, 34s–40s. [Google Scholar] [CrossRef] [PubMed]
  4. Könönen, E.; Gursoy, M.; Gursoy, U.K. Periodontitis: A multifaceted disease of tooth-supporting tissues. J. Clin. Med. 2019, 8, 1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kim, J.H.; Lee, D.E.; Gunawardhana, K.S.; Choi, S.H.; Woo, G.H.; Cha, J.H.; Bak, E.J.; Yoo, Y.J. Effect of the interaction between periodontitis and type 1 diabetes mellitus on alveolar bone, mandibular condyle and tibia. Acta Odontol. Scand. 2014, 72, 265–273. [Google Scholar] [CrossRef] [PubMed]
  6. Duarte, P.M.; Bezerra, J.P.; Miranda, T.S.; Feres, M.; Chambrone, L.; Shaddox, L.M. Local levels of inflammatory mediators in uncontrolled type 2 diabetic subjects with chronic periodontitis. J. Clin. Periodontol. 2014, 41, 11–18. [Google Scholar] [CrossRef] [PubMed]
  7. Pałka, Ł.; Nowakowska-Toporowska, A.; Dalewski, B. Is chlorhexidine in dentistry an ally or a foe? A narrative review. Healthcare 2022, 10, 764. [Google Scholar] [CrossRef]
  8. Haque, M.; Sartelli, M.; Haque, S.Z. Dental infection and resistance-global health consequences. Dent. J. 2019, 7, 22. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, S.; Hu, R.; Geng, Y.; Chen, K.; Wang, L.; Imam, M.U. The regulatory effects and the signaling pathways of natural bioactive compounds on ferroptosis. Foods 2021, 10, 2952. [Google Scholar] [CrossRef]
  10. Usmani, J.; Khan, T.; Ahmad, R.; Sharma, M. Potential role of herbal medicines as a novel approach in sepsis treatment. Biomed. Pharmacother. 2021, 144, 112337. [Google Scholar] [CrossRef]
  11. Mahdavi, A.; Bagherniya, M.; Fakheran, O.; Reiner, Ž.; Xu, S.; Sahebkar, A. Medicinal plants and bioactive natural compounds as inhibitors of hmg-coa reductase: A literature review. Biofactors 2020, 46, 906–926. [Google Scholar] [CrossRef]
  12. Infante-Garcia, C.; Garcia-Alloza, M. Review of the effect of natural compounds and extracts on neurodegeneration in animal models of diabetes mellitus. Int. J. Mol. Sci. 2019, 20, 2533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Choudhury, H.; Pandey, M.; Hua, C.K.; Mun, C.S.; Jing, J.K.; Kong, L.; Ern, L.Y.; Ashraf, N.A.; Kit, S.W.; Yee, T.S.; et al. An update on natural compounds in the remedy of diabetes mellitus: A systematic review. J. Tradit. Complement. Med. 2018, 8, 361–376. [Google Scholar] [CrossRef] [PubMed]
  14. Patle, D.; Vyas, M.; Khatik, G.L. A review on natural products and herbs used in the management of diabetes. Curr. Diabetes Rev. 2021, 17, 186–197. [Google Scholar] [CrossRef] [PubMed]
  15. Fakheran, O.; Khademi, A.; Bagherniya, M.; Sathyapalan, T.; Sahebkar, A. The effects of nutraceuticals and bioactive natural compounds on chronic periodontitis: A clinical review. Adv. Exp. Med. Biol. 2021, 1328, 59–80. [Google Scholar]
  16. Chen, Y.; Wong, R.W.; McGrath, C.; Hagg, U.; Seneviratne, C.J. Natural compounds containing mouthrinses in the management of dental plaque and gingivitis: A systematic review. Clin. Oral Investig. 2014, 18, 1–16. [Google Scholar] [CrossRef]
  17. Javid, A.Z.; Hormoznejad, R.; Yousefimanesh, H.A.; Haghighi-Zadeh, M.H.; Zakerkish, M. Impact of resveratrol supplementation on inflammatory, antioxidant, and periodontal markers in type 2 diabetic patients with chronic periodontitis. Diabetes Metab. Syndr. 2019, 13, 2769–2774. [Google Scholar] [CrossRef]
  18. Zare Javid, A.; Hormoznejad, R.; Yousefimanesh, H.A.; Zakerkish, M.; Haghighi-Zadeh, M.H.; Dehghan, P.; Ravanbakhsh, M. The impact of resveratrol supplementation on blood glucose, insulin, insulin resistance, triglyceride, and periodontal markers in type 2 diabetic patients with chronic periodontitis. Phytother. Res. 2017, 31, 108–114. [Google Scholar] [CrossRef]
  19. Giménez-Siurana, A.; Gómez García, F.; Pagan Bernabeu, A.; Lozano-Pérez, A.A.; Aznar-Cervantes, S.D.; Cenis, J.L.; López-Jornet, P. Chemoprevention of experimental periodontitis in diabetic rats with silk fibroin nanoparticles loaded with resveratrol. Antioxidants 2020, 9, 85. [Google Scholar] [CrossRef] [Green Version]
  20. Cirano, F.R.; Molez, A.M.; Ribeiro, F.V.; Tenenbaum, H.C.; Casati, M.Z.; Corrêa, M.G.; Pimentel, S.P. Resveratrol and insulin association reduced alveolar bone loss and produced an antioxidant effect in diabetic rats. J. Periodontol. 2021, 92, 748–759. [Google Scholar] [CrossRef]
  21. Zhen, L.; Fan, D.S.; Zhang, Y.; Cao, X.M.; Wang, L.M. Resveratrol ameliorates experimental periodontitis in diabetic mice through negative regulation of tlr4 signaling. Acta Pharmacol. Sin. 2015, 36, 221–228. [Google Scholar] [CrossRef]
  22. Deng, J.; Golub, L.M.; Lee, H.M.; Raja, V.; Johnson, F.; Kucine, A.; Lee, W.; Xu, T.M.; Gu, Y. A novel modified-curcumin promotes resolvin-like activity and reduces bone loss in diabetes-induced experimental periodontitis. J. Inflamm. Res. 2021, 14, 5337–5347. [Google Scholar] [CrossRef] [PubMed]
  23. Elburki, M.S.; Moore, D.D.; Terezakis, N.G.; Zhang, Y.; Lee, H.M.; Johnson, F.; Golub, L.M. A novel chemically modified curcumin reduces inflammation-mediated connective tissue breakdown in a rat model of diabetes: Periodontal and systemic effects. J. Periodontal Res. 2017, 52, 186–200. [Google Scholar] [CrossRef] [PubMed]
  24. Elburki, M.S.; Rossa, C., Jr.; Guimarães-Stabili, M.R.; Lee, H.M.; Curylofo-Zotti, F.A.; Johnson, F.; Golub, L.M. A chemically modified curcumin (cmc 2.24) inhibits nuclear factor κb activation and inflammatory bone loss in murine models of lps-induced experimental periodontitis and diabetes-associated natural periodontitis. Inflammation 2017, 40, 1436–1449. [Google Scholar] [CrossRef] [PubMed]
  25. Ivanaga, C.A.; Miessi, D.M.J.; Nuernberg, M.A.A.; Claudio, M.M.; Garcia, V.G.; Theodoro, L.H. Antimicrobial photodynamic therapy (apdt) with curcumin and led, as an enhancement to scaling and root planing in the treatment of residual pockets in diabetic patients: A randomized and controlled split-mouth clinical trial. Photodiagn. Photodyn. Ther. 2019, 27, 388–395. [Google Scholar] [CrossRef]
  26. Iova, G.M.; Calniceanu, H.; Popa, A.; Szuhanek, C.A.; Marcu, O.; Ciavoi, G.; Scrobota, I. The antioxidant effect of curcumin and rutin on oxidative stress biomarkers in experimentally induced periodontitis in hyperglycemic wistar rats. Molecules 2021, 26, 1332. [Google Scholar] [CrossRef]
  27. Pimentel, S.P.; Casati, M.Z.; Ribeiro, F.V.; Corrêa, M.G.; Franck, F.C.; Benatti, B.B.; Cirano, F.R. Impact of natural curcumin on the progression of experimental periodontitis in diabetic rats. J. Periodontal Res. 2020, 55, 41–50. [Google Scholar] [CrossRef] [PubMed]
  28. Anton, D.M.; Martu, M.A.; Maris, M.; Maftei, G.A.; Sufaru, I.G.; Tatarciuc, D.; Luchian, I.; Ioanid, N.; Martu, S. Study on the effects of melatonin on glycemic control and periodontal parameters in patients with type ii diabetes mellitus and periodontal disease. Medicina 2021, 57, 140. [Google Scholar] [CrossRef]
  29. Zare Javid, A.; Hosseini, S.A.; Gholinezhad, H.; Moradi, L.; Haghighi-Zadeh, M.H.; Bazyar, H. Antioxidant and anti-inflammatory properties of melatonin in patients with type 2 diabetes mellitus with periodontal disease under non-surgical periodontal therapy: A double-blind, placebo-controlled trial. Diabetes Metab. Syndr. Obes. 2020, 13, 753–761. [Google Scholar] [CrossRef] [Green Version]
  30. Bazyar, H.; Gholinezhad, H.; Moradi, L.; Salehi, P.; Abadi, F.; Ravanbakhsh, M.; Zare Javid, A. The effects of melatonin supplementation in adjunct with non-surgical periodontal therapy on periodontal status, serum melatonin and inflammatory markers in type 2 diabetes mellitus patients with chronic periodontitis: A double-blind, placebo-controlled trial. Inflammopharmacology 2019, 27, 67–76. [Google Scholar]
  31. Cutando, A.; Montero, J.; Gómez-de Diego, R.; Ferrera, M.J.; Lopez-Valverde, A. Effect of topical application of melatonin on serum levels of c-reactive protein (crp), interleukin-6 (il-6) and tumor necrosis factor-alpha (tnf-α) in patients with type 1 or type 2 diabetes and periodontal disease. J. Clin. Exp. Dent. 2015, 7, e628–e633. [Google Scholar] [CrossRef]
  32. Cutando, A.; López-Valverde, A.; de Diego, R.G.; de Vicente, J.; Reiter, R.; Fernández, M.H.; Ferrera, M.J. Effect of topical application of melatonin to the gingiva on salivary osteoprotegerin, rankl and melatonin levels in patients with diabetes and periodontal disease. Odontology 2014, 102, 290–296. [Google Scholar] [CrossRef] [PubMed]
  33. El-Sharkawy, H.M.; Anees, M.M.; Van Dyke, T.E. Propolis improves periodontal status and glycemic control in patients with type 2 diabetes mellitus and chronic periodontitis: A randomized clinical trial. J. Periodontol. 2016, 87, 1418–1426. [Google Scholar] [CrossRef] [PubMed]
  34. Kızıldağ, A.; Arabacı, T.; Albayrak, M.; Balseven, H.M.; Aksu Kızıldağ, C.; Tasdemir, U. A biochemical and immunohistochemical study of the effects of caffeic acid phenethyl ester on alveolar bone loss and oxidative stress in diabetic rats with experimental periodontitis. Biotech. Histochem. 2020, 95, 456–463. [Google Scholar] [CrossRef] [PubMed]
  35. Aral, C.A.; Kesim, S.; Greenwell, H.; Kara, M.; Çetin, A.; Yakan, B. Alveolar bone protective and hypoglycemic effects of systemic propolis treatment in experimental periodontitis and diabetes mellitus. J. Med. Food 2015, 18, 195–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Toraman, A.; Arabaci, T.; Aytekin, Z.; Albayrak, M.; Bayir, Y. Effects of vitamin c local application on ligature-induced periodontitis in diabetic rats. J. Appl. Oral. Sci. 2020, 28, e20200444. [Google Scholar] [CrossRef] [PubMed]
  37. Kunsongkeit, P.; Okuma, N.; Rassameemasmaung, S.; Chaivanit, P. Effect of vitamin c as an adjunct in nonsurgical periodontal therapy in uncontrolled type 2 diabetes mellitus patients. Eur. J. Dent. 2019, 13, 444–449. [Google Scholar] [CrossRef] [Green Version]
  38. Gokhale, N.H.; Acharya, A.B.; Patil, V.S.; Trivedi, D.J.; Thakur, S.L. A short-term evaluation of the relationship between plasma ascorbic acid levels and periodontal disease in systemically healthy and type 2 diabetes mellitus subjects. J. Diet. Suppl. 2013, 10, 93–104. [Google Scholar] [CrossRef]
  39. Gadagi, J.S.; Chava, V.K.; Reddy, V.R. Green tea extract as a local drug therapy on periodontitis patients with diabetes mellitus: A randomized case-control study. J. Indian Soc. Periodontol. 2013, 17, 198–203. [Google Scholar]
  40. Catanzaro, D.P.; Mena Laura, E.E.; Cestari, T.M.; Arantes, R.V.N.; Garlet, G.P.; Taga, R.; Assis, G.F. Green tea prevents vascular disturbs and attenuates periodontal breakdown in long-term hyperglycaemia in t1d rats. J. Clin. Periodontol. 2018, 45, 557–569. [Google Scholar] [CrossRef]
  41. Gennaro, G.; Claudino, M.; Cestari, T.M.; Ceolin, D.; Germino, P.; Garlet, G.P.; de Assis, G.F. Green tea modulates cytokine expression in the periodontium and attenuates alveolar bone resorption in type 1 diabetic rats. PLoS ONE 2015, 10, e0134784. [Google Scholar] [CrossRef] [Green Version]
  42. Zare Javid, A.; Bazyar, H.; Gholinezhad, H.; Rahimlou, M.; Rashidi, H.; Salehi, P.; Haghighi-Zadeh, M.H. The effects of ginger supplementation on inflammatory, antioxidant, and periodontal parameters in type 2 diabetes mellitus patients with chronic periodontitis under non-surgical periodontal therapy. A double-blind, placebo-controlled trial. Diabetes Metab. Syndr. Obes. 2019, 12, 1751–1761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Pradeep, A.R.; Garg, V.; Raju, A.; Singh, P. Adjunctive local delivery of aloe vera gel in patients with type 2 diabetes and chronic periodontitis: A randomized, controlled clinical trial. J. Periodontol. 2016, 87, 268–274. [Google Scholar] [CrossRef] [PubMed]
  44. Hatipoglu, M.; Alptekin, N.; Avunduk, M.C. Effects of alpha-tocopherol on gingival expression of inducible nitric oxide synthase in the rats with experimental periodontitis and diabetes. Niger J. Clin. Pract. 2016, 19, 480–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Singh, V.P.; Bali, A.; Singh, N.; Jaggi, A.S. Advanced glycation end products and diabetic complications. Korean J. Physiol. Pharmacol. 2014, 18, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Negre-Salvayre, A.; Salvayre, R.; Augé, N.; Pamplona, R.; Portero-Otín, M. Hyperglycemia and glycation in diabetic complications. Antioxid. Redox Signal. 2009, 11, 3071–3109. [Google Scholar] [CrossRef] [PubMed]
  47. Preshaw, P.M.; Alba, A.L.; Herrera, D.; Jepsen, S.; Konstantinidis, A.; Makrilakis, K.; Taylor, R. Periodontitis and diabetes: A two-way relationship. Diabetologia 2012, 55, 21–31. [Google Scholar] [CrossRef] [Green Version]
  48. Taylor, G.W.; Burt, B.A.; Becker, M.P.; Genco, R.J.; Shlossman, M. Glycemic control and alveolar bone loss progression in type 2 diabetes. Ann. Periodontol. 1998, 3, 30–39. [Google Scholar] [CrossRef] [Green Version]
  49. Salvi, G.E.; Collins, J.G.; Yalda, B.; Arnold, R.R.; Lang, N.P.; Offenbacher, S. Monocytic tnf alpha secretion patterns in iddm patients with periodontal diseases. J. Clin. Periodontol. 1997, 24, 8–16. [Google Scholar] [CrossRef]
  50. Okazaki, R.; Totsuka, Y.; Hamano, K.; Ajima, M.; Miura, M.; Hirota, Y.; Hata, K.; Fukumoto, S.; Matsumoto, T. Metabolic improvement of poorly controlled noninsulin-dependent diabetes mellitus decreases bone turnover. J. Clin. Endocrinol. Metab. 1997, 82, 2915–2920. [Google Scholar] [CrossRef]
  51. Haskins, K.; Bradley, B.; Powers, K.; Fadok, V.; Flores, S.; Ling, X.; Pugazhenthi, S.; Reusch, J.; Kench, J. Oxidative stress in type 1 diabetes. Ann. N. Y. Acad. Sci. 2003, 1005, 43–54. [Google Scholar] [CrossRef]
  52. Takagi, M.; Kasayama, S.; Yamamoto, T.; Motomura, T.; Hashimoto, K.; Yamamoto, H.; Sato, B.; Okada, S.; Kishimoto, T. Advanced glycation endproducts stimulate interleukin-6 production by human bone-derived cells. J. Bone Miner. Res. 1997, 12, 439–446. [Google Scholar] [CrossRef] [PubMed]
  53. Mócsai, A. Diverse novel functions of neutrophils in immunity, inflammation, and beyond. J. Exp. Med. 2013, 210, 1283–1299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Naguib, G.; Al-Mashat, H.; Desta, T.; Graves, D.T. Diabetes prolongs the inflammatory response to a bacterial stimulus through cytokine dysregulation. J. Investig. Dermatol. 2004, 123, 87–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schmidt, A.M.; Yan, S.D.; Wautier, J.L.; Stern, D. Activation of receptor for advanced glycation end products: A mechanism for chronic vascular dysfunction in diabetic vasculopathy and atherosclerosis. Circ. Res. 1999, 84, 489–497. [Google Scholar] [CrossRef] [Green Version]
  56. Mayadas, T.N.; Cullere, X.; Lowell, C.A. The multifaceted functions of neutrophils. Annu. Rev. Pathol. 2014, 9, 181–218. [Google Scholar] [CrossRef] [Green Version]
  57. Delima, A.J.; Van Dyke, T.E. Origin and function of the cellular components in gingival crevice fluid. Periodontology 2000 2003, 31, 55–76. [Google Scholar] [CrossRef]
  58. Roomi, M.W.; Kalinovsky, T.; Rath, M.; Niedzwiecki, A. Modulation of mmp-2 and mmp-9 secretion by cytokines, inducers and inhibitors in human glioblastoma t-98g cells. Oncol. Rep. 2017, 37, 1907–1913. [Google Scholar] [CrossRef] [Green Version]
  59. Ohta, K.; Naruse, T.; Ishida, Y.; Shigeishi, H.; Nakagawa, T.; Fukui, A.; Nishi, H.; Sasaki, K.; Ogawa, I.; Takechi, M. Tnf-α-induced il-6 and mmp-9 expression in immortalized ameloblastoma cell line established by htert. Oral Dis. 2017, 23, 199–209. [Google Scholar] [CrossRef]
  60. Tiranathanagul, S.; Yongchaitrakul, T.; Pattamapun, K.; Pavasant, P. Actinobacillus actinomycetemcomitans lipopolysaccharide activates matrix metalloproteinase-2 and increases receptor activator of nuclear factor-kappab ligand expression in human periodontal ligament cells. J. Periodontol. 2004, 75, 1647–1654. [Google Scholar] [CrossRef]
  61. Wahl, L.M.; Corcoran, M.L. Regulation of monocyte/macrophage metalloproteinase production by cytokines. J. Periodontol. 1993, 64, 467–473. [Google Scholar]
  62. Ries, C.; Petrides, P.E. Cytokine regulation of matrix metalloproteinase activity and its regulatory dysfunction in disease. Biol. Chem. Hoppe Seyler 1995, 376, 345–355. [Google Scholar] [PubMed]
  63. Frankwich, K.; Tibble, C.; Torres-Gonzalez, M.; Bonner, M.; Lefkowitz, R.; Tyndall, M.; Schmid-Schönbein, G.W.; Villarreal, F.; Heller, M.; Herbst, K. Proof of concept: Matrix metalloproteinase inhibitor decreases inflammation and improves muscle insulin sensitivity in people with type 2 diabetes. J. Inflamm. 2012, 9, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Derosa, G.; Ferrari, I.; D’Angelo, A.; Tinelli, C.; Salvadeo, S.A.; Ciccarelli, L.; Piccinni, M.N.; Gravina, A.; Ramondetti, F.; Maffioli, P.; et al. Matrix metalloproteinase-2 and -9 levels in obese patients. Endothelium 2008, 15, 219–224. [Google Scholar] [CrossRef] [PubMed]
  65. Kanzaki, H.; Shinohara, F.; Kajiya, M.; Fukaya, S.; Miyamoto, Y.; Nakamura, Y. Nuclear nrf2 induction by protein transduction attenuates osteoclastogenesis. Free Radic. Biol. Med. 2014, 77, 239–248. [Google Scholar] [CrossRef] [Green Version]
  66. Patil, V.S.; Patil, V.P.; Gokhale, N.; Acharya, A.; Kangokar, P. Chronic periodontitis in type 2 diabetes mellitus: Oxidative stress as a common factor in periodontal tissue injury. J. Clin. Diagn. Res. 2016, 10, BC12-6. [Google Scholar] [CrossRef]
  67. Lambeth, J.D. Nox enzymes and the biology of reactive oxygen. Nat. Rev. Immunol. 2004, 4, 181–189. [Google Scholar] [CrossRef]
  68. Franchini, A.M.; Hunt, D.; Melendez, J.A.; Drake, J.R. Fcγr-driven release of il-6 by macrophages requiRes. nox2-dependent production of reactive oxygen species. J. Biol. Chem. 2013, 288, 25098–25108. [Google Scholar] [CrossRef] [Green Version]
  69. Sima, C.; Aboodi, G.M.; Lakschevitz, F.S.; Sun, C.; Goldberg, M.B.; Glogauer, M. Nuclear factor erythroid 2-related factor 2 down-regulation in oral neutrophils is associated with periodontal oxidative damage and severe chronic periodontitis. Am. J. Pathol. 2016, 186, 1417–1426. [Google Scholar] [CrossRef] [Green Version]
  70. Mealey, B.L.; Rose, L.F. Diabetes mellitus and inflammatory periodontal diseases. Curr. Opin. Endocrinol. Diabetes Obes. 2008, 15, 135–141. [Google Scholar] [CrossRef]
  71. Sbordone, L.; Ramaglia, L.; Barone, A.; Ciaglia, R.N.; Iacono, V.J. Periodontal status and subgingival microbiota of insulin-dependent juvenile diabetics: A 3-year longitudinal study. J. Periodontol. 1998, 69, 120–128. [Google Scholar] [CrossRef]
  72. Taylor, G.W.; Burt, B.A.; Becker, M.P.; Genco, R.J.; Shlossman, M.; Knowler, W.C.; Pettitt, D.J. Severe periodontitis and risk for poor glycemic control in patients with non-insulin-dependent diabetes mellitus. J. Periodontol. 1996, 67, 1085–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Vallianou, N.G.; Evangelopoulos, A.; Kazazis, C. Resveratrol and diabetes. Rev. Diabet. Stud. 2013, 10, 236–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Brasnyó, P.; Molnár, G.A.; Mohás, M.; Markó, L.; Laczy, B.; Cseh, J.; Mikolás, E.; Szijártó, I.A.; Mérei, A.; Halmai, R.; et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the akt pathway in type 2 diabetic patients. Br. J. Nutr. 2011, 106, 383–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Park, H.J.; Jeong, S.K.; Kim, S.R.; Bae, S.K.; Kim, W.S.; Jin, S.D.; Koo, T.H.; Jang, H.O.; Yun, I.; Kim, K.W.; et al. Resveratrol inhibits porphyromonas gingivalis lipopolysaccharide-induced endothelial adhesion molecule expression by suppressing nf-kappab activation. Arch. Pharm. Res. 2009, 32, 583–591. [Google Scholar] [CrossRef] [PubMed]
  76. Lv, J.S.; Jiang, X.W.; Zhang, Y.; Zhen, L. Effect of resveratrol on expression of tlr4 and inflammatory factors in gingival epithelial cells under high glucose environment. Shanghai Kou Qiang Yi Xue 2017, 26, 32–36. [Google Scholar] [PubMed]
  77. Becerik, S.; Ozsan, N.; Gürkan, A.; Oztürk, V.; Atilla, G.; Emingil, G. Toll like receptor 4 and membrane-bound cd14 expressions in gingivitis, periodontitis and csa-induced gingival overgrowth. Arch. Oral. Biol. 2011, 56, 456–465. [Google Scholar] [CrossRef]
  78. Promsudthi, A.; Poomsawat, S.; Limsricharoen, W. The role of toll-like receptor 2 and 4 in gingival tissues of chronic periodontitis subjects with type 2 diabetes. J. Periodontal Res. 2014, 49, 346–354. [Google Scholar] [CrossRef]
  79. Yang, X.; Zhang, J.; Ni, J.; Ouyang, B.; Wang, D.; Luo, S.; Xie, B.; Xuan, D. Toll-like receptor 4-mediated hyper-responsiveness of gingival epithelial cells to lipopolysaccharide in high-glucose environments. J. Periodontol. 2014, 85, 1620–1628. [Google Scholar] [CrossRef]
  80. Jakus, P.B.; Kalman, N.; Antus, C.; Radnai, B.; Tucsek, Z.; Gallyas, F., Jr.; Sumegi, B.; Veres, B. Traf6 is functional in inhibition of tlr4-mediated nf-κb activation by resveratrol. J. Nutr. Biochem. 2013, 24, 819–823. [Google Scholar] [CrossRef]
  81. Cirano, F.R.; Casarin, R.C.; Ribeiro, F.V.; Casati, M.Z.; Pimentel, S.P.; Taiete, T.; Bernardi, M.M. Effect of resveratrol on periodontal pathogens during experimental periodontitis in rats. Braz. Oral Res. 2016, 30, e128. [Google Scholar] [CrossRef] [Green Version]
  82. He, T.; Guan, X.; Wang, S.; Xiao, T.; Yang, K.; Xu, X.; Wang, J.; Zhao, J. Resveratrol prevents high glucose-induced epithelial-mesenchymal transition in renal tubular epithelial cells by inhibiting nadph oxidase/ros/erk pathway. Mol. Cell. Endocrinol. 2015, 402, 13–20. [Google Scholar] [CrossRef] [PubMed]
  83. Singh, V.; Ubaid, S. Role of silent information regulator 1 (sirt1) in regulating oxidative stress and inflammation. Inflammation 2020, 43, 1589–1598. [Google Scholar] [CrossRef] [PubMed]
  84. Fang, W.J.; Wang, C.J.; He, Y.; Zhou, Y.L.; Peng, X.D.; Liu, S.K. Resveratrol alleviates diabetic cardiomyopathy in rats by improving mitochondrial function through pgc-1α deacetylation. Acta Pharmacol. Sin. 2018, 39, 59–73. [Google Scholar] [CrossRef] [PubMed]
  85. Rivera, L.; Morón, R.; Zarzuelo, A.; Galisteo, M. Long-term resveratrol administration reduces metabolic disturbances and lowers blood pressure in obese zucker rats. Biochem. Pharmacol. 2009, 77, 1053–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Guzik, T.J.; Mussa, S.; Gastaldi, D.; Sadowski, J.; Ratnatunga, C.; Pillai, R.; Channon, K.M. Mechanisms of increased vascular superoxide production in human diabetes mellitus: Role of nad(p)h oxidase and endothelial nitric oxide synthase. Circulation 2002, 105, 1656–1662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Yeh, T.C.; Shin, C.S.; Chen, H.H.; Lai, C.C.; Sun, G.C.; Tseng, C.J.; Cheng, P.W. Resveratrol regulates blood pressure by enhancing ampk signaling to downregulate a rac1-derived nadph oxidase in the central nervous system. J. Appl. Physiol. 2018, 125, 40–48. [Google Scholar] [CrossRef] [PubMed]
  88. Tamaki, N.; Cristina Orihuela-Campos, R.; Inagaki, Y.; Fukui, M.; Nagata, T.; Ito, H.O. 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. 2014, 75, 222–229. [Google Scholar] [CrossRef]
  89. Lozano-Pérez, A.A.; Rivero, H.C.; Pérez Hernández, M.D.C.; Pagán, A.; Montalbán, M.G.; Víllora, G.; Cénis, J.L. Silk fibroin nanoparticles: Efficient vehicles for the natural antioxidant quercetin. Int. J. Pharm. 2017, 518, 11–19. [Google Scholar] [CrossRef]
  90. Lozano-Pérez, A.A.; Gil, A.L.; Pérez, S.A.; Cutillas, N.; Meyer, H.; Pedreño, M.; Aznar-Cervantes, S.D.; Janiak, C.; Cenis, J.L.; Ruiz, J. Antitumor properties of platinum(iv) prodrug-loaded silk fibroin nanoparticles. Dalton Trans. 2015, 44, 13513–13521. [Google Scholar] [CrossRef]
  91. Lozano-Pérez, A.A.; Rodriguez-Nogales, A.; Ortiz-Cullera, V.; Algieri, F.; Garrido-Mesa, J.; Zorrilla, P.; Rodriguez-Cabezas, M.E.; Garrido-Mesa, N.; Utrilla, M.P.; De Matteis, L.; et al. Silk fibroin nanoparticles constitute a vector for controlled release of resveratrol in an experimental model of inflammatory bowel disease in rats. Int. J. Nanomed. 2014, 9, 4507–4520. [Google Scholar]
  92. Corrêa, M.G.; Pires, P.R.; Ribeiro, F.V.; Pimentel, S.Z.; Casarin, R.C.; Cirano, F.R.; Tenenbaum, H.T.; Casati, M.Z. Systemic treatment with resveratrol and/or curcumin reduces the progression of experimental periodontitis in rats. J. Periodontal Res. 2017, 52, 201–209. [Google Scholar] [CrossRef] [PubMed]
  93. Movahed, A.; Nabipour, I.; Lieben Louis, X.; Thandapilly, S.J.; Yu, L.; Kalantarhormozi, M.; Rekabpour, S.J.; Netticadan, T. Antihyperglycemic effects of short term resveratrol supplementation in type 2 diabetic patients. Evid. Based Complement. Alternat. Med. 2013, 2013, 851267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Bhatt, J.K.; Thomas, S.; Nanjan, M.J. Resveratrol supplementation improves glycemic control in type 2 diabetes mellitus. Nutr. Res. 2012, 32, 537–541. [Google Scholar] [CrossRef] [PubMed]
  95. Apostolidou, C.; Adamopoulos, K.; Iliadis, S.; Kourtidou-Papadeli, C. Alterations of antioxidant status in asymptomatic hypercholesterolemic individuals after resveratrol intake. Int. J. Food Sci. Nutr. 2015, 67, 541–552. [Google Scholar] [CrossRef] [PubMed]
  96. Ghanim, H.; Sia, C.L.; Korzeniewski, K.; Lohano, T.; Abuaysheh, S.; Marumganti, A.; Chaudhuri, A.; Dandona, P. A resveratrol and polyphenol preparation suppresses oxidative and inflammatory stress response to a high-fat, high-carbohydrate meal. J. Clin. Endocrinol. Metab. 2011, 96, 1409–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Jagetia, G.C.; Aggarwal, B.B. “Spicing up” of the immune system by curcumin. J. Clin. Immunol. 2007, 27, 19–35. [Google Scholar] [CrossRef]
  98. Chougala, M.B.; Bhaskar, J.J.; Rajan, M.G.; Salimath, P.V. Effect of curcumin and quercetin on lysosomal enzyme activities in streptozotocin-induced diabetic rats. Clin. Nutr. 2012, 31, 749–755. [Google Scholar] [CrossRef]
  99. Sylvester, J.; Liacini, A.; Li, W.Q.; Dehnade, F.; Zafarullah, M. Tripterygium wilfordii hook f extract suppresses proinflammatory cytokine-induced expression of matrix metalloproteinase genes in articular chondrocytes by inhibiting activating protein-1 and nuclear factor-kappab activities. Mol. Pharmacol. 2001, 59, 1196–1205. [Google Scholar] [CrossRef] [Green Version]
  100. Mengshol, J.A.; Vincenti, M.P.; Coon, C.I.; Barchowsky, A.; Brinckerhoff, C.E. Interleukin-1 induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requiRes. p38, c-jun n-terminal kinase, and nuclear factor kappab: Differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum. 2000, 43, 801–811. [Google Scholar] [CrossRef]
  101. Sakai, T.; Kambe, F.; Mitsuyama, H.; Ishiguro, N.; Kurokouchi, K.; Takigawa, M.; Iwata, H.; Seo, H. Tumor necrosis factor alpha induces expression of genes for matrix degradation in human chondrocyte-like hcs-2/8 cells through activation of nf-kappab: Abrogation of the tumor necrosis factor alpha effect by proteasome inhibitors. J. Bone Miner. Res. 2001, 16, 1272–1280. [Google Scholar] [CrossRef]
  102. Liacini, A.; Sylvester, J.; Li, W.Q.; Zafarullah, M. Inhibition of interleukin-1-stimulated map kinases, activating protein-1 (ap-1) and nuclear factor kappa b (nf-kappa b) transcription factors down-regulates matrix metalloproteinase gene expression in articular chondrocytes. Matrix Biol. 2002, 21, 251–262. [Google Scholar] [CrossRef]
  103. Deng, J.; Gu, Y.; Lee, H.; Raja, V.; Johnson, F.; Golub, L.J.J.D.R. Novel modified-curcumin: Resolution of cytokines and mmps in cell culture. In Proceedings of the 2018 AADR/CADR Annual Meeting, Fort Lauderdale, FL, USA, 21–24 March 2018; p. 129. [Google Scholar]
  104. Yin, Y.; Chen, F.; Wang, W.; Wang, H.; Zhang, X. Resolvin d1 inhibits inflammatory response in stz-induced diabetic retinopathy rats: Possible involvement of nlrp3 inflammasome and nf-κb signaling pathway. Mol. Vis. 2017, 23, 242–250. [Google Scholar] [PubMed]
  105. Gounden, S.; Chuturgoon, A. Curcumin upregulates antioxidant defense, lon protease, and heat-shock protein 70 under hyperglycemic conditions in human hepatoma cells. J. Med. Food 2017, 20, 465–473. [Google Scholar] [CrossRef] [PubMed]
  106. Milne, J.C.; Lambert, P.D.; Schenk, S.; Carney, D.P.; Smith, J.J.; Gagne, D.J.; Jin, L.; Boss, O.; Perni, R.B.; Vu, C.B.; et al. Small molecule activators of sirt1 as therapeutics for the treatment of type 2 diabetes. Nature 2007, 450, 712–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Zhou, T.; Chen, D.; Li, Q.; Sun, X.; Song, Y.; Wang, C. Curcumin inhibits inflammatory response and bone loss during experimental periodontitis in rats. Acta Odontol. Scand. 2013, 71, 349–356. [Google Scholar] [CrossRef] [PubMed]
  108. Tomofuji, T.; Ekuni, D.; Irie, K.; Azuma, T.; Endo, Y.; Tamaki, N.; Sanbe, T.; Murakami, J.; Yamamoto, T.; Morita, M. Preventive effects of a cocoa-enriched diet on gingival oxidative stress in experimental periodontitis. J. Periodontol. 2009, 80, 1799–1808. [Google Scholar] [CrossRef]
  109. De Araújo Júnior, R.F.; Souza, T.O.; de Moura, L.M.; Torres, K.P.; de Souza, L.B.; Alves Mdo, S.; Rocha, H.O.; de Araújo, A.A. Atorvastatin decreases bone loss, inflammation and oxidative stress in experimental periodontitis. PLoS ONE 2013, 8, e75322. [Google Scholar]
  110. Stehle, J.H.; Saade, A.; Rawashdeh, O.; Ackermann, K.; Jilg, A.; Sebestény, T.; Maronde, E. A survey of molecular details in the human pineal gland in the light of phylogeny, structure, function and chronobiological diseases. J. Pineal Res. 2011, 51, 17–43. [Google Scholar] [CrossRef]
  111. Murch, S.J.; Simmons, C.B.; Saxena, P.K. Melatonin in feverfew and other medicinal plants. Lancet 1997, 350, 1598–1599. [Google Scholar] [CrossRef]
  112. Khalil, M.; Power, N.; Graham, E.; Deschenes, S.S.; Schmitz, N. The association between sleep and diabetes outcomes—A systematic review. Diabetes Res. Clin. Pract. 2020, 161, 108035. [Google Scholar] [CrossRef]
  113. Peivandi, S.; Habibi, A.; Hosseini, S.H.; Khademloo, M.; Raisian, M.; Pournorouz, H. Evaluation of sleep quality and duration in pregnancy and risk of gestational diabetes mellitus: A prospective follow-up study. Biomedicine 2021, 11, 24–29. [Google Scholar] [CrossRef]
  114. Galano, A.; Tan, D.X.; Reiter, R.J. Melatonin as a natural ally against oxidative stress: A physicochemical examination. J. Pineal Res. 2011, 51, 1–16. [Google Scholar] [CrossRef] [PubMed]
  115. Poeggeler, B.; Saarela, S.; Reiter, R.J.; Tan, D.X.; Chen, L.D.; Manchester, L.C.; Barlow-Walden, L.R. Melatonin—A highly potent endogenous radical scavenger and electron donor: New aspects of the oxidation chemistry of this indole accessed in vitro. Ann. N. Y. Acad. Sci. 1994, 738, 419–420. [Google Scholar] [CrossRef] [PubMed]
  116. Reiter, R.J.; Tan, D.X.; Manchester, L.C.; Qi, W. Biochemical reactivity of melatonin with reactive oxygen and nitrogen species: A review of the evidence. Cell Biochem. Biophys. 2001, 34, 237–256. [Google Scholar] [CrossRef]
  117. Reiter, R.J.; Calvo, J.R.; Karbownik, M.; Qi, W.; Tan, D.X. Melatonin and its relation to the immune system and inflammation. Ann. N. Y. Acad. Sci. 2000, 917, 376–386. [Google Scholar] [CrossRef] [PubMed]
  118. Radogna, F.; Diederich, M.; Ghibelli, L. Melatonin: A pleiotropic molecule regulating inflammation. Biochem. Pharmacol. 2010, 80, 1844–1852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Koyama, H.; Nakade, O.; Takada, Y.; Kaku, T.; Lau, K.H. Melatonin at pharmacologic doses increases bone mass by suppressing resorption through down-regulation of the rankl-mediated osteoclast formation and activation. J. Bone Miner. Res. 2002, 17, 1219–1229. [Google Scholar] [CrossRef]
  120. Ostrowska, Z.; Ziora, K.; Kos-Kudła, B.; Swietochowska, E.; Oświecimska, J.; Dyduch, A.; Wołkowska-Pokrywa, K.; Szapska, B. Melatonin, the rankl/rank/opg system, and bone metabolism in girls with anorexia nervosa. Endokrynol. Pol. 2010, 61, 117–123. [Google Scholar]
  121. Satomura, K.; Tobiume, S.; Tokuyama, R.; Yamasaki, Y.; Kudoh, K.; Maeda, E.; Nagayama, M. Melatonin at pharmacological doses enhances human osteoblastic differentiation in vitro and promotes mouse cortical bone formation in vivo. J. Pineal Res. 2007, 42, 231–239. [Google Scholar] [CrossRef]
  122. Liu, D.; Xu, J.K.; Figliomeni, L.; Huang, L.; Pavlos, N.J.; Rogers, M.; Tan, A.; Price, P.; Zheng, M.H. Expression of rankl and opg mrna in periodontal disease: Possible involvement in bone destruction. Int. J. Mol. Med. 2003, 11, 17–21. [Google Scholar] [CrossRef]
  123. Crotti, T.; Smith, M.D.; Hirsch, R.; Soukoulis, S.; Weedon, H.; Capone, M.; Ahern, M.J.; Haynes, D. Receptor activator nf kappab ligand (rankl) and osteoprotegerin (opg) protein expression in periodontitis. J. Periodontal Res. 2003, 38, 380–387. [Google Scholar] [CrossRef] [PubMed]
  124. Naderi-Behdani, F.; Heydari, F.; Ala, S.; Moradi, S.; Abediankenari, S.; Asgarirad, H.; Khodabakhsh, E. Effect of melatonin on stress-induced hyperglycemia and insulin resistance in critically-ill patients: A randomized double-blind, placebo-controlled clinical trial. Casp. J. Intern. Med. 2022, 13, 51–60. [Google Scholar]
  125. Rubio-Sastre, P.; Scheer, F.A.; Gómez-Abellán, P.; Madrid, J.A.; Garaulet, M. Acute melatonin administration in humans impairs glucose tolerance in both the morning and evening. Sleep 2014, 37, 1715–1719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Kampmann, U.; Lauritzen, E.S.; Grarup, N.; Jessen, N.; Hansen, T.; Møller, N.; Støy, J. Acute metabolic effects of melatonin-a randomized crossover study in healthy young men. J. Pineal Res. 2021, 70, e12706. [Google Scholar] [CrossRef]
  127. Cagnacci, A.; Arangino, S.; Renzi, A.; Paoletti, A.M.; Melis, G.B.; Cagnacci, P.; Volpe, A. Influence of melatonin administration on glucose tolerance and insulin sensitivity of postmenopausal women. Clin. Endocrinol. 2001, 54, 339–346. [Google Scholar] [CrossRef]
  128. Rybka, J.; Kędziora-Kornatowska, K.; Kupczyk, D.; Muszalik, M.; Kornatowski, M.; Kędziora, J. Antioxidant effect of immediate- versus sustained-release melatonin in type 2 diabetes mellitus and healthy controls. Drug Deliv. 2016, 23, 814–817. [Google Scholar] [CrossRef]
  129. Ramch; Padalkar, R.K.; Shinde, A.V.; Patil, S.M. Lipid profile, serum malondialdehyde, superoxide dismutase in chronic kidney diseases and type 2 diabetes mellitus. Biomed. Res. 2012, 23, 207–210. [Google Scholar]
  130. Rodriguez, C.; Mayo, J.C.; Sainz, R.M.; Antolín, I.; Herrera, F.; Martín, V.; Reiter, R.J. Regulation of antioxidant enzymes: A significant role for melatonin. J. Pineal Res. 2004, 36, 1–9. [Google Scholar] [CrossRef]
  131. Prado, N.J.; Ferder, L.; Manucha, W.; Diez, E.R. Anti-inflammatory effects of melatonin in obesity and hypertension. Curr. Hypertens. Rep. 2018, 20, 45. [Google Scholar] [CrossRef]
  132. Lauritzen, E.S.; Kampmann, U.; Smedegaard, S.B.; Støy, J. Effects of daily administration of melatonin before bedtime on fasting insulin, glucose and insulin sensitivity in healthy adults and patients with metabolic diseases. A systematic review and meta-analysis. Clin. Endocrinol. 2021, 95, 691–701. [Google Scholar] [CrossRef]
  133. Pierce, M.; Linnebur, S.A.; Pearson, S.M.; Fixen, D.R. Optimal melatonin dose in older adults: A clinical review of the literature. Sr. Care Pharm. 2019, 34, 419–431. [Google Scholar] [CrossRef] [PubMed]
  134. Velazquez, C.; Navarro, M.; Acosta, A.; Angulo, A.; Dominguez, Z.; Robles, R.; Robles-Zepeda, R.; Lugo, E.; Goycoolea, F.M.; Velazquez, E.F.; et al. Antibacterial and free-radical scavenging activities of sonoran propolis. J. Appl. Microbiol. 2007, 103, 1747–1756. [Google Scholar] [CrossRef] [PubMed]
  135. Russo, A.; Longo, R.; Vanella, A. Antioxidant activity of propolis: Role of caffeic acid phenethyl ester and galangin. Fitoterapia 2002, 73 (Suppl. 1), S21–S29. [Google Scholar] [CrossRef]
  136. Kishimoto, N.; Kakino, Y.; Iwai, K.; Mochida, K.; Fujita, T. In vitro antibacterial, antimutagenic and anti-influenza virus activity of caffeic acid phenethyl esters. Biocontrol. Sci. 2005, 10, 155–161. [Google Scholar] [CrossRef] [Green Version]
  137. Nna, V.U.; Abu Bakar, A.B.; Md Lazin, M.; Mohamed, M. Antioxidant, anti-inflammatory and synergistic anti-hyperglycemic effects of malaysian propolis and metformin in streptozotocin-induced diabetic rats. Food Chem. Toxicol. 2018, 120, 305–320. [Google Scholar] [CrossRef]
  138. Orban, Z.; Mitsiades, N.; Burke, T.R., Jr.; Tsokos, M.; Chrousos, G.P. Caffeic acid phenethyl ester induces leukocyte apoptosis, modulates nuclear factor-kappa b and suppresses acute inflammation. Neuroimmunomodulation 2000, 7, 99–105. [Google Scholar] [CrossRef] [Green Version]
  139. Koltuksuz, U.; Ozen, S.; Uz, E.; Aydinç, M.; Karaman, A.; Gültek, A.; Akyol, O.; Gürsoy, M.H.; Aydin, E. Caffeic acid phenethyl ester prevents intestinal reperfusion injury in rats. J. Pediatr. Surg. 1999, 34, 1458–1462. [Google Scholar] [CrossRef]
  140. Santos, V.R.; Lima, J.A.; Gonçalves, T.E.; Bastos, M.F.; Figueiredo, L.C.; Shibli, J.A.; Duarte, P.M. Receptor activator of nuclear factor-kappa b ligand/osteoprotegerin ratio in sites of chronic periodontitis of subjects with poorly and well-controlled type 2 diabetes. J. Periodontol. 2010, 81, 1455–1465. [Google Scholar] [CrossRef]
  141. Araújo, A.A.; Souza, T.O.; Moura, L.M.; Brito, G.A.; Aragão, K.S.; Araújo, L.S.; Medeiros, C.A.; Alves, M.S.; Araújo, R.F., Jr. Effect of telmisartan on levels of il-1, tnf-α, down-regulated cox-2, mmp-2, mmp-9 and rankl/rank in an experimental periodontitis model. J. Clin. Periodontol. 2013, 40, 1104–1111. [Google Scholar] [CrossRef] [Green Version]
  142. Ha, J.; Choi, H.S.; Lee, Y.; Lee, Z.H.; Kim, H.H. Caffeic acid phenethyl ester inhibits osteoclastogenesis by suppressing nf kappab and downregulating nfatc1 and c-fos. Int. Immunopharmacol. 2009, 9, 774–780. [Google Scholar] [CrossRef]
  143. Vo, T.T.T.; Lee, C.W.; Chiang, Y.C.; Chen, Y.W.; Yu, Y.H.; Tuan, V.P.; Wu, C.Z.; Lee, I.T. Protective mechanisms of taiwanese green propolis toward high glucose-induced inflammation via nlrp3 inflammasome signaling pathway in human gingival fibroblasts. J. Periodontal Res. 2021, 56, 804–818. [Google Scholar] [CrossRef] [PubMed]
  144. Hsieh, C.Y.; Li, L.H.; Rao, Y.K.; Ju, T.C.; Nai, Y.S.; Chen, Y.W.; Hua, K.F. Mechanistic insight into the attenuation of gouty inflammation by taiwanese green propolis via inhibition of the nlrp3 inflammasome. J. Cell Physiol. 2019, 234, 4081–4094. [Google Scholar] [CrossRef] [PubMed]
  145. Cao, X. Self-regulation and cross-regulation of pattern-recognition receptor signalling in health and disease. Nat. Rev. Immunol. 2016, 16, 35–50. [Google Scholar] [CrossRef] [PubMed]
  146. Bauernfeind, F.G.; Horvath, G.; Stutz, A.; Alnemri, E.S.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B.G.; Fitzgerald, K.A.; et al. Cutting edge: Nf-kappab activating pattern recognition and cytokine receptors license nlrp3 inflammasome activation by regulating nlrp3 expression. J. Immunol. 2009, 183, 787–791. [Google Scholar] [CrossRef]
  147. Ceriello, A. Oxidative stress and glycemic regulation. Metabolism 2000, 49, 27–29. [Google Scholar] [CrossRef]
  148. Mohammed, B.M.; Fisher, B.J.; Kraskauskas, D.; Ward, S.; Wayne, J.S.; Brophy, D.F.; Fowler, A.A., 3rd; Yager, D.R.; Natarajan, R. Vitamin c promotes wound healing through novel pleiotropic mechanisms. Int. Wound J. 2016, 13, 572–584. [Google Scholar] [CrossRef]
  149. Chapple, I.L. Potential mechanisms underpinning the nutritional modulation of periodontal inflammation. J. Am. Dent. Assoc. 2009, 140, 178–184. [Google Scholar] [CrossRef] [Green Version]
  150. De Morais, E.F.; Pinheiro, J.C.; Leite, R.B.; Santos, P.P.A.; Barboza, C.A.G.; Freitas, R.A. Matrix metalloproteinase-8 levels in periodontal disease patients: A systematic review. J. Periodontal Res. 2018, 53, 156–163. [Google Scholar] [CrossRef]
  151. Hantke, B.; Lahmann, C.; Venzke, K.; Fischer, T.; Kocourek, A.; Windsor, L.J.; Bergemann, J.; Stäb, F.; Tschesche, H. Influence of flavonoids and vitamins on the mmp- and timp-expression of human dermal fibroblasts after uva irradiation. Photochem. Photobiol. Sci 2002, 1, 826–833. [Google Scholar] [CrossRef]
  152. Borutta, A. Vitamin c intake and periodontal disease. Br. Dent. J. 2005, 199, 210. [Google Scholar] [CrossRef] [Green Version]
  153. Mizuno, H.; Ekuni, D.; Maruyama, T.; Kataoka, K.; Yoneda, T.; Fukuhara, D.; Sugiura, Y.; Tomofuji, T.; Wada, J.; Morita, M. The effects of non-surgical periodontal treatment on glycemic control, oxidative stress balance and quality of life in patients with type 2 diabetes: A randomized clinical trial. PLoS ONE 2017, 12, e0188171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Afkhami-Ardekani, M.; Shojaoddiny-Ardekani, A. Effect of vitamin c on blood glucose, serum lipids & serum insulin in type 2 diabetes patients. Indian J. Med. Res. 2007, 126, 471–474. [Google Scholar] [PubMed]
  155. Sinclair, A.J.; Taylor, P.B.; Lunec, J.; Girling, A.J.; Barnett, A.H. Low plasma ascorbate levels in patients with type 2 diabetes mellitus consuming adequate dietary vitamin c. Diabet. Med. 1994, 11, 893–898. [Google Scholar] [CrossRef] [PubMed]
  156. Yussif, N.; Aziz, M.; Reda, A. Evaluation of the anti-inflammatory effect of locally delivered vitamin c in the treatment of persistent gingival inflammation: Clinical and histopathological study. J. Nutr. Metab. 2016, 2016, 2978741. [Google Scholar] [CrossRef] [Green Version]
  157. Higdon, J.V.; Frei, B. Tea catechins and polyphenols: Health effects, metabolism, and antioxidant functions. Crit. Rev. Food Sci. Nutr. 2003, 43, 89–143. [Google Scholar] [CrossRef]
  158. Inaba, H.; Tagashira, M.; Honma, D.; Kanda, T.; Kou, Y.; Ohtake, Y.; Amano, A. Identification of hop polyphenolic components which inhibit prostaglandin e2 production by gingival epithelial cells stimulated with periodontal pathogen. Biol. Pharm. Bull. 2008, 31, 527–530. [Google Scholar] [CrossRef] [Green Version]
  159. Miura, T.; Koike, T.; Ishida, T. Antidiabetic activity of green tea (Thea sinensis L.) in genetically type 2 diabetic mice. J. Health Sci. 2005, 51, 708–710. [Google Scholar] [CrossRef] [Green Version]
  160. Lee, M.J.; Lambert, J.D.; Prabhu, S.; Meng, X.; Lu, H.; Maliakal, P.; Ho, C.T.; Yang, C.S. Delivery of tea polyphenols to the oral cavity by green tea leaves and black tea extract. Cancer Epidemiol. Biomark. Prev. 2004, 13, 132–137. [Google Scholar] [CrossRef] [Green Version]
  161. Hirasawa, M.; Takada, K.; Makimura, M.; Otake, S. Improvement of periodontal status by green tea catechin using a local delivery system: A clinical pilot study. J. Periodontal Res. 2002, 37, 433–438. [Google Scholar] [CrossRef]
  162. Yang, F.; de Villiers, W.J.; McClain, C.J.; Varilek, G.W. Green tea polyphenols block endotoxin-induced tumor necrosis factor-production and lethality in a murine model. J. Nutr. 1998, 128, 2334–2340. [Google Scholar] [CrossRef]
  163. Rogers, J.; Perkins, I.; van Olphen, A.; Burdash, N.; Klein, T.W.; Friedman, H. Epigallocatechin gallate modulates cytokine production by bone marrow-derived dendritic cells stimulated with lipopolysaccharide or muramyldipeptide, or infected with legionella pneumophila. Exp. Biol. Med. 2005, 230, 645–651. [Google Scholar] [CrossRef] [PubMed]
  164. Ishida, I.; Kohda, C.; Yanagawa, Y.; Miyaoka, H.; Shimamura, T. Epigallocatechin gallate suppresses expression of receptor activator of nf-kappab ligand (rankl) in staphylococcus aureus infection in osteoblast-like nrg cells. J. Med. Microbiol. 2007, 56, 1042–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Claudino, M.; Garlet, T.P.; Cardoso, C.R.; de Assis, G.F.; Taga, R.; Cunha, F.Q.; Silva, J.S.; Garlet, G.P. Down-regulation of expression of osteoblast and osteocyte markers in periodontal tissues associated with the spontaneous alveolar bone loss of interleukin-10 knockout mice. Eur. J. Oral Sci. 2010, 118, 19–28. [Google Scholar] [CrossRef] [PubMed]
  166. Liu, R.; Bal, H.S.; Desta, T.; Krothapalli, N.; Alyassi, M.; Luan, Q.; Graves, D.T. Diabetes enhances periodontal bone loss through enhanced resorption and diminished bone formation. J. Dent. Res. 2006, 85, 510–514. [Google Scholar] [CrossRef] [PubMed]
  167. Nitta, H.; Katagiri, S.; Nagasawa, T.; Izumi, Y.; Ishikawa, I.; Izumiyama, H.; Uchimura, I.; Kanazawa, M.; Chiba, H.; Matsuo, A.; et al. The number of microvascular complications is associated with an increased risk for severity of periodontitis in type 2 diabetes patients: Results of a multicenter hospital-based cross-sectional study. J. Diabetes Investig. 2017, 8, 677–686. [Google Scholar] [CrossRef] [PubMed]
  168. Chiarelli, F.; Spagnoli, A.; Basciani, F.; Tumini, S.; Mezzetti, A.; Cipollone, F.; Cuccurullo, F.; Morgese, G.; Verrotti, A. Vascular endothelial growth factor (vegf) in children, adolescents and young adults with type 1 diabetes mellitus: Relation to glycaemic control and microvascular complications. Diabet. Med. 2000, 17, 650–656. [Google Scholar] [CrossRef]
  169. Seckin, D.; Ilhan, N.; Ilhan, N.; Ertugrul, S. Glycaemic control, markers of endothelial cell activation and oxidative stress in children with type 1 diabetes mellitus. Diabetes Res. Clin. Pract. 2006, 73, 191–197. [Google Scholar] [CrossRef]
  170. Shukla, Y.; Singh, M. Cancer preventive properties of ginger: A brief review. Food Chem. Toxicol 2007, 45, 683–690. [Google Scholar] [CrossRef]
  171. Semwal, R.B.; Semwal, D.K.; Combrinck, S.; Viljoen, A.M. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry 2015, 117, 554–568. [Google Scholar] [CrossRef]
  172. Rehman, M.U.; Rashid, S.M.; Rasool, S.; Shakeel, S.; Ahmad, B.; Ahmad, S.B.; Madkhali, H.; Ganaie, M.A.; Majid, S.; Bhat, S.A. Zingerone (4-(4-hydroxy-3-methylphenyl)butan-2-one) ameliorates renal function via controlling oxidative burst and inflammation in experimental diabetic nephropathy. Arch. Physiol. Biochem. 2019, 125, 201–209. [Google Scholar] [CrossRef]
  173. Kota, N.; Panpatil, V.V.; Kaleb, R.; Varanasi, B.; Polasa, K. Dose-dependent effect in the inhibition of oxidative stress and anticlastogenic potential of ginger in stz induced diabetic rats. Food Chem. 2012, 135, 2954–2959. [Google Scholar] [CrossRef] [PubMed]
  174. Al Hroob, A.M.; Abukhalil, M.H.; Alghonmeen, R.D.; Mahmoud, A.M. Ginger alleviates hyperglycemia-induced oxidative stress, inflammation and apoptosis and protects rats against diabetic nephropathy. Biomed. Pharmacother. 2018, 106, 381–389. [Google Scholar] [CrossRef] [PubMed]
  175. Nonaka, K.; Bando, M.; Sakamoto, E.; Inagaki, Y.; Naruishi, K.; Yumoto, H.; Kido, J.I. 6-shogaol inhibits advanced glycation end-products-induced il-6 and icam-1 expression by regulating oxidative responses in human gingival fibroblasts. Molecules 2019, 24, 3705. [Google Scholar] [CrossRef] [Green Version]
  176. Dugasani, S.; Pichika, M.R.; Nadarajah, V.D.; Balijepalli, M.K.; Tandra, S.; Korlakunta, J.N. Comparative antioxidant and anti-inflammatory effects of [6]-gingerol, [8]-gingerol, [10]-gingerol and [6]-shogaol. J. Ethnopharmacol. 2010, 127, 515–520. [Google Scholar] [CrossRef] [PubMed]
  177. Kim, J.K.; Jang, H.D. 6-shogaol attenuates h(2)o(2)-induced oxidative stress via upregulation of nrf2-mediated γ-glutamylcysteine synthetase and heme oxygenase expression in hepg2 cells. Food Sci. Biotechnol. 2016, 25, 319–327. [Google Scholar] [CrossRef]
  178. Zhao, D.; Gu, M.Y.; Xu, J.L.; Zhang, L.J.; Ryu, S.Y.; Yang, H.O. Anti-neuroinflammatory effects of 12-dehydrogingerdione in lps-activated microglia through inhibiting akt/ikk/nf-κb pathway and activating nrf-2/ho-1 pathway. Biomol. Ther. 2019, 27, 92–100. [Google Scholar] [CrossRef]
  179. Bak, M.J.; Ok, S.; Jun, M.; Jeong, W.S. 6-shogaol-rich extract from ginger up-regulates the antioxidant defense systems in cells and mice. Molecules 2012, 17, 8037–8055. [Google Scholar] [CrossRef]
  180. Davis, R.H.; Rosenthal, K.Y.; Cesario, L.R.; Rouw, G.A. Processed aloe vera administered topically inhibits inflammation. J. Am. Podiatr. Med. Assoc. 1989, 79, 395–397. [Google Scholar]
  181. Yagi, A.; Kabash, A.; Okamura, N.; Haraguchi, H.; Moustafa, S.M.; Khalifa, T.I. Antioxidant, free radical scavenging and anti-inflammatory effects of aloesin derivatives in aloe vera. Planta Med. 2002, 68, 957–960. [Google Scholar] [CrossRef]
  182. Lorenzetti, L.J.; Salisbury, R.; Beal, J.L.; Baldwin, J.N. Bacteriostatic property of aloe vera. J. Pharm. Sci. 1964, 53, 1287. [Google Scholar] [CrossRef]
  183. Choudhary, M.; Kochhar, A.; Sangha, J. Hypoglycemic and hypolipidemic effect of aloe vera l. In non-insulin dependent diabetics. J. Food Sci. Technol. 2014, 51, 90–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Fani, M.; Kohanteb, J. Inhibitory activity of aloe vera gel on some clinically isolated cariogenic and periodontopathic bacteria. J. Oral Sci. 2012, 54, 15–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Tanko, Y.; Salisu, A.I.; Mohammed, K.A.; Musa, S.A.; Jimoh, A.; Yusuf, R. Anti-hyperglycaemic effects of rutin on blood glucose, oxidative stress biomarkers and lipid peroxidation in alloxan-induced hyperglycaemic wistar rats. Niger J. Physiol. Sci. 2017, 32, 91–96. [Google Scholar] [PubMed]
  186. Zhang, R.; Zhang, B.L.; He, T.; Yi, T.; Yang, J.P.; He, B. Increase of rutin antioxidant activity by generating maillard reaction products with lysine. Bioorg. Med. Chem. Lett. 2016, 26, 2680–2684. [Google Scholar] [CrossRef] [PubMed]
  187. Mascaraque, C.; Aranda, C.; Ocón, B.; Monte, M.J.; Suárez, M.D.; Zarzuelo, A.; Marín, J.J.; Martínez-Augustin, O.; de Medina, F.S. Rutin has intestinal antiinflammatory effects in the cd4+ cd62l+ t cell transfer model of colitis. Pharmacol. Res. 2014, 90, 48–57. [Google Scholar] [CrossRef] [PubMed]
  188. Webster, R.P.; Gawde, M.D.; Bhattacharya, R.K. Protective effect of rutin, a flavonol glycoside, on the carcinogen-induced DNA damage and repair enzymes in rats. Cancer Lett. 1996, 109, 185–191. [Google Scholar] [CrossRef]
  189. Mauludin, R.; Müller, R.H.; Keck, C.M. Development of an oral rutin nanocrystal formulation. Int. J. Pharm. 2009, 370, 202–209. [Google Scholar] [CrossRef]
  190. Rizvi, S.; Raza, S.T.; Ahmed, F.; Ahmad, A.; Abbas, S.; Mahdi, F. The role of vitamin e in human health and some diseases. Sultan Qaboos Univ. Med. J. 2014, 14, e157–e165. [Google Scholar]
  191. Bas, N.; Kayar, N.A.; Baba, Z.F.; Avunduk, M.C.; Haliloğlu, S.; Alptekin, N. Systemic treatment with alpha-tocopherol and/or sodium selenite decreases the progression of experimental periodontitis. Clin. Oral Investig. 2021, 25, 2677–2688. [Google Scholar] [CrossRef]
  192. Chapple, I.L.; Matthews, J.B. The role of reactive oxygen and antioxidant species in periodontal tissue destruction. Periodontology 2000 2007, 43, 160–232. [Google Scholar] [CrossRef]
  193. Grisham, M.B.; Jourd’Heuil, D.; Wink, D.A. Nitric oxide. I. Physiological chemistry of nitric oxide and its metabolites:Implications in inflammation. Am. J. Physiol. 1999, 276, G315–G321. [Google Scholar] [PubMed]
  194. Rizk, M.; Witte, M.B.; Barbul, A. Nitric oxide and wound healing. World J. Surg. 2004, 28, 301–306. [Google Scholar] [CrossRef] [PubMed]
  195. Popkov, V.L.; Fil’chukova, I.A.; Lapina, N.V.; Galenko-Yaroshevskii, V.P.; Dukhanin, A.S. Activity of nitric oxide synthase and concentration of nitric oxide end metabolites in the gingiva under experimental pathological conditions. Bull. Exp. Biol. Med. 2005, 140, 391–393. [Google Scholar] [CrossRef] [PubMed]
  196. Shaker, O.; Ghallab, N.A.; Hamdy, E.; Sayed, S. Inducible nitric oxide synthase (inos) in gingival tissues of chronic periodontitis with and without diabetes: Immunohistochemistry and rt-pcr study. Arch. Oral Biol. 2013, 58, 1397–1406. [Google Scholar] [CrossRef] [PubMed]
  197. Pan, Z.; Guzeldemir, E.; Toygar, H.; Bal, N.; Bulut, S. Nitric oxide synthase in gingival tissues of patients with chronic periodontitis and with and without diabetes. J. Periodontol. 2010, 81, 109–120. [Google Scholar] [CrossRef] [Green Version]
  198. Struillou, X.; Boutigny, H.; Soueidan, A.; Layrolle, P. Experimental animal models in periodontology: A review. Open Dent. J. 2010, 4, 37–47. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Study selection flow chart.
Figure 1. Study selection flow chart.
Jcm 11 03614 g001
Figure 2. Summary of the pathogenesis of diabetes-associated periodontitis (DP).
Figure 2. Summary of the pathogenesis of diabetes-associated periodontitis (DP).
Jcm 11 03614 g002
Table 1. Summary of study types for each natural compound and its clinical evaluation.
Table 1. Summary of study types for each natural compound and its clinical evaluation.
Type of Natural ProductsStudies Type IncludedApplicable to Clinical Studies Only
Route of TreatmentDosageImprovements in:
ANICLINTOPSYS Periodontitis
(PD/CAL/ABL)
DM (FBG/HbA1c/Insulin Resistance)
Resveratrol//-/480 mg/day-
30 days
//
Curcumin///-100 m/L as irrigant-
single session
/N.A.
CMC2.24/-N.A.N.A.N.A.N.A.
Melatonin-///Top. cream 1%-
20 days
/-
Syst-3–6 mg/day-
8 weeks
//
Propolis//-/400 mg/day-
6 months
//
Vitamin C//-/450–500 mg/day–
2 weeks–2 months
--
Green Tea///-0.204 g strip-
single session
/N.A.
Ginger-/-/2 g/day-
8 weeks
/N.A.
Aloe Vera-//-Top. gel-N.A. dose
single session
/N.A.
Rutin/-N.A.N.A.N.A.N.A.
Vitamin E/ N.A.N.A.N.A.N.A.
ABL, alveolar bone loss; ANI, animal study; CAL, clinical attachment loss; CLIN, clinical study; FBG, fasting blood glucose; HbA1c, hemoglobin A1c; N.A., not available; PD, pocket depth; SYS, systemic; TOP, topical.
Table 2. Studies on the use of resveratrol (RSV) in diabetes-associated periodontitis (DP).
Table 2. Studies on the use of resveratrol (RSV) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
Zare Javid et al. (2019)
[17]
CLIN
(chronic P: moderate)
480 mg/day RSV
orally
30 days
(1)
SRP + Placebo
(2)
SRP + RSV
Both groups ↑ CAL gain, but no significant difference between them.
RSV ↓ IL-6, ↑ TAC.
Zare Javid et al. (2017)
[18]
CLIN
(chronic P: all stages)
480 mg/day RSV
orally
30 days
(1)
SRP + Placebo
(2)
SRP + RSV
RSV improved insulin level, insulin resistance and PD.
RSV ⬌ made to FBG and TG.
Giménez- Siurana et al. (2019)
[19]
ANI
(rats, STZ-induced DM; 50 mg/kg single dose)
3 mg/day RSV-SFN via oral gavage30 days
(1)
CMC
(2)
CMC + SF
(3)
CMC + RSV-SFN
(4)
CMC + SF + RSV-SFN
(5)
Water
Group 3: lowest area of inflammatory focus.
Group 4: greatest drop in IL-1β, IL-6 and has lowest epithelium thickness.
Overall, liquid SF + RSV-SFN work synergically in controlling DP.
Cirano et al. (2021)
[20]
ANI
(rats, STZ-induced DM; 60 mg/kg single dose)
10 mg/kg/day RSV via oral gavage30 days
(1)
DM + Placebo
(2)
DM + INS
(3)
DM + RSV
(4)
DM + RSV + INS
(5)
Non-DM
RSV alone ↓ NADPH oxidase activity.
RSV+ INS ↓ ABL and IL-1β, IL-6, IL-17, oxidative stress markers.
RSV, alone or combined with insulin ↑ anti-oxidants, SOD and SIRT l in DP.
Zhen et al. (2015)
[21]
ANI
(DM type 2 genetic mice)
20 mg/kg/day RSV via oral gavage30 days
(1)
DM
(2)
DP
(3)
DP + RSV
RSV ↓ FBG, ABL in DP.
mRNA, protein levels of TLR4 ↑ in DP.
RSV ↓ the inflammatory response via ↓ TLR4 expression and ↓ activation of TLR4 downstream signaling.
IV
(GEC of DM type 2 genetic mice)
10 µM RSV24 h prior to stimulation
(1)
Non-HG
(2)
HG
(3)
HG + RSV
↑ up-regulate; ↓ down-regulate; ⬌ no change; ANI, animal study; CLIN, clinical study; ABL, alveolar bone loss; CAL, clinical attachment level; DM, diabetes mellitus; FBG, fasting blood glucose; GEC, gingival epithelial cells; HG, high glucose; IL, interleukin; INS, insulin; NADPH, nicotinamide adenine dinucleotide phosphate; PD, pocket depth; SIRT, silent information regulator; SOD 1, superoxide dismutase; TAC, total anti-oxidant capacity; TG, triglyceride; TLR, Toll-like receptor.
Table 3. Studies on the use of curcumin (CUR) in diabetes-associated periodontitis (DP).
Table 3. Studies on the use of curcumin (CUR) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
CMC2.24
Deng et al. (2021)
[22]
ANI
(rats, STZ-induced DM; 70 mg/kg single dose)
30 mg/kg/day CMC2.24 via oral gavage3 weeks
(1)
DP
(2)
DP + CMC2.24
CMC2.24 ⬌on BG, HbA1c.
CMC2.24 ↓ ABL, DM-induced macrophage accumulation and impaired chemotactic activity.
CMC2.24 is anti-inflammatory, ↓ MMP-9, pro-resolvin.
Elburki et al.
(2017)
[24]
ANI
(rats, STZ-induced DM; 70 mg/kg single dose)
30 mg/kg/day CMC2.24 via oral gavage3 weeks
(1)
DP
(2)
DP + CMC2.24
CMC 2.24 ↓ ABL and ↓ IL-1β, IL-6.
CMC2.24 ↓ MMP-9, NF-κB (p65) and p38 MAPK.
Elburki et al. (2016)
[23]
ANI
(rats, STZ-induced DM; 70 mg/kg single dose, DM type 1)
30 mg/kg/day CMC2.24 via oral gavage3 weeks
(1)
DP
(2)
DP + CMC2.24
CMC 2.24 has ⬌ on BG, ↓ ABL.
CMC 2.24 ↓ IL-1β, IL-6 (x significant).
CMC 2.24 ↓ MMPs to near normal levels.
CUR
Ivanaga et al. (2019)
[25]
CLIN
(chronic P: all stages)
100 mg/L CUR solution via irrigation Single session
(1)
SRP
(2)
SRP + CUR
(3)
SRP + LED
(4)
SRP + CUR+ LED
CUR alone and with LED irradiation to SRP ↑ PD gain at 3- and 6-months post-therapy.
CUR + LED irradiation to SRP provide CAL gain at 3 months.
Iova et al. (2021)
[26]
ANI
(rats, STZ-induced DM; 30 mg/kg & 30% glucose single dose)
75 mg/kg/day CUR via oral gavage10 weeks
(1)
DP
(2)
DP + CUR
(3)
DP + Rutin
(4)
DP + CUR+ Rutin
CUR, single or paired with Rutin, ↓ oxidative stress both in gingival tissue and blood.
CUR ↑ anti-oxidant status in DP rats.
Pimentel et al. (2020)
[27]
ANI
(rats, STZ-induced DM; 60 mg/kg single dose)
100 mg/kg/day CUR via oral gavage30 days
(1)
DP + placebo
(2)
DP + CUR
(3)
DP + INS
(4)
DP + CURC + INS
CUR ↓ BG, similar to INS effect.
CUR ↑ Runx2 and SIRT gene.
CUR + INS synergically ↓ BG, ABL.
CUR +INS ↓ IL-6, IL-1β, RANKL/OPG ratio.
↑ up-regulate; ↓ down-regulate; ⬌ no change; ANI, animal study; CLIN, clinical study; ABL, alveolar bone loss; BG, blood glucose; CAL, clinical attachment level; CMC2.24, chemically modified curcumin; DM, diabetes mellitus; HbA1c, hemoglobin A1c; IL, interleukin; INS, insulin; MAPK, mitogen-activated protein kinase; MMPs, matrix metalloproteinases; PD, pocket depth; RANKL/OPG, receptor activator of nuclear factor-kappa B ligand/osteoprotegerin; SIRT, silent information regulator; sost, sclerostin; SRP, non-surgical scaling and root planning.
Table 4. Studies on the use of melatonin (MLT) in diabetes-associated periodontitis (DP).
Table 4. Studies on the use of melatonin (MLT) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
Anton et al. (2021)
[28]
CLIN
(chronic P: all stages)
3 mg/day MLT orally8 weeks
(1)
SRP + Placebo
(2)
SRP + MLT
MLT improved PD, CAL and glycemic control.
BOP, hygiene level improved more significant in MLT group.
Javid (2020)
[29]
CLIN
(chronic P: mild, moderate)
6 mg/day MLT orally8 weeks
(1)
SRP + Placebo
(2)
SRP + MLT
MLT ↓ IL-1β and MDA.
MLT ↑ serum levels of TAC, SOD, CAT, GPx.
Bazyar et al. (2019)
[30]
CLIN
(chronic P: mild, moderate)
6 mg/day MLT orally8 weeks
(1)
SRP + Placebo
(2)
SRP + MLT
MLT ↑ serum MLT, ↓ IL-6 and hs-CRP and improved PD, CAL.
Cutando et al. (2015)
[31]
CLIN
(P: all stages)
Topical MLT (1% orabase cream formula) 20 days
(1)
Placebo
(2)
MLT
MLT ↓ GI, PD and ↓ IL-6 and CRP serum levels.
Cutando et al. (2014)
[32]
CLIN
(P: all stages)
Topical MLT (1% orabase cream formula) 20 days
(1)
Placebo
(2)
MLT
MLT ↓ GI, PD, salivary levels of RANKL, and ↑ salivary OPG.
↑ up-regulate; ↓ down-regulate; CLIN, clinical study; BOP, bleeding on probing; CAL, clinical attachment level; CAT, catalase; CRP, C-reactive protein; GPx, glutathione peroxidase; hs-CRP, high-sensitivity C-reactive protein; IL, interleukin; MDA, malondialdehyde; PD, pocket depth; RANKL, receptor activator of nuclear factor-kappa B ligand; SRP, non-surgical scaling and root planning.
Table 5. Studies on the use of propolis (PP) in diabetes-associated periodontitis (DP).
Table 5. Studies on the use of propolis (PP) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
El-Sharkaw et al. (2016)
[33]
CLIN
(chronic P: moderate to severe)
400 mg/day PP orally6 months
(1)
SRP
(2)
SRP + PP
PP ↓ HbA1c level, FBG, CML both 3 and 6 months after SRP.
PP improved PD, CAL at 3 and 6 months.
Kızıldağ et al. (2020)
[34]
ANI
(rats, STZ-induced DM; 50 mg/kg single dose)
10 mM/kg/day CAPE via oral gavage15 days
(1)
DP
(2)
DP + CAPE
CAPE ↓ alveolar ABL, OSI and IL-1β.
CAPE ↓ histochemistry score and RANKL-stained cells.
CAPE ↓oxidative stress via ↑ TAS and ↓ TOS, OSI, RANKL and IL-1β levels.
Aral et al. (2015)
[35]
ANI
(rats, STZ-induced DM; 150 mg/kg single dose)
100 mg/kg/day PP orally3 weeks
(1)
DP
(2)
DP + PP
PP ↓ BG, ABL in DP.
PP ⬌ plasma IL-1β, TNF-α, MMP-8.
↑ up-regulate; ↓ down-regulate; ⬌ no change; ANI, animal study; CLIN, clinical study; ABL, alveolar bone loss; BG, blood glucose; CAPE, caffeic acid phenethyl ester; IL, interleukin; MMP-8, matrix metalloproteinase-8; OSI, oxidative stress index; P, periodontitis; RANKL, receptor activator of nuclear factor-kappa B ligand; TNF-α, tumor necrosis factor alpha.
Table 6. Studies on the use of vitamin C (Vit. C) in diabetes-associated periodontitis (DP).
Table 6. Studies on the use of vitamin C (Vit. C) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
Toraman et al. (2020)
[36]
ANI
(rats, ALX- induced DM; 150 mg/kg single dose)
50 μLVit C injected to buccal gingiva 3 times with 2 days interval after ligature removal.
(1)
DP + placebo
(2)
DP + Vit C
Vit. C ↓ ABL and ↓ CTX, 8-OHdG, and AGE ↓ in the DP group
Kunsongkeit et al. (2019)
[37]
CLIN
(chronic P: moderate to severe)
500 mg/day Vit. C orally2 months
(1)
SRP + placebo
(2)
SRP + Vit. C
Vit. C ↑ plasma vit. C, ⬌ FBG and HbA1c.
All periodontal parameters were improved in both groups, but no significant difference between them.
Gokhale et al. (2013)
[38]
CLIN
(chronic P: moderate to severe)
450 mg of AA orally2 weeks
(1)
SRP + placebo
(2)
SRP + AA
AAL plasma levels were ↓ in DP patients.
Vit. C ↓ sulcus bleeding index in DP, ⬌ in PI, PD
↑ up-regulate; ↓ down-regulate; ⬌ no change; ANI, animal study; CLIN, clinical study; AA, ascorbic acid; ABL, alveolar bone loss; AGE, advanced glycation end-products; ALX, alloxan monohydrate; CTX, C-terminal telopeptide fragments; DM, diabetes mellitus; FBG, fasting blood glucose; HbA1c, hemoglobin A1c; P, periodontitis; PD, pocket depth; PI, plaque index; SRP, non-surgical scaling and root planing.
Table 7. Studies on the use of green tea (GT) in diabetes-associated periodontitis (DP).
Table 7. Studies on the use of green tea (GT) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
Gadagi et al. (2013)
[39]
CLIN
(chronic P: all stages)
Local delivery of 0.204 g of GT strips Single
session
(1)
SRP + placebo
(2)
SRP + GT strips
10.67% of GT is released at first 30 min and ended at 120 min with full release.
GT improved GI, PD and CAL.
Catanzaro et al. (2018)
[40]
ANI
(rats, STZ-induced DM; 47 mg/kg single dose, DM type 1)
7 g GT diluted in 1 L distilled water ad libitum3 months
(1)
water
(2)
GT
GT ↓ FBG, ↓ DM-induced increased MVD, ↓ number of CD31 and VEGF-stained cells.
GT ↓ dental plaque accumulation and severity of periodontitis.
Gennaro et al. (2015)
[41]
ANI
(rats, STZ; 50 mg/kg single dose, DM type 1)
7 g GT diluted in 1 L distilled water ad libitum3 months
(1)
water
(2)
GT
GT ↓ TNF-α, RANKL.
GT ↑ IL-10, RUNX-2 and OPG.
↑ up-regulate; ↓ down-regulate; ANI, animal study; CLIN, clinical study; CAL, clinical attachment level; DM, diabetes mellitus; FBG, fasting blood glucose; GI, gingival index; IL-10, interleukin-10; MVD, microvascular density; OPG, osteoprotegerin; P, periodontitis; PD, pocket depth; RANKL, receptor activator of nuclear factor-kappa B ligand; RUNX-2, Runt-related transcription factor 2; TNF-α, tumor necrosis factor alpha; VEGF, vascular endothelial growth factor.
Table 8. Studies on the use of ginger (GG) in diabetes-associated periodontitis (DP).
Table 8. Studies on the use of ginger (GG) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
Zare Javid et al. (2019)
[42]
CLIN
(chronic P: mild to moderate)
2 g/day ginger orally8 weeks
  • SRP + placebo
  • SRP + GG
GG ↓ IL-6, hs-CRP, TNF-α, CAL, and PD.
GG ↑ SOD and GPx, however SOD difference between groups was not significant.
↑ up-regulate; ↓ down-regulate; CLIN, clinical study; CAL, clinical attachment level; GPx, glutathione peroxidase; hs-CRP, high-sensitivity C-reactive protein; IL-6, interleukin-6; PD, pocket depth; SOD, superoxide dismutase; SRP, non-surgical scaling and root planing; TNF-α, tumor necrosis factor alpha.
Table 9. Studies on the use of aloe vera (AV), rutin (RT), and vitamin E (Vit. E) in diabetes-associated periodontitis (DP).
Table 9. Studies on the use of aloe vera (AV), rutin (RT), and vitamin E (Vit. E) in diabetes-associated periodontitis (DP).
Author
(Year)
Types of StudyDosage & Administration RouteTreatment DurationStudy vs. Control GroupsStudy Outcomes
Pradeep et al. (2015)
[43]
CLIN
(chronic P: moderate to severe)
AV gel (injected to periodontal pocket)Single session
(1)
SRP + placebo
(2)
SRP + AV gel
AV group showed greater ↓ in PI, mSBI, and PD (3 months)
AV ↑ CAL gain at 3 and 6 months.
Iova et al. (2021)
[26]
ANI
(rats, STZ-induced DM; 30 mg/kg & 30% glucose single dose)
75 mg/kg/day RT via oral gavage10 weeks
(1)
DP
(2)
DP + CUR
(3)
DP + Rutin
(4)
DP + CUR+ RT
RT, single or combined with CUR, ↓ oxidative stress in gingival tissue and blood and ↑ anti-oxidant status.
Hatipoglu et al. (2016)
[44]
ANI
(rats, STZ-induced DM; 50 mg/kg single dose)
40 mg/kg/daily Vit. E (α-tocopherol) via intraperitoneal3 weeks
(1)
DP + saline
(2)
DP + α-tocopherol
α-tocopherol↓ number of iNOS positive cells.
↑ up-regulate; ↓ down-regulate; ANI, animal study; CLIN, clinical study; CAL, clinical attachment level; CAT, catalase; CUR, curcumin; mSBI, modified sulcus bleeding index; PD, pocket depth; PI, plaque index.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ng, M.Y.; Lin, T.; Chao, S.-C.; Chu, P.-M.; Yu, C.-C. Potential Therapeutic Applications of Natural Compounds in Diabetes-Associated Periodontitis. J. Clin. Med. 2022, 11, 3614. https://doi.org/10.3390/jcm11133614

AMA Style

Ng MY, Lin T, Chao S-C, Chu P-M, Yu C-C. Potential Therapeutic Applications of Natural Compounds in Diabetes-Associated Periodontitis. Journal of Clinical Medicine. 2022; 11(13):3614. https://doi.org/10.3390/jcm11133614

Chicago/Turabian Style

Ng, Min Yee, Taichen Lin, Shih-Chi Chao, Pei-Ming Chu, and Cheng-Chia Yu. 2022. "Potential Therapeutic Applications of Natural Compounds in Diabetes-Associated Periodontitis" Journal of Clinical Medicine 11, no. 13: 3614. https://doi.org/10.3390/jcm11133614

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

Article Metrics

Back to TopTop