Next Article in Journal
Massa Medicata Fermentata, a Functional Food for Improving the Metabolic Profile via Prominent Anti-Oxidative and Anti-Inflammatory Effects
Previous Article in Journal
Astaxanthin Supplementation Effects in Right Ventricle of Rats Exposed to Chronic Intermittent Hypobaric Hypoxia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis

1
Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
2
Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy
3
Department of Clinical Specialistic and Dental Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
4
Dentistry Clinic, National Institute of Health and Science of Aging, IRCCS INRCA, 60126 Ancona, Italy
5
Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, 60126 Ancona, Italy
6
IRCCS INRCA, 60124 Ancona, Italy
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(10), 1270; https://doi.org/10.3390/antiox13101270
Submission received: 7 October 2024 / Revised: 15 October 2024 / Accepted: 17 October 2024 / Published: 18 October 2024

Abstract

:
Periodontitis affects up to 40% of adults over 60 years old and is a consequence of gingivitis. Periodontitis is characterized by a chronic inflammation, periodontal damage, and alveolar bone resorption. The nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 (KEAP1) (NRF2/KEAP1) signaling pathway plays a key role in periodontitis by modulating redox balance and inflammation of the periodontium. However, NRF2 expression is decreased in gingival tissues of patients with periodontitis while oxidative stress is significantly increased in this pathology. Oxidative stress and lipopolysaccharide (LPS) produced by gram-negative bacteria favor the production of inflammatory causing periodontal inflammation and favoring alveolar bone. In this review, we analyzed the current literature regarding the role of natural and synthetic compounds in modulating the NRF2/KEAP1 pathway in in vitro and in vivo models of periodontitis in order to evaluate new potential treatments of periodontitis that can improve the outcome of this disease.

1. Introduction

Periodontitis is a chronic multifactorial inflammatory disease associated with dysbiotic plaque biofilms and characterized by a progressive destruction of the tooth-supporting apparatus [1,2]. Its main clinical features include the loss of periodontal tissue, due to the clinical attachment loss and the alveolar bone loss, and the presence of periodontal pocketing and gingival bleeding [3,4]. Periodontitis represents a major public health problem that contributes to the global burden of chronic non-communicable diseases. According to the Global Burden of Disease, periodontitis was ranked as one of the most prevalent conditions of humankind between 1990 to 2010, and a recent update confirmed that its prevalence is still substantial and worrisome, with an overall prevalence equal to 61.6% [5]. Periodontitis accounts for a substantial proportion of edentulism and masticatory dysfunction, resulting in significant dental care costs and a negative impact on general health [6]. It may lead to tooth loss and instability, negatively affect chewing function and aesthetics, be a source of social inequality, and impair quality of life [6].
According to the new classification of periodontal and peri-implant diseases, stages III and IV represent the most complex cases of periodontitis, due to the presence of angular defects, furcation involvements, tooth mobility, extensive tooth loss, and loss of function. In these stages, several intrinsic or environmental risk factors adversely affect the ability of the host to respond to the bacterial infection and to contain the tissue damage. Moreover, a significantly rapid progressive damage to the attachment apparatus is appreciated. Fortunately, a relative limited proportion of the population suffers from severe periodontitis (10–12%) and, usually, only a few teeth per person are involved [7,8,9].
The main risk factors associated with the periodontitis are the smoking and the diabetes mellitus. Other modifiable and non-modifiable risk factors may contribute to the developmental of periodontal disease, such as alcohol abuse, systemic diseases (cardiovascular, immunological, metabolic, and hematological diseases, as well as malnutrition), polypharmacological therapies, stress conditions, a sedentary lifestyle, and genetic predisposition. The progression of the periodontitis seems to be dependent on an abnormal host response to sub-gingival plaque biofilm. Over the past few years, strong evidence has emerged to implicate oxidative stress or presence of specific pathogens in pathogenesis of periodontitis. Since the nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 (KEAP1) (NRF2/KEAP1) signaling is the main pathway involved in the regulation of cellular redox homeostasis, several studies have investigated the role of this pathway in periodontitis pathophysiology. In fact, it has been demonstrated that NRF2/KEAP1 signaling is involved in the regulation of many cell processes that are altered in periodontitis.
Dental plaque harbors several bacterial pathogens which stimulate host cells to release various pro-inflammatory cytokines, leading to the hyper production of proteolytic enzymes and O2 by oxidative burst [10]. Porphyromonas gingivalis (P. gingivalis), Tannerella forsythia (T. forsythia) and Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) are the most common pathogens associated to periodontitis [11]. These periodontal pathogens are equipped with several virulence factors including fimbriae, adhesins, lipopolysaccharides, hemagglutinins, proteinases, and toxic products that favor pathogen survival and proliferation [12].
Reactive Oxygen Species (ROS) [13] play a key role in the pathophysiology of several diseases [14,15], including cancer [16,17,18,19,20], inflammatory diseases [21,22], and oral cavity diseases such as periodontitis [23,24]. The antioxidant defenses of the cell are able to mitigate the negative effects of high ROS levels, although harmful effects will take place if ROS overwhelm the antioxidant capacity of the cell [14,15,25]. Neutrophils play a key role in periodontitis since they represent the first defense line against pathogenic biofilm during periodontitis but also the major producers of ROS since they use the latter to fight the pathogenic bacteria [26]. In addition, lipopolysaccharide (LPS), produced by gram-negative bacteria during periodontitis, induces periodontal inflammation characterized by high levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1, as well as of nuclear factor-κB ligand (RANKL) which, in turn, causes an excessive osteoclast formation and activation, leading to alveolar bone loss [27,28,29]. In fact, RANKL is produced by osteoblasts, periodontal ligament fibroblasts and inflammatory cells under pro-inflammatory stimuli and plays a key role in osteoclastogenesis [27,30]. Inflammation may also be favored by the increased blood vessels found in gingiva of patients with periodontitis [31,32,33]. However, this mechanism in periodontitis requires more evidence and it is not widely recognized.
A schematic representation of periodontitis pathogenesis is shown in Figure 1.
The nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2)/Kelch-like ECH-Associated Protein 1 (KEAP1) (NRF2/KEAP1) signaling is a master antioxidant pathway in cells reducing ROS through the induction of genes encoding several antioxidant and phase II detoxifying enzymes [34,35]. Normally, NRF2 is present in the cytoplasm as an inactive complex and is bound to its repressor KEAP1. The latter is part of the Cullin 3 (Cul3)/RING box protein 1 (RBX1) E3-ubiquitin ligase complex that favors NRF2 ubiquitination and proteasomal degradation [35,36,37]. However, KEAP1 contains many reactive cysteine residues that act as sensors of intracellular redox conditions. In fact, modification (oxidation) of these cysteine residues (under oxidant stimuli) causes KEAP1 conformational changes that inhibit NRF2 proteasomal degradation and cause its nuclear translocation, allowing its binding to the antioxidant response elements (AREs) in the promoter of antioxidant enzyme genes, thus inducing their transcription [35,37,38] (Figure 2).
The multifaceted role of NRF2/KEAP1 signaling has been widely demonstrated in several cancerous [20,35,37] and non-cancerous [34,36,39,40,41] diseases, including periodontitis [42,43]. It is known that NRF2 expression is decreased in gingival tissues of patients with severe periodontitis [44] and, accordingly, that increased ROS levels during periodontitis worsens periodontal inflammation [45].
The effects of the NRF2/KEAP1 pathway have also been proven in NRF2 knockdown mice model of periodontitis where NRF2 absence caused a more severe alveolar bone loss as well as an increased oxidative stress in periodontal tissue [46]. The important role of this signaling has also been demonstrated in human periodontal ligament stem cells (hPDLSCs) where NRF2 overexpression attenuated apoptosis in oxidative stress conditions by activating the expression of antioxidant enzymes [47].
The NRF2/KEAP1 signaling pathway plays a key role also in osteoclastogenesis since mouse macrophages exposed to RANKL showed a decreased expression of NRF2 and NRF2-dependent antioxidant enzymes such as Heme-oxygenase 1 (HO-1) and NAD(P)H:quinone oxidoreductase (NQO1) [48]. These data demonstrated that NRF2 is an important inhibitor of osteoclastogenesis. In fact, it has been found that NRF2 overexpression induced the expression of the antioxidant enzymes HO-1, γ-glutamylcysteine synthetase (GCS) and NQO1 reducing osteoclast differentiation and then bone destruction [48].
Looking at the beneficial effects of NRF2/KEAP1 signaling activation reported in several studies, it is reasonable to think that increasing NRF2 expression in oral cavity could be an efficient therapeutic strategy for the prevention or treatment of patients with periodontitis.
The purpose of this review is to explore the potential role of natural compounds to promote NRF2/KEAP1 signaling activation by in vitro and in vivo models of periodontitis in order to suggest the use of these compounds to prevent or treat periodontitis, thus preventing or improving the outcome of this disease.

2. NRF2/KEAP1 Signaling Activation by Phytotherapeutics in Periodontitis Models

Natural compounds are biological substances used by plants (e.g., polyphenols, carotenoids, flavonoids, and anthocyanins) to protect themself from predators or external influences. Natural compounds can also be isolated from bacteria, fungi, and marine organisms [35,38,49,50,51]. Several natural compounds have shown important beneficial effects in many diseases, often for their antioxidant and anti-inflammatory effects, and are therefore used as supplements in natural medicine [35,38,49,50]. For these reasons, these compounds could also be used as supplement to protect oral tissues from oxidative stress and inflammation during periodontitis. Moreover, the presence of these compounds in chewing sticks, used as tools for oral hygiene, may show significant beneficial effects in preventing/treat periodontitis.
The studies discussed in this section are summarized in Table 1.
Quercetin is a natural compound with important antioxidant effects that can be found in many fruits and vegetables [52]. It has been reported that quercetin treatment of H2O2-exposed human periodontal ligament cells (hPDLCs) increased NRF2, NQO1, catalase (CAT), and HO-1 expression, reducing ROS, DNA damage, and cellular senescence. Quercetin also favored osteogenesis in H2O2-exposed hPDLCs. Moreover, quercetin treatment of periodontitis in mice increased NRF2 and SOD expression while reduced alveolar bone loss. Thus, quercetin can significantly improve antioxidant status and alveolar bone loss in periodontitis [53].
Biochanin A (BA) is an isoflavone present in several herbal products and has important anti-inflammatory and antioxidant effects [54]. Zhang et al. evaluated the effects of BA in rats with experimental periodontitis and found that BA treatment alleviated alveolar bone resorption and reduced interleukin (IL)-1β, Tumor Necrosis Factor (TNF)-α and ROS levels, as well as increased NRF2 protein expression demonstrating that BA can inhibit inflammation and bone loss in periodontitis [55].
Curcumin is a natural polyphenolic phytochemical widely used for its antioxidant, anticancer, and anti-inflammatory effects [49,56,57,58]. It can also reduce bone loss, inhibiting the proliferation and differentiation of osteoclasts and promoting their apoptosis [59,60]. A key role in osteogenic differentiation is played by the PI3K/AKT signaling pathway since it can favor this process [61,62]. An interesting study found that curcumin promoted osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) and activated the PI3K/AKT/NRF2 signaling pathway, favoring AKT phosphorylation and inducing NRF2 expression and its nuclear translocation. Interestingly, the inhibition of the PI3K/AKT signaling with the inhibitor LY294002 significantly decreased NRF2 expression. Moreover, the silencing of NRF2 (by siRNA) significantly reversed curcumin-induced osteogenic differentiation of hPDLSCs [63]. Thus, curcumin can induce the osteogenesis modulating PI3K/AKT/NRF2 signaling pathway. Additionally, curcumin pretreatment of an H400 oral epithelial cell line exposed to Fusobacterium nucleatum reduced IL-1β, TNF-α, and IL-8 expression while increasing NRF2 and HO-1 expression [64].
10-oxo-trans-11-octadecenoic acid (KetoC) is a bioactive metabolite generated from linoleic acid (abundant in sunflower seeds, walnuts, soybeans, corn, olives, and their oils) by intestinal microorganisms with important antioxidant and anti-inflammatory effects [65,66]. It has been found that KetoC treatment of gingival epithelial cells (GECs) increased the expression of NRF2, HO-1, and NQO1, decreasing ROS levels and demonstrating a protective function against the oxidative stress [67].
Caffeic acid phenethyl ester (CAPE) is a natural compound found in several plants that has important anti-inflammatory effects on periodontal inflammation [68,69]. It has been reported that primary murine macrophages, RAW 264.7 cells and primary human gingival fibroblasts exposed to CAPE showed an increased HO-1 expression. Moreover, NRF2 silencing attenuated CAPE-induced HO1 expression in macrophages and CAPE reduced IL-1α and IL-1β levels in primary murine macrophages and RAW 264.7 cells exposed to periodontal pathogens. Blocking HO-1 by its specific inhibitor (SnPP) decreased the antioxidative activity and attenuated the anti-inflammatory activity of CAPE. Thus, CAPE exerted its antioxidant and anti-inflammatory effects through the modulation of NRF2/HO-1 pathway [70].
Paeonol is a natural phenolic compound isolated from the root bark of Paeonia suffruticosa Andrews, a shrub widely used in traditional Chinese herbal medicine for its important anti-inflammatory and antioxidant activity [71,72]. Li et al. showed that paeonol treatment of periodontitis-induced rats decreased RANKL expression inhibiting osteoclasts formation. Moreover, paeonol reduced ROS, pro-inflammatory cytokines levels (IL-1β, IL-6 and TNF-α), and NF-κB activation, and alleviated oxidative stress, increasing HO-1 expression and glutathione (GSH) levels in gingival tissues. Importantly, paeonol increased NRF2 expression while NRF2 silencing decreased the inhibitory effect of paeonol on NF-κB activation, suggesting that the protective effect of paeonol against periodontitis-induced osteoclastogenesis and alveolar bone loss is mediated by the regulating of the NRF2/NF-κB signaling pathway [73].
Euphorbia factor L1 (EFL1) is a diterpenoid isolated from Euphorbia lathyris with several beneficial effects [74]. It has been reported that EFL1 treatment of mouse bone marrow-derived macrophages (used as osteoclast precursor) suppressed osteoclast formation and bone resorption inhibiting RANKL-induced c-Fos expression. Moreover, EFL1 decreased ROS levels, activating NRF2 signaling and increasing the expression of sulfiredoxin (SRX), peroxiredoxins (PRXs), and thioredoxins (TRXs). EFL1 also induced apoptosis in differentiated osteoclasts and inhibited inflammation-induced bone erosion in mice, suggesting that EFL1 regulates osteoclast differentiation by modulating redox status and inducing apoptosis in osteoclasts [75].
Resveratrol is a stilbenoid with important antioxidant and anti-inflammatory properties that can be found in grape, blueberries, raspberries, and mulberries [76]. Tamaki et al. showed that resveratrol could prevent the progression of periodontitis and reduce systemic oxidative stress. In fact, resveratrol administration to rats with periodontitis relieved alveolar bone resorption and activated the sirtuin 1 (SIRT1)/AMP-activated protein kinase (AMPK) and the NRF2 pathways in inflamed gingival tissues. Furthermore, resveratrol decreased TNF-α, IL-1, and IL-6 levels in rats with periodontitis [77]. In a mouse model of periodontitis, resveratrol reduced alveolar bone loss and oxidative stress in the periodontium. Interesting, NRF2 knockout reversed these results, demonstrating the involvement of NRF2 in periodontal bone healing in periodontitis [78].
Sulforaphane (SFN), a natural product found in cruciferous vegetables, increased intracellular reduced the glutathione (GSH)/oxidized glutathione (GSSG) ratio and the neutrophil respiratory burst in primary neutrophils from patients with periodontitis and controls. Interestingly, the chronic inflammation found in periodontitis is mainly due to the production of high levels of reactive oxygen species by neutrophils [79]. Moreover, SFN increased the expression of NRF2, NQO1, glutamate cysteine ligase catalytic (GCLC), and modifier (GCLM) subunits while reduced extracellular O2 (. -) production [80]. An important antioxidant effect of SFN has also been reported in gingival epithelial cells (GECs). In fact, SFN treatment of GECs significantly increased NRF2 and HO-1 expression [81].
Table 1. NRF2 modulators in periodontitis models.
Table 1. NRF2 modulators in periodontitis models.
ModulatorStructureModel UsedResultsRef.
QuercetinAntioxidants 13 01270 i001H2O2-exposed hPDLCs
Periodontitis mice model
Quercetin treatment increased NRF2, NQO1, CAT, and HO-1 expression, reducing ROS, DNA damage, and cellular senescence. Quercetin increased NRF2 and SOD expression, favored osteogenesis, and reduced alveolar bone loss.[53]
Biochanin A (BA)Antioxidants 13 01270 i002Periodontitis rat modelBA alleviated alveolar bone resorption and reduced IL-1β, TNF-α, and ROS levels, as well as increased NRF2 protein expression[55]
CurcuminAntioxidants 13 01270 i003hPDLSCsCurcumin induced AKT phosphorylation, NRF2 expression, and nuclear translocation. Inhibition of PI3K/AKT signaling decreased NRF2 expression while NRF2 silencing reversed curcumin-induced osteogenic differentiation.[63]
F. nucleatum-exposed H400 cell lineCurcumin reduced IL-1β, TNF-α, and IL-8 expression while increasing NRF2 and HO-1 expression.[64]
10-oxo-trans-11-octadecenoic acid (KetoC)Antioxidants 13 01270 i004GECsKetoC increased the expression of NRF2, HO-1, and NQO1, thus decreasing ROS levels.[67]
Caffeic acid phenethyl ester (CAPE)Antioxidants 13 01270 i005Primary murine macrophages
RAW 264.7 cells
Primary human gingival fibroblasts
CAPE increased HO-1 expression and reduced IL-1α and IL-1β levels. NRF2 silencing attenuated CAPE-induced HO-1 expression in macrophages. Inhibition of HO-1 by SnPP decreased the antioxidative activity and attenuated the anti-inflammatory activity of CAPE.[70]
PaeonolAntioxidants 13 01270 i006Periodontitis rat modelPaeonol decreased RANKL and inhibited osteoclasts formation. Paeonol reduced IL-1β, IL-6, and TNF-α, increased HO-1 expression and GSH levels, and reduced ROS levels in gingival tissues. Paeonol increased NRF2 expression while NRF2 silencing favored NF-κB activation by increasing pp65 subunit phosphorylation, thus abrogating the anti-inflammatory effect of paeonol.[73]
Euphorbia factor L1 (EFL1)Antioxidants 13 01270 i007Mice bone marrow-derived macrophages (used as osteoclast precursor).EFL1 treatment suppressed osteoclast formation and bone resorption, inhibiting RANKL-induced c-Fos expression. EFL1 decreased ROS levels activating NRF2 signaling and increasing SRX, PRXs, and TRXs expression. EFL1 induced apoptosis in differentiated osteoclasts and inhibited inflammation-induced bone erosion in mice.[75]
ResveratrolAntioxidants 13 01270 i008Periodontitis rat modelResveratrol administration relieved alveolar bone resorption and activated the Sirt1/AMP-activated protein kinase (AMPK) and the NRF2 pathways in inflamed gingival tissues. Resveratrol decreased TNF-α, IL-1, and IL-6 levels.[77]
Periodontitis mouse model.Resveratrol reduced alveolar bone loss and oxidative stress in the periodontium. NRF2 knockout reversed resveratrol effects.[78]
Sulforaphane (SFN)Antioxidants 13 01270 i009Differentiated HL60 cells (as a neutrophil model)
Primary neutrophils from patients.
SFN increased the intracellular GSH/GSSG ratio and reduced the neutrophil respiratory burst. SFN increased the expression of NRF2, NQO1, GCLC, and GCLM.[80]
GECsSFN increased NRF2 and HO-1 expression[81]
hPDLCs (human periodontal ligament cells); PDLSCs (human periodontal ligament stem cells); GECs (gingival epithelial cells). The chemical structures of the compounds illustrated in this table have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).

2.1. Effects of NRF2 Activation on RANKL-Induced Osteoclastogenesis in Periodontitis

The presence of LPS during periodontitis induces the production of pro-osteoclastogenetic cytokines such as TNF-α, IL-1, IL-6 by osteoblasts and periodontal ligament fibroblasts, T cells, and B cells, leading to the production RANKL, which has a pivotal function in osteoclast differentiation [27].
Dehydrocostus lactone (DL) is a natural sesquiterpene lactone derived from medicinal plants such as Inulahelenium L. and Saussurea lappa with antioxidant and anti-inflammatory properties [82,83,84,85]. Lee et al. found that RANKL-stimulated RAW 264.7 cells treated with DL attenuated NR-κB activation while increased NRF2, NQO1, sulfiredoxin (SRX), and peroxiredoxin-1 (PRX1) expression reducing ROS levels. Interestingly, NRF2 silencing promoted osteoclast differentiation, suggesting that DL attenuates osteoclast differentiation modulating NRF2 and NF-κB signaling pathways [86].
Hesperetin is a flavanone glycoside with anti-inflammatory and antioxidant activity that can be found in citrus fruits, grapefruits, and lemons [87]. It has been reported that hesperetin could be a potential therapeutic compound for periodontitis since it suppressed RANKL-induced osteoclastogenesis, osteoclastic bone resorption, and the activation of NF-κB and MAPK signaling in RAW 264.7 cells. Moreover, hesperetin increased NRF2, HO-1, and NQO1 expression scavenging ROS [88].
Thus, the activation of NRF2/KEAP1 signaling can significantly inhibit RANKL-induced osteoclast formation reducing inflammation and alveolar bone resorption.
The molecular structures of dehydrocostus lactone and hesperetin are shown in Figure 3.

2.2. Role of NRF2 Activation in LPS-Exposed Animal and Cell Models of Periodontitis

LPS released from gram-negative bacteria such as P. gingivalis stimulates ROS production from periodontal tissue, favoring the production of inflammatory cytokines such as TNF-α, IL-1, and IL-6 by inflammatory cells and causing periodontal inflammation and alveolar bone loss due to an excessive osteoclast formation and activation, since these cytokines also exert pro-osteoclastogenetic effects [27,89,90,91].
In this section, we discuss the effects of natural compounds on NRF2 activation in LPS-exposed animal and cell models of periodontitis.
The studies discussed in this section are summarized in Table 2.
Notopterol is a type of furanocoumarin isolated from Notopterygium incisum with important anti-inflammatory activities [92]. This compound may exert important function in periodontitis since notopterol treatment of LPS-stimulated human gingival fibroblasts (HGFs) significantly decreased IL-1β, IL-32, and IL-8 levels by inhibiting the activation of the NF-κB signaling pathway (inhibiting p65 subunit phosphorylation), a known pro-inflammatory signaling pathway [93]. Moreover, notopterol increased AKT and PI3K phosphorylation, as well as NRF2 expression. Notopterol also increased the expression of antioxidant enzymes such as HO-1, NQO1, CAT, and glutathione reductase (GSR), thus decreasing ROS levels. These effects were attenuated by the AKT inhibitor MK-2206.
Thus, notopterol relieved periodontal inflammation by inhibiting the activation the NF-κB and exerted antioxidant capacity activating the NRF2 and PI3K/AKT signaling pathways [94]. It is interesting to note that, according to other studies [95], the increased NRF2 expression was mediated by PI3K/AKT signaling since these effects were inhibited by MK-2206.
Isorhamnetin is a flavonoid isolated from Hippophae fhamnoides L. fruit with known anti-inflammatory effects. It has been reported that isorhamnetin treatment attenuated LPS-induced release of nitric oxide (NO), prostaglandin E2 (PGE2), IL-6, and IL-8 in human gingival fibroblasts (HGFs). Moreover, isorhamnetin inhibited LPS-induced activation of NF-κB (inhibiting p65 subunit phosphorylation), while increasing the expression of NRF2 and its downstream target HO-1. Importantly, NRF2 silencing, using siRNA, reversed the anti-inflammatory effects of isorhamnetin, suggesting that isorhamnetin inhibited LPS-induced inflammation in HGFs through the activation of the NRF2 signaling pathway [96]. This mechanism of action may be explained by the fact that silencing NRF2 decreased HO-1 expression, which inhibits NF-κB activation (a key player in inflammatory response), therefore favoring inflammation. This inhibitory effect of HO-1 on NF-κB activation may be due to the products derived from HO-1 activity such as CO, as reported in literature [97].
Magnolol is the main bioactive compound of Magnolia officinalis, a Chinese medicinal herb, and has important antioxidant and anti-inflammatory effects [98,99]. It has been found that magnolol treatment of RAW 264.7 macrophages exposed to LPS of P. gingivalis significantly reduced LPS-induced inflammation (reduced TNF-α, IL-1β levels), NF-κB activation, and increased NRF2 and HO-1 expression. Interestingly, NRF2/HO-1 activation by magnolol was decreased by blocking p38 MAPK activity with the specific inhibitor SB203580. This effect can be explained by the fact that MAPKs (p38, ERK, and JUK) can stimulate HO-1 expression by phosphorylating NRF2, thus favoring NRF2 nuclear translocation [100]. Inhibiting HO-1 activity by the SnPP inhibitor reversed the anti-inflammatory effects of magnolol proving that magnolol inhibits P. gingivalis LPS-induced inflammation in macrophages activating the NRF2/HO-1 axis, suggesting a possible use of magnolol in treatment of periodontitis [101].
Resveratrol, whose beneficial effects have been already discussed in the paragraph 2, also showed important effects against LPS exposure. In fact, it has been demonstrated that resveratrol treatment of LPS-stimulated human gingival fibroblasts (hGFs) reduced the expression of cyclooxygenase-2 (COX-2), matrix metalloproteinase (MMP)-2, MMP-9, and Toll-like receptor-4 (TLR4). Moreover, resveratrol reduced the activation of the MAPK signaling pathway while activating the NRF2/HO-1 axis, reducing ROS levels. Finally, resveratrol protected a periodontitis rat model against alveolar bone loss, inhibiting inflammation and osteoclast formation and increasing NRF2 and HO-1 expression in the gingiva of rat periodontitis [102]. Ma et al. confirmed these results, demonstrating that resveratrol treatment of LPS-stimulated hPDLSCs reduced IL-1β and IL-6 levels by reducing NF-κB activation (by decreasing p65/p50 subunits nuclear translocation and reducing p50 subunit expression) and increasing NRF2 and HO-1 expression. Moreover, resveratrol favored osteogenic differentiation of LPS-stimulated hPDLSCs [103]. Thus, oral administration of resveratrol may prevent the progression of periodontitis.
Lindenenyl acetate (LA) is one of the major constituents of Lindera strychnifolia Vill. (Lauraceae), a shrub native of Southeast Asia, used for its antioxidant and anti-diabetic properties [104,105]. Jeong et al. found that LA treatment of LPS-stimulated hPDLCs inhibited LPS-induced inducible nitric oxide synthase (iNOS), nitric oxide (NO), cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2) production. LA also attenuated the production of LPS-induced TNF-α, IL-1β, IL-6, and IL-12. Moreover, LA increased HO-1 expression and enzyme activity. Pretreatment with the HO-1 inhibitor (SnPP) decreased the inhibitory activities of LA on LPS-induced PGE2, IL-1β, TNF-α, IL-6, and IL-12 production. Interestingly, the authors found that the increased expression of HO-1 was due to the activation (nuclear translocation) of NRF2. Notably, the same authors found that LA up-regulated the levels of phosphorylated c-Jun N-terminal Kinase (JNK) while JNK pathway inhibition abolished LA-induced HO-1 expression by the synthetic inhibitor SP600125. Thus, the anti-inflammatory activity of LA in HPDL cells were mediated by the HO-1, JNK, and NRF2 pathways, suggesting LA as a potential therapeutic agent in periodontal disease [106].
Macrophages play a key role in periodontal lesions, regulating the production of pro-inflammatory cytokines involved in tissue and bone destruction during periodontitis [107]. Schisandrin is a natural compound isolated from the dried fruits of Schisandra chinensis [108,109] that has shown important anti-inflammatory effects on RAW 264.7 macrophages stimulated with LPS from P. gingivalis. In fact, schisandrin significantly inhibited the secretion of LPS-induced proinflammatory cytokines such as TNF-α, IL-1β, and IL-6, suppressing the activation of NF-kB (by inhibiting p65 subunit expression). HO-1 inhibition (by SnPP or siRNA) inhibited the anti-inflammatory activity of schisandrin. Furthermore, schisandrin induced HO-1 expression by increasing the expression of NRF2, PI3K/Akt, and ERK activation [110]. Thus, in addition to its antioxidant effects, HO-1 can be considered also a potent anti-inflammatory molecule able to regulate pro-inflammatory mediators. The anti-inflammatory effects of schisandrin may therefore be helpful for preventing/treating periodontitis.
Sappanchalcone is a natural flavonoid isolated from Caesalpinia sappan L., a plant used in traditional Chinese medicine. It has been demonstrated that sappanchalcone exerts important anti-inflammatory and neuroprotective effects [111,112]. An interesting study reported that sappanchalcone treatment of human dental pulp cells (HDPCs) and human periodontal ligament cells (hPDLCs) increased HO-1 expression and enzyme activity in both HDPCs and hPDLCs. Moreover, sappanchalcone protected HDPCs from H2O2-induced cytotoxicity and ROS production. In addition, these authors demonstrated that the cytoprotective effect of sappanchalcone was due to the inhibition of LPS-stimulated NO, PGE2, IL-1β, TNF-α, IL-6, and IL-12 release. These anti-inflammatory effects were partly inhibited by SnPP, a specific inhibitor of HO-1. Interestingly, the authors proved that the increased HO-1 expression was due to the activation of NRF2 and c-Jun NH2-terminal kinase (JNK). Thus, sappanchalcone protected HDPCs and hPDLCs from oxidative stress and inflammation activating NRF2 pathway and JNK suggesting a potential use as therapeutic compound for periodontal, pulpal, and periapical inflammatory lesion [113].
Table 2. NRF2 modulators in LPS-exposed models of periodontitis.
Table 2. NRF2 modulators in LPS-exposed models of periodontitis.
ModulatorStructureModel UsedResultsRef.
NotopterolAntioxidants 13 01270 i010LPS-stimulated HGFsNotopterol decreased IL-1β, IL-32, and IL-8 levels by inhibiting the activation of the NF-κB signaling pathway inhibiting the phosphorylation of p65 subunit. Notopterol increased AKT and PI3K phosphorylation and NRF2 expression. Notopterol increased HO-1, NQO1, CAT, and GSR expression and decreased ROS levels. These effects were attenuated by the AKT inhibitor MK-2206.[94]
IsorhamnetinAntioxidants 13 01270 i011LPS-stimulated HGFsIsorhamnetin attenuated LPS-induced release of PGE2, NO, IL-6, and IL-8, and inhibited NF-κB activation by inhibiting the phosphorylation of p65 subunit. Isorhamnetin increased the expression of NRF2 and HO-1. Silencing of NRF2 reversed the anti-inflammatory effects of isorhamnetin.[96]
MagnololAntioxidants 13 01270 i012LPS-stimulated RAW 264.7Magnolol reduced TNF-α and IL-1β levels and NF-κB activation (by inhibiting the phosphorylation of p65 subunit) while increasing NRF2 and HO-1 expression. NRF2/HO-1 activation by magnolol was diminished by blocking p38 MAPK activity. Inhibiting HO-1 activity by SnPP reversed the anti-inflammatory effects of magnolol.[101]
ResveratrolAntioxidants 13 01270 i013LPS-stimulated HGFs and Rats with periodontitisResveratrol reduced COX2, MMP-2, MMP-9, TLR4 expression and the activation of MAPK signaling pathway while activating the NRF2/HO-1 axis, reducing ROS levels. Resveratrol protected a periodontitis rat model against alveolar bone loss, inhibiting inflammation and osteoclast formation and increasing NRF2 and HO-1 expression in the gingiva of rats.[102]
LPS-stimulated hPDLSCsResveratrol reduced IL-1β and IL-6 levels and NF-κB activation by decreasing p65/p50 subunits nuclear translocation and p50 subunit expression. Resveratrol increased NRF2 and HO-1 expression and favored osteogenic differentiation.[103]
Lindenenyl acetate (LA)Antioxidants 13 01270 i014LPS-stimulated hPDLCsLA inhibited LPS-induced iNOS, NO, COX-2, and PGE2 production, and attenuated TNF-α, IL-1β, IL-6, and IL-12 secretion. LA increased NRF2 nuclear translocation and HO-1 expression and activity. HO-1 inhibition by SnPP decreased the inhibitory activities of LA on LPS-induced inflammatory cytokines production.[106]
SchisandrinAntioxidants 13 01270 i015LPS-stimulated RAW 264.7Schisandrin blocked the production of LPS-induced TNF-α, IL-1β, and IL-6, suppressing the activation of NF-kB signaling by inhibiting p65 subunit expression. Moreover, Schisandrin increased the expression of HO-1 and NRF2 and activated PI3K/Akt and ERK. Inhibiting HO-1 activity by SnPP reversed the surfactin-mediated inhibition of pro-inflammatory cytokines.[110]
SappanchalconeAntioxidants 13 01270 i016LPS-stimulated HDPCs and hPDLCsIncreased HO-1 expression and enzyme activity in both HDPCs and hPDLCs protected them from H2O2-induced ROS production. Sappanchalcone inhibited LPS-stimulated NO, PGE2, IL-1β, TNF-α, IL-6, and IL-12 release. The increased HO-1 expression was due to the activation of NRF2 and JNK.[113]
hPDLCs (human periodontal ligament cells); HGFs (human gingival fibroblasts); human dental pulp (HDPCs); human periodontal ligament stem cells (hPDLSCs). The chemical structures of the compounds illustrated in this table have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).

3. Role of NRF2 as Anti-Pyroptotic Target in Periodontitis

Pyroptosis is a physiological pro-inflammatory programmed death and the NOD-like receptor protein 3 (NLRP3) inflammasome pathway plays a key role in this process. NLRP3 is activated by external pathogens and danger signals, recruiting both apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) and caspase-1 to form the NLRP3 inflammasome. Once activated, the NLRP3 inflammasome induces caspase-1 activation and the secretion of pro-inflammatory cytokines such as IL-1β and IL-18, leading to cell death [114]. The NLRP3 inflammasome can also be activated by ROS and plays a key role in inflammatory diseases including periodontitis [114]. In fact, it has been demonstrated that NLRP3-mediated pyroptosis can induce inflammation, osteoclastogenesis, and alveolar bone loss [115].
The studies discussed in this section are summarized in Table 3.
Kynurenic acid (KA) is a metabolite of tryptophan, an amino acid present in many protein-based foods such as almonds [116,117], with important anti-inflammatory potential. It has been reported that KA treatment significantly improved the LPS-induced THP-1 macrophage viability, preventing pyroptosis through the reduction of NLRP3 and Caspase-1 expression, as well as IL-1β, IL-18, and TNF-α levels. Other authors have demonstrated that KA suppressed the NLRP3 inflammasome activation through the activation of NRF2/HO-1 axis, which led to ROS inhibition. These anti-pyroptotic and antioxidant effects of KA could be reversed by the inhibition of NRF2 (by using the synthetic inhibitor ML385). These data demonstrate that KA exerts its anti-pyroptotic effects through the activation of the NRF2 pathway [118].
In addition to KA, four other compounds (epigallocatechin-3-gallate, silibinin, chlorogenic acid, and eldecalcitol) have been found to be involved in the modulation of the NRF2/HO-1/NLRP3 axis.
Epigallocatechin-3-gallate (EGCG) is a polyphenol contained in green tea, with important anti-bacterial, anti-inflammatory, and antioxidant capacity [119]. It has been reported that EGCG significantly reduced alveolar bone loss in a periodontitis rat model. Moreover, EGCG decreased IL-1β, IL-18, TNF-α levels, decreasing NLRP3 expression and NF-κB activation. Additionally, the authors showed that EGCG increased NRF2, HO-1 and SOD expression decreasing oxidative stress [120].
Silibinin (SB) is a natural compound that can be found in silymarin, also called milk thistle or Silybum marianum, with important antioxidant and anti-inflammatory effects [121]. Li et al. found that SB reduced alveolar bone loss, oxidative stress, NF-κB and NLRP3 expression, and TNF-α, IL-1β, and IL-6, while increasing NRF2 expression in the periodontitis rat model. Thus, SB exhibited important anti-inflammatory and antioxidative properties by regulating both NRF2 and NF-κB signaling, suggesting a promising potential clinical application of this compound in treatment of periodontitis [122].
Chlorogenic acid (CA) is a natural compound isolated from Coffea canephora, Coffea arabica L. and Lonicerae japonicae with anticancer, antioxidant and anti-inflammatory effects [123,124,125]. It has been found that CA treatment of LPS-induced Human gingival fibroblasts (HGFs) inhibited NLRP3 expression and reduced IL-1β and IL-18 levels while increasing NRF2 and HO-1 expression and reducing oxidative stress, suggesting that CA could attenuate inflammation in HGFs during periodontitis [126]. These results are in agreement with another study that evaluated the effects of CA in LPS-induced immortalized human oral keratinocytes (IHOKs). In this study, the authors found that CA treatment attenuated LPS-induced inflammatory mediators release (e.g., PGE2), ROS production, and NF-κB activation, reducing inflammation. Moreover, CA promoted NRF2 translocation and HO-1 expression. Thus, coffee consumption may be beneficial for alleviating periodontitis [127].
Table 3. Modulators targeting NRF2 to inhibit pyroptosis in periodontitis.
Table 3. Modulators targeting NRF2 to inhibit pyroptosis in periodontitis.
ModulatorStructureModel UsedResultsRef.
Kynurenic acid (KA)Antioxidants 13 01270 i017LPS-induced THP-1 macrophageKA improved cell viability preventing pyroptosis through the reduction of NLRP3 and Caspase-1 expression, as well as IL-1β, IL-18, and TNF-α levels. KA suppressed NLRP3 inflammasome activation through the activation of the NRF2/HO-1 axis, which led to ROS inhibition. These effects were reversed by the inhibition of NRF2.[118]
Epigallocatechin-3-gallate (EGCG)Antioxidants 13 01270 i018Periodontitis rat modelEGCG reduced alveolar bone loss, lowered IL-1β, IL-18, TNF-α levels, and decreased NLRP3 expression and NF-κB activation (inhibiting p65 subunit expression). EGCG increased NRF2 and HO-1 expression, decreasing oxidative stress.[120]
Silibinin (SB)Antioxidants 13 01270 i019Periodontitis rat modelSB reduced alveolar bone loss, oxidative stress, NF-κB (inhibiting p65 subunit expression), NLRP3 expression, and TNF-α, IL-1β, and IL-6 levels while increasing NRF2 expression in the periodontium.[122]
Chlorogenic acid (CA)Antioxidants 13 01270 i020LPS-induced HGFsCA treatment inhibited the contents of IL-1β and IL-18 while increasing NRF2 and HO-1 expression, reducing oxidative stress.[126]
LPS-induced IHOKsCA treatment attenuated LPS-induced inflammatory mediators release (PGE2), ROS production, and NF-κB activation (by inhibiting p65 subunit phosphorylation), reducing inflammation. Moreover, CA promoted NRF2 translocation and HO-1 expression.[127]
HGFs (human gingival fibroblasts); IHOKs (immortalized human oral keratinocytes). The chemical structures of the compounds illustrated in this table have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).

4. Role of NRF2 in Periodontitis Complicated by Diabetes

Diabetes mellitus (DM) is a complex metabolic disorder characterized by an impaired glucose tolerance and hyperglycemia as a result of insulin deficiency or resistance [128]. Chronic hyperglycemia can result in a damage of several organs and tissues, including the retina, kidneys, heart, and blood vessels. Moreover, DM has been associated with an increased risk of developing cardiovascular disease (CVD) [129,130].
DM is a well-recognized risk factor of periodontitis. It is formally included in the grading classification system of periodontitis because the prevalence and severity of periodontitis in people with DM are significantly higher than in normoglycemic people. Although the correlation between DM and periodontitis is still not clear, it has been found that oxidative stress is a key pathogenic factor of diabetic periodontitis [131,132]. The role of DM in periodontitis has also been elucidated in diabetic rats. In fact, induction of periodontitis in diabetic rats led to a more severe alveolar bone loss and periodontal cell apoptosis than in normoglycemic rats. Moreover, in diabetic rats with induced periodontitis there was an increased local and systemic oxidative damage associated to a decreased NRF2 expression and increased levels of oxidative stress markers such as 3-NT-, 4-HNE-, MDA, and 8-OHdG in periodontal lesions. Thus, the enhanced local and systemic oxidative damage correlates to the downregulation of NRF2, favoring the development and progression of diabetic periodontitis [133].
Chronic periodontitis with diabetes mellitus (CPDM) has been correlated with the high glucose levels present in diabetic patients [134]. Moreover, CPDM onset is also favored by the increased ROS levels found in diabetic patients [135]. An interesting study found that human gingival epithelial cells (hGECs) treated with high glucose concentrations or P. gingivalis LPS have increased ROS levels while the mRNA levels of NRF2, catalase (CAT), glutamate-cysteine ligase catalytic subunit (GCLC), superoxide dismutase 1 (SOD1), and SOD2 were decreased [136]. However, ROS levels decreased after pretreatment with baicalein (BCI), a natural polyphenolic flavonoid that can be found in Scutellaria baicalensis Georgi (a flowering plant) [137]. In addition, BCI promoted the nucleus translocation of NRF2 inducing the expression of its target gene (CAT, GCLC, SOD1, and SOD2). Finally, a CPDM rat model treated with BCI showed an increased expression of NRF2 in periodontal tissue and mitigated the alveolar bone loss. Thus, BCI treatment may have beneficial effects in CPDM patients [136].
Diabetes plays also a key role in cell senescence, altering tissue repair. In fact, it has been reported that a high glucose microenvironment induces hPDLSC senescence, and senescent hPDLSCs show a diminished abilities to proliferate and differentiate impairing periodontal tissue repair and regeneration ability [138]. However, activation of NRF2/KEAP1 signaling can prevent cell senescence [139]. In a high glucose microenvironment, KEAP1 expression was increased while NRF2, HO-1, and NQO1 expression were significantly decreased, leading to increased oxidative stress [138]. Thus, therapies targeting NRF2 activation can reduce cell senescence, favoring periodontal tissue repair and regeneration in periodontitis by favoring in osteoblast or cementoblast hPDLSCs differentiation.
Uncontrolled DM leads to the accumulation of advanced glycation end-products (AGEs) through glycation reaction, causing several DM-associated complications such as diabetic retinopathy, peripheral neuropathy, and peripheral vascular diseases [140]. AGEs levels are significantly increased type-2 DM patients with chronic periodontitis compared to healthy individuals with or without periodontitis [141]. Moreover, AGEs trigger ROS accumulation, increasing oxidative stress and inflammation [142,143,144].
Magnolol, a natural compound found in Magnolia officinalis, showed protective effects in AGEs-exposed HGF, reducing ROS and increasing NRF2 and HO-1 expression. Moreover, magnolol significantly reduced AGEs-induced IL-6 and IL-8 production, demonstrating that magnolol has anti-inflammatory and antioxidant effects in AGEs-exposed HGF [145].
Thus, baicalein and magnolol may be used as a potential therapeutic approach for treatment of diabetes-associated periodontitis.
The molecular structures of the natural compounds described above are shown in Figure 4.

5. Conclusions and Further Remarks

Periodontitis is a microbially-associated disease characterized by a host-mediated inflammation leading to the activation of host-derived proteinases, that result in loss of marginal periodontal ligament fibers, apical migration of the junctional epithelium, and apical spread of the bacterial biofilm along the root surface. Its progression depends on dysbiotic ecological modifications in response to nutrients from gingival inflammation and tissue damage.
The NRF2/KEAP1 signaling pathway is a promising target for future research since it can significantly improve several cellular processes. In this review, we discussed many studies highlighting the multifaceted role of this pathway in regulating many important processes, including inflammation and osteogenesis, and how this pathway can be modulated by a variety of natural compounds. In particular, it has been highlighted that certain compounds can reduce inflammation by inhibiting NF-κB signaling activation, a key pathway involved in cytokine production [93], thus reducing cytokine levels. In particular, the inhibition of NF-κB signaling activation was due to the increased HO-1 activity (a NRF2-dependent enzyme). In fact, it has been reported that this inhibitory effect of HO-1 on NF-κB signaling activation may be due to the products derived from HO-1 activity, such as CO [97].
The compounds discussed in this review activate NRF2/KEAP1 signaling, favoring the expression of several antioxidant enzymes that significantly reduce oxidative stress in the periodontium. The activation of this pathway also favors osteogenesis by inhibiting RANKL-induced osteoclast formation, thus inhibiting alveolar bone loss (as demonstrated in the in vivo model of periodontitis).
Overall, NRF2/KEAP1 signaling can be modulated by several natural compounds that could be used in combination with classical periodontitis treatments. In addition, since these compounds are safe for human consumption, they could be used as preventive treatment during gingivitis, since the worsening of this pathology leads to periodontitis (Figure 1). A schematic representation of NRF2/KEAP1 signaling modulation by phytotherapeutics in periodontitis is shown in Figure 5.
Since NRF2 activation plays a key function on protecting cells from oxidative stress and inflammation, as well as alveolar bone loss, the development of specific drugs or food supplements based on the natural compounds with this function may have a significant clinical impact not only in periodontitis treatment but also in the prevention of this disease.
It deserves to be pointed out that all the studies discussed in this review have been performed on cell lines or animal periodontitis models (mice and rats). Thus, specific clinical trials are necessary to evaluate the dose/effect relationship in humans in order to evaluate the beneficial effects of these phototherapeutics in patients with periodontitis.

Author Contributions

Conceptualization, software, writing—original draft preparation G.T.; writing—review and editing, S.F., D.M., L.T., F.O. and A.S.; supervision, project administration, funding acquisition M.R.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by PON Ministero dello Sviluppo Economico (MiSe) 2018 project “ChAALenge”—F/180016/01-05/X43.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Nunn, M.E. Understanding the etiology of periodontitis: An overview of periodontal risk factors. Periodontol. 2000 2003, 32, 11–23. [Google Scholar] [CrossRef] [PubMed]
  2. Abdulkareem, A.A.; Al-Taweel, F.B.; Al-Sharqi, A.J.B.; Gul, S.S.; Sha, A.; Chapple, I.L.C. Current concepts in the pathogenesis of periodontitis: From symbiosis to dysbiosis. J. Oral Microbiol. 2023, 15, 2197779. [Google Scholar] [CrossRef] [PubMed]
  3. Papapanou, P.N.; Sanz, M.; Buduneli, N.; Dietrich, T.; Feres, M.; Fine, D.H.; Flemmig, T.F.; Garcia, R.; Giannobile, W.V.; Graziani, F.; et al. Periodontitis: Consensus report of workgroup 2 of the 2017 World Workshop on the Classification of Periodontal and Peri-Implant Diseases and Conditions. J. Clin. Periodontol. 2018, 45 (Suppl. 20), S162–S170. [Google Scholar] [CrossRef] [PubMed]
  4. Tonetti, M.S.; Greenwell, H.; Kornman, K.S. Staging and grading of periodontitis: Framework and proposal of a new classification and case definition. J. Periodontol. 2018, 89 (Suppl. 1), S159–S172. [Google Scholar] [CrossRef]
  5. Trindade, D.; Carvalho, R.; Machado, V.; Chambrone, L.; Mendes, J.J.; Botelho, J. Prevalence of periodontitis in dentate people between 2011 and 2020: A systematic review and meta-analysis of epidemiological studies. J. Clin. Periodontol. 2023, 50, 604–626. [Google Scholar] [CrossRef]
  6. Caton, J.G.; Armitage, G.; Berglundh, T.; Chapple, I.L.C.; Jepsen, S.; Kornman, K.S.; Mealey, B.L.; Papapanou, P.N.; Sanz, M.; Tonetti, M.S. A new classification scheme for periodontal and peri-implant diseases and conditions—Introduction and key changes from the 1999 classification. J. Clin. Periodontol. 2018, 45 (Suppl. 20), S1–S8. [Google Scholar] [CrossRef]
  7. Billings, M.; Holtfreter, B.; Papapanou, P.N.; Mitnik, G.L.; Kocher, T.; Dye, B.A. Age-dependent distribution of periodontitis in two countries: Findings from NHANES 2009 to 2014 and SHIP-TREND 2008 to 2012. J. Clin. Periodontol. 2018, 45 (Suppl. 20), S130–S148. [Google Scholar] [CrossRef]
  8. Needleman, I.; Garcia, R.; Gkranias, N.; Kirkwood, K.L.; Kocher, T.; Iorio, A.D.; Moreno, F.; Petrie, A. Mean annual attachment, bone level, and tooth loss: A systematic review. J. Periodontol. 2018, 89 (Suppl. 1), S120–S139. [Google Scholar] [CrossRef]
  9. Kassebaum, N.J.; Bernabe, E.; Dahiya, M.; Bhandari, B.; Murray, C.J.; Marcenes, W. Global burden of severe periodontitis in 1990-2010: A systematic review and meta-regression. J. Dent. Res. 2014, 93, 1045–1053. [Google Scholar] [CrossRef]
  10. Dahiya, P.; Kamal, R.; Gupta, R.; Bhardwaj, R.; Chaudhary, K.; Kaur, S. Reactive oxygen species in periodontitis. J. Indian Soc. Periodontol. 2013, 17, 411–416. [Google Scholar] [CrossRef]
  11. Van Dyke, T.E.; Sheilesh, D. Risk factors for periodontitis. J. Int. Acad. Periodontol. 2005, 7, 3–7. [Google Scholar] [PubMed]
  12. Shahoumi, L.A.; Saleh, M.H.A.; Meghil, M.M. Virulence Factors of the Periodontal Pathogens: Tools to Evade the Host Immune Response and Promote Carcinogenesis. Microorganisms 2023, 11, 115. [Google Scholar] [CrossRef] [PubMed]
  13. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
  14. Emanuelli, M.; Sartini, D.; Molinelli, E.; Campagna, R.; Pozzi, V.; Salvolini, E.; Simonetti, O.; Campanati, A.; Offidani, A. The Double-Edged Sword of Oxidative Stress in Skin Damage and Melanoma: From Physiopathology to Therapeutical Approaches. Antioxidants 2022, 11, 612. [Google Scholar] [CrossRef]
  15. Sartini, D.; Campagna, R.; Lucarini, G.; Pompei, V.; Salvolini, E.; Mattioli-Belmonte, M.; Molinelli, E.; Brisigotti, V.; Campanati, A.; Bacchetti, T.; et al. Differential immunohistochemical expression of paraoxonase-2 in actinic keratosis and squamous cell carcinoma. Hum. Cell 2021, 34, 1929–1931. [Google Scholar] [CrossRef]
  16. Campagna, R.; Belloni, A.; Pozzi, V.; Salvucci, A.; Notarstefano, V.; Togni, L.; Mascitti, M.; Sartini, D.; Giorgini, E.; Salvolini, E.; et al. Role Played by Paraoxonase-2 Enzyme in Cell Viability, Proliferation and Sensitivity to Chemotherapy of Oral Squamous Cell Carcinoma Cell Lines. Int. J. Mol. Sci. 2022, 24, 338. [Google Scholar] [CrossRef]
  17. Campagna, R.; Pozzi, V.; Salvucci, A.; Togni, L.; Mascitti, M.; Sartini, D.; Salvolini, E.; Santarelli, A.; Lo Muzio, L.; Emanuelli, M. Paraoxonase-2 expression in oral squamous cell carcinoma. Hum. Cell 2023, 36, 1211–1213. [Google Scholar] [CrossRef]
  18. Togni, L.; Mascitti, M.; Sartini, D.; Campagna, R.; Pozzi, V.; Salvolini, E.; Offidani, A.; Santarelli, A.; Emanuelli, M. Nicotinamide N-Methyltransferase in Head and Neck Tumors: A Comprehensive Review. Biomolecules 2021, 11, 1594. [Google Scholar] [CrossRef]
  19. Fantone, S.; Piani, F.; Olivieri, F.; Rippo, M.R.; Sirico, A.; Di Simone, N.; Marzioni, D.; Tossetta, G. Role of SLC7A11/xCT in Ovarian Cancer. Int. J. Mol. Sci. 2024, 25, 587. [Google Scholar] [CrossRef]
  20. Marzioni, D.; Mazzucchelli, R.; Fantone, S.; Tossetta, G. NRF2 modulation in TRAMP mice: An in vivo model of prostate cancer. Mol. Biol. Rep. 2023, 50, 873–881. [Google Scholar] [CrossRef]
  21. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [PubMed]
  22. Campagna, R.; Mateuszuk, L.; Wojnar-Lason, K.; Kaczara, P.; Tworzydlo, A.; Kij, A.; Bujok, R.; Mlynarski, J.; Wang, Y.; Sartini, D.; et al. Nicotinamide N-methyltransferase in endothelium protects against oxidant stress-induced endothelial injury. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 119082. [Google Scholar] [CrossRef] [PubMed]
  23. Shang, J.; Liu, H.; Zheng, Y.; Zhang, Z. Role of oxidative stress in the relationship between periodontitis and systemic diseases. Front. Physiol. 2023, 14, 1210449. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, Y.; Andrukhov, O.; Rausch-Fan, X. Oxidative Stress and Antioxidant System in Periodontitis. Front. Physiol. 2017, 8, 910. [Google Scholar] [CrossRef] [PubMed]
  25. Campagna, R.; Pozzi, V.; Giorgini, S.; Morichetti, D.; Goteri, G.; Sartini, D.; Serritelli, E.N.; Emanuelli, M. Paraoxonase-2 is upregulated in triple negative breast cancer and contributes to tumor progression and chemoresistance. Hum. Cell 2023, 36, 1108–1119. [Google Scholar] [CrossRef]
  26. Miyasaki, K.T. The neutrophil: Mechanisms of controlling periodontal bacteria. J. Periodontol. 1991, 62, 761–774. [Google Scholar] [CrossRef]
  27. AlQranei, M.S.; Chellaiah, M.A. Osteoclastogenesis in periodontal diseases: Possible mediators and mechanisms. J. Oral Biosci. 2020, 62, 123–130. [Google Scholar] [CrossRef]
  28. Zhu, L.; Zhou, C.; Chen, S.; Huang, D.; Jiang, Y.; Lan, Y.; Zou, S.; Li, Y. Osteoporosis and Alveolar Bone Health in Periodontitis Niche: A Predisposing Factors-Centered Review. Cells 2022, 11, 3380. [Google Scholar] [CrossRef]
  29. Bhuyan, R.; Bhuyan, S.K.; Mohanty, J.N.; Das, S.; Juliana, N.; Juliana, I.F. Periodontitis and Its Inflammatory Changes Linked to Various Systemic Diseases: A Review of Its Underlying Mechanisms. Biomedicines 2022, 10, 2659. [Google Scholar] [CrossRef]
  30. Tsukasaki, M. RANKL and osteoimmunology in periodontitis. J. Bone Miner. Metab. 2021, 39, 82–90. [Google Scholar] [CrossRef]
  31. Zoellner, H.; Hunter, N. Vascular expansion in chronic periodontitis. J. Oral. Pathol. Med. 1991, 20, 433–437. [Google Scholar] [CrossRef] [PubMed]
  32. Gurav, A.N. The implication of periodontitis in vascular endothelial dysfunction. Eur. J. Clin. Investig. 2014, 44, 1000–1009. [Google Scholar] [CrossRef] [PubMed]
  33. Aspriello, S.D.; Zizzi, A.; Spazzafumo, L.; Rubini, C.; Lorenzi, T.; Marzioni, D.; Bullon, P.; Piemontese, M. Effects of enamel matrix derivative on vascular endothelial growth factor expression and microvessel density in gingival tissues of periodontal pocket: A comparative study. J. Periodontol. 2011, 82, 606–612. [Google Scholar] [CrossRef] [PubMed]
  34. Tossetta, G.; Fantone, S.; Piani, F.; Crescimanno, C.; Ciavattini, A.; Giannubilo, S.R.; Marzioni, D. Modulation of NRF2/KEAP1 Signaling in Preeclampsia. Cells 2023, 12, 1545. [Google Scholar] [CrossRef] [PubMed]
  35. Tossetta, G.; Marzioni, D. Targeting the NRF2/KEAP1 pathway in cervical and endometrial cancers. Eur. J. Pharmacol. 2023, 941, 175503. [Google Scholar] [CrossRef]
  36. Szczesny-Malysiak, E.; Stojak, M.; Campagna, R.; Grosicki, M.; Jamrozik, M.; Kaczara, P.; Chlopicki, S. Bardoxolone Methyl Displays Detrimental Effects on Endothelial Bioenergetics, Suppresses Endothelial ET-1 Release, and Increases Endothelial Permeability in Human Microvascular Endothelium. Oxidative Med. Cell. Longev. 2020, 2020, 4678252. [Google Scholar] [CrossRef]
  37. Tossetta, G.; Fantone, S.; Marzioni, D.; Mazzucchelli, R. Cellular Modulators of the NRF2/KEAP1 Signaling Pathway in Prostate Cancer. Front. Biosci. (Landmark Ed.) 2023, 28, 143. [Google Scholar] [CrossRef]
  38. Tossetta, G.; Fantone, S.; Marzioni, D.; Mazzucchelli, R. Role of Natural and Synthetic Compounds in Modulating NRF2/KEAP1 Signaling Pathway in Prostate Cancer. Cancers 2023, 15, 3037. [Google Scholar] [CrossRef]
  39. Chen, J.; Wang, Q.; Li, R.; Li, Z.; Jiang, Q.; Yan, F.; Ye, J. The role of Keap1-Nrf2 signaling pathway during the progress and therapy of diabetic retinopathy. Life Sci. 2024, 338, 122386. [Google Scholar] [CrossRef]
  40. Cai, S.; Sun, Y.; Wang, Y.; Lin, Z. Exploring the effect of LncRNA DANCR to regulate the Keap1-Nrf2/ARE pathway on oxidative stress in rheumatoid arthritis. Immun. Inflamm. Dis. 2024, 12, e1163. [Google Scholar] [CrossRef]
  41. Ward, K.W. Targeting the NRF2 pathway: A promising approach for corneal endothelial dysfunction. Curr. Opin. Pharmacol. 2024, 74, 102429. [Google Scholar] [CrossRef] [PubMed]
  42. Ma, F.; Luo, S.; Lu, C.; Jiang, X.; Chen, K.; Deng, J.; Ma, S.; Li, Z. The role of Nrf2 in periodontal disease by regulating lipid peroxidation, inflammation and apoptosis. Front. Endocrinol. (Lausanne) 2022, 13, 963451. [Google Scholar] [CrossRef] [PubMed]
  43. Chiu, A.V.; Saigh, M.A.; McCulloch, C.A.; Glogauer, M. The Role of NrF2 in the Regulation of Periodontal Health and Disease. J. Dent. Res. 2017, 96, 975–983. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, W.Y.; Meng, X.; Wang, Y.R.; Wang, Q.Q.; He, X.; Sun, X.Y.; Cheng, N.; Zhang, L. PRDX6 alleviates lipopolysaccharide-induced inflammation and ferroptosis in periodontitis. Acta Odontol. Scand. 2022, 80, 535–546. [Google Scholar] [CrossRef] [PubMed]
  45. Petelin, M.; Pavlica, Z.; Ivanusa, T.; Sentjurc, M.; Skaleric, U. Local delivery of liposome-encapsulated superoxide dismutase and catalase suppress periodontal inflammation in beagles. J. Clin. Periodontol. 2000, 27, 918–925. [Google Scholar] [CrossRef]
  46. 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]
  47. Liu, Y.; Yang, H.; Wen, Y.; Li, B.; Zhao, Y.; Xing, J.; Zhang, M.; Chen, Y. Nrf2 Inhibits Periodontal Ligament Stem Cell Apoptosis under Excessive Oxidative Stress. Int. J. Mol. Sci. 2017, 18, 1076. [Google Scholar] [CrossRef]
  48. Kanzaki, H.; Shinohara, F.; Kajiya, M.; Kodama, T. The Keap1/Nrf2 protein axis plays a role in osteoclast differentiation by regulating intracellular reactive oxygen species signaling. J. Biol. Chem. 2013, 288, 23009–23020. [Google Scholar] [CrossRef]
  49. Tossetta, G.; Fantone, S.; Giannubilo, S.R.; Marzioni, D. The Multifaced Actions of Curcumin in Pregnancy Outcome. Antioxidants 2021, 10, 126. [Google Scholar] [CrossRef]
  50. Tossetta, G.; Fantone, S.; Licini, C.; Marzioni, D.; Mattioli-Belmonte, M. The multifaced role of HtrA1 in the development of joint and skeletal disorders. Bone 2022, 157, 116350. [Google Scholar] [CrossRef]
  51. Bacchetti, T.; Campagna, R.; Sartini, D.; Cecati, M.; Morresi, C.; Bellachioma, L.; Martinelli, E.; Rocchetti, G.; Lucini, L.; Ferretti, G.; et al. C. spinosa L. subsp. rupestris Phytochemical Profile and Effect on Oxidative Stress in Normal and Cancer Cells. Molecules 2022, 27, 6488. [Google Scholar] [CrossRef] [PubMed]
  52. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef] [PubMed]
  53. Wei, Y.; Fu, J.; Wu, W.; Ma, P.; Ren, L.; Yi, Z.; Wu, J. Quercetin Prevents Oxidative Stress-Induced Injury of Periodontal Ligament Cells and Alveolar Bone Loss in Periodontitis. Drug Des. Devel Ther. 2021, 15, 3509–3522. [Google Scholar] [CrossRef] [PubMed]
  54. Feng, Z.J.; Lai, W.F. Chemical and Biological Properties of Biochanin A and Its Pharmaceutical Applications. Pharmaceutics 2023, 15, 1105. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, S.; Niu, Y.; Yang, Z.; Zhang, Y.; Guo, Q.; Yang, Y.; Zhou, X.; Ding, Y.; Liu, C. Biochanin A alleviates gingival inflammation and alveolar bone loss in rats with experimental periodontitis. Exp. Ther. Med. 2020, 20, 251. [Google Scholar] [CrossRef]
  56. Perugini, J.; Di Mercurio, E.; Tossetta, G.; Severi, I.; Monaco, F.; Reguzzoni, M.; Tomasetti, M.; Dani, C.; Cinti, S.; Giordano, A. Biological Effects of Ciliary Neurotrophic Factor on hMADS Adipocytes. Front. Endocrinol. 2019, 10, 768. [Google Scholar] [CrossRef]
  57. Tossetta, G.; Fantone, S.; Busilacchi, E.M.; Di Simone, N.; Giannubilo, S.R.; Scambia, G.; Giordano, A.; Marzioni, D. Modulation of matrix metalloproteases by ciliary neurotrophic factor in human placental development. Cell Tissue Res. 2022, 390, 113–129. [Google Scholar] [CrossRef]
  58. Ahmad, I.; Ahmad, S.; Ahmad, A.; Zughaibi, T.A.; Alhosin, M.; Tabrez, S. Curcumin, its derivatives, and their nanoformulations: Revolutionizing cancer treatment. Cell Biochem. Funct. 2024, 42, e3911. [Google Scholar] [CrossRef]
  59. Yang, M.W.; Wang, T.H.; Yan, P.P.; Chu, L.W.; Yu, J.; Gao, Z.D.; Li, Y.Z.; Guo, B.L. Curcumin improves bone microarchitecture and enhances mineral density in APP/PS1 transgenic mice. Phytomedicine 2011, 18, 205–213. [Google Scholar] [CrossRef]
  60. Hatefi, M.; Ahmadi, M.R.H.; Rahmani, A.; Dastjerdi, M.M.; Asadollahi, K. Effects of Curcumin on Bone Loss and Biochemical Markers of Bone Turnover in Patients with Spinal Cord Injury. World Neurosurg. 2018, 114, e785–e791. [Google Scholar] [CrossRef]
  61. Guntur, A.R.; Rosen, C.J. The skeleton: A multi-functional complex organ: New insights into osteoblasts and their role in bone formation: The central role of PI3Kinase. J. Endocrinol. 2011, 211, 123–130. [Google Scholar] [CrossRef] [PubMed]
  62. Tsai, K.S.; Kao, S.Y.; Wang, C.Y.; Wang, Y.J.; Wang, J.P.; Hung, S.C. Type I collagen promotes proliferation and osteogenesis of human mesenchymal stem cells via activation of ERK and Akt pathways. J. Biomed. Mater. Res. Part A 2010, 94, 673–682. [Google Scholar] [CrossRef] [PubMed]
  63. Xiong, Y.; Zhao, B.; Zhang, W.; Jia, L.; Zhang, Y.; Xu, X. Curcumin promotes osteogenic differentiation of periodontal ligament stem cells through the PI3K/AKT/Nrf2 signaling pathway. Iran. J. Basic. Med. Sci. 2020, 23, 954–960. [Google Scholar] [CrossRef] [PubMed]
  64. Grant, M.M.; Scott, A.E.; Matthews, J.B.; Griffiths, H.R.; Chapple, I.L.C. Pre-conditioning of gingival epithelial cells with sub-apoptotic concentrations of curcumin prevents pro-inflammatory cytokine release. J. Periodontal Res. 2023, 58, 634–645. [Google Scholar] [CrossRef]
  65. Furumoto, H.; Nanthirudjanar, T.; Kume, T.; Izumi, Y.; Park, S.B.; Kitamura, N.; Kishino, S.; Ogawa, J.; Hirata, T.; Sugawara, T. 10-Oxo-trans-11-octadecenoic acid generated from linoleic acid by a gut lactic acid bacterium Lactobacillus plantarum is cytoprotective against oxidative stress. Toxicol. Appl. Pharmacol. 2016, 296, 1–9. [Google Scholar] [CrossRef]
  66. Sulijaya, B.; Takahashi, N.; Yamada, M.; Yokoji, M.; Sato, K.; Aoki-Nonaka, Y.; Nakajima, T.; Kishino, S.; Ogawa, J.; Yamazaki, K. The anti-inflammatory effect of 10-oxo-trans-11-octadecenoic acid (KetoC) on RAW 264.7 cells stimulated with Porphyromonas gingivalis lipopolysaccharide. J. Periodontal Res. 2018, 53, 777–784. [Google Scholar] [CrossRef]
  67. Yokoji-Takeuchi, M.; Takahashi, N.; Yamada-Hara, M.; Sulijaya, B.; Tsuzuno, T.; Aoki-Nonaka, Y.; Tabeta, K.; Kishino, S.; Ogawa, J.; Yamazaki, K. A bacterial metabolite induces Nrf2-mediated anti-oxidative responses in gingival epithelial cells by activating the MAPK signaling pathway. Arch. Oral Biol. 2020, 110, 104602. [Google Scholar] [CrossRef]
  68. 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]
  69. Ercan, N.; Erdemir, E.O.; Ozkan, S.Y.; Hendek, M.K. The comparative effect of propolis in two different vehicles; mouthwash and chewing-gum on plaque accumulation and gingival inflammation. Eur. J. Dent. 2015, 9, 272–276. [Google Scholar] [CrossRef]
  70. Stahli, A.; Maheen, C.U.; Strauss, F.J.; Eick, S.; Sculean, A.; Gruber, R. Caffeic acid phenethyl ester protects against oxidative stress and dampens inflammation via heme oxygenase 1. Int. J. Oral. Sci. 2019, 11, 6. [Google Scholar] [CrossRef]
  71. Meng, L.; Xu, W.; Guo, L.; Ning, W.; Zeng, X. Paeonol Inhibits the Proliferation, Invasion, and Inflammatory Reaction Induced by TNF-alpha in Vascular Smooth Muscle Cells. Cell Biochem. Biophys. 2015, 73, 495–503. [Google Scholar] [CrossRef] [PubMed]
  72. Li, P.; Shen, J.; Wang, Z.; Liu, S.; Liu, Q.; Li, Y.; He, C.; Xiao, P. Genus Paeonia: A comprehensive review on traditional uses, phytochemistry, pharmacological activities, clinical application, and toxicology. J. Ethnopharmacol. 2021, 269, 113708. [Google Scholar] [CrossRef] [PubMed]
  73. Li, J.; Li, Y.; Pan, S.; Zhang, L.; He, L.; Niu, Y. Paeonol attenuates ligation-induced periodontitis in rats by inhibiting osteoclastogenesis via regulating Nrf2/NF-kappaB/NFATc1 signaling pathway. Biochimie 2019, 156, 129–137. [Google Scholar] [CrossRef] [PubMed]
  74. Zhu, A.; Zhang, T.; Wang, Q. The phytochemistry, pharmacokinetics, pharmacology and toxicity of Euphorbia semen. J. Ethnopharmacol. 2018, 227, 41–55. [Google Scholar] [CrossRef]
  75. Hong, S.E.; Lee, J.; Seo, D.H.; In Lee, H.; Ri Park, D.; Lee, G.R.; Jo, Y.J.; Kim, N.; Kwon, M.; Shon, H.; et al. Euphorbia factor L1 inhibits osteoclastogenesis by regulating cellular redox status and induces Fas-mediated apoptosis in osteoclast. Free Radic. Biol. Med. 2017, 112, 191–199. [Google Scholar] [CrossRef]
  76. Silva, P.M.; Goncalves, C.; Pastrana, L.M.; Coimbra, M.A.; Vicente, A.A.; Cerqueira, M.A. Recent advances in oral delivery systems of resveratrol: Foreseeing their use in functional foods. Food Funct. 2023, 14, 10286–10313. [Google Scholar] [CrossRef]
  77. 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]
  78. Ikeda, E.; Tanaka, D.; Glogauer, M.; Tenenbaum, H.C.; Ikeda, Y. Healing effects of monomer and dimer resveratrol in a mouse periodontitis model. BMC Oral Health 2022, 22, 460. [Google Scholar] [CrossRef]
  79. Chapple, I.L.; Matthews, J.B. The role of reactive oxygen and antioxidant species in periodontal tissue destruction. Periodontol. 2000 2007, 43, 160–232. [Google Scholar] [CrossRef]
  80. Dias, I.H.; Chapple, I.L.; Milward, M.; Grant, M.M.; Hill, E.; Brown, J.; Griffiths, H.R. Sulforaphane restores cellular glutathione levels and reduces chronic periodontitis neutrophil hyperactivity in vitro. PLoS ONE 2013, 8, e66407. [Google Scholar] [CrossRef]
  81. Mineo, S.; Takahashi, N.; Yamada-Hara, M.; Tsuzuno, T.; Aoki-Nonaka, Y.; Tabeta, K. Rice bran-derived protein fractions enhance sulforaphane-induced anti-oxidative activity in gingival epithelial cells. Arch. Oral Biol. 2021, 129, 105215. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, J.; Yu, Z.; Wang, C.; Tian, X.; Huo, X.; Wang, Y.; Sun, C.; Feng, L.; Ma, J.; Zhang, B.; et al. Dehydrocostus lactone, a natural sesquiterpene lactone, suppresses the biological characteristics of glioma, through inhibition of the NF-kappaB/COX-2 signaling pathway by targeting IKKbeta. Am. J. Cancer Res. 2017, 7, 1270–1284. [Google Scholar] [PubMed]
  83. Paulsen, E. The sesquiterpene lactone mix: A review of past, present and future aspects. Contact Dermat. 2023, 89, 434–441. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, L.; Yang, M.; Zhang, C.; Huang, F. The protective effects of dehydrocostus lactone against TNF-alpha-induced degeneration of extracellular matrix (ECM) in SW1353 cells. Aging 2020, 12, 17137–17149. [Google Scholar] [CrossRef] [PubMed]
  85. Seo, M.S.; Choi, E.M. The effects of dehydrocostus lactone on osteoblastic MC3T3-E1 cells in redox changes and PI3K/Akt/CREB. Immunopharmacol. Immunotoxicol. 2012, 34, 810–814. [Google Scholar] [CrossRef]
  86. Lee, H.I.; Lee, G.R.; Lee, J.; Kim, N.; Kwon, M.; Kim, H.J.; Kim, N.Y.; Park, J.H.; Jeong, W. Dehydrocostus lactone inhibits NFATc1 via regulation of IKK, JNK, and Nrf2, thereby attenuating osteoclastogenesis. BMB Rep. 2020, 53, 218–222. [Google Scholar] [CrossRef]
  87. Wdowiak, K.; Walkowiak, J.; Pietrzak, R.; Bazan-Wozniak, A.; Cielecka-Piontek, J. Bioavailability of Hesperidin and Its Aglycone Hesperetin-Compounds Found in Citrus Fruits as a Parameter Conditioning the Pro-Health Potential (Neuroprotective and Antidiabetic Activity)-Mini-Review. Nutrients 2022, 14, 2647. [Google Scholar] [CrossRef]
  88. Liu, H.; Dong, Y.; Gao, Y.; Zhao, L.; Cai, C.; Qi, D.; Zhu, M.; Zhao, L.; Liu, C.; Guo, F.; et al. Hesperetin suppresses RANKL-induced osteoclastogenesis and ameliorates lipopolysaccharide-induced bone loss. J. Cell Physiol. 2019, 234, 11009–11022. [Google Scholar] [CrossRef]
  89. Golz, L.; Memmert, S.; Rath-Deschner, B.; Jager, A.; Appel, T.; Baumgarten, G.; Gotz, W.; Frede, S. LPS from P. gingivalis and hypoxia increases oxidative stress in periodontal ligament fibroblasts and contributes to periodontitis. Mediat. Inflamm. 2014, 2014, 986264. [Google Scholar] [CrossRef]
  90. Manolagas, S.C. Birth and death of bone cells: Basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr. Rev. 2000, 21, 115–137. [Google Scholar] [CrossRef]
  91. Rodan, G.A.; Martin, T.J. Therapeutic approaches to bone diseases. Science 2000, 289, 1508–1514. [Google Scholar] [CrossRef] [PubMed]
  92. Inthanon, S.; Dejkriengkraikul, P.; Yodkeeree, S. Notopterol Suppresses IL-17-Induced Proliferation and Invasion of A549 Lung Adenocarcinoma Cells via Modulation of STAT3, NF-kappaB, and AP-1 Activation. Int. J. Mol. Sci. 2023, 24, 5057. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed]
  94. Zhou, J.; Shi, P.; Ma, R.; Xie, X.; Zhao, L.; Wang, J. Notopterol Inhibits the NF-kappaB Pathway and Activates the PI3K/Akt/Nrf2 Pathway in Periodontal Tissue. J. Immunol. 2023, 211, 1516–1525. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, L.; Chen, Y.; Sternberg, P.; Cai, J. Essential roles of the PI3 kinase/Akt pathway in regulating Nrf2-dependent antioxidant functions in the RPE. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1671–1678. [Google Scholar] [CrossRef]
  96. Qi, F.; Sun, J.H.; Yan, J.Q.; Li, C.M.; Lv, X.C. Anti-inflammatory effects of isorhamnetin on LPS-stimulated human gingival fibroblasts by activating Nrf2 signaling pathway. Microb. Pathog. 2018, 120, 37–41. [Google Scholar] [CrossRef]
  97. Lee, T.S.; Tsai, H.L.; Chau, L.Y. Induction of heme oxygenase-1 expression in murine macrophages is essential for the anti-inflammatory effect of low dose 15-deoxy-Delta 12,14-prostaglandin J2. J. Biol. Chem. 2003, 278, 19325–19330. [Google Scholar] [CrossRef]
  98. Shih, C.Y.; Chou, T.C. The antiplatelet activity of magnolol is mediated by PPAR-beta/gamma. Biochem. Pharmacol. 2012, 84, 793–803. [Google Scholar] [CrossRef]
  99. Lee, Y.J.; Lee, Y.M.; Lee, C.K.; Jung, J.K.; Han, S.B.; Hong, J.T. Therapeutic applications of compounds in the Magnolia family. Pharmacol. Ther. 2011, 130, 157–176. [Google Scholar] [CrossRef]
  100. Furukawa, Y.; Urano, T.; Minamimura, M.; Nakajima, M.; Okuyama, S.; Furukawa, S. 4-Methylcatechol-induced heme oxygenase-1 exerts a protective effect against oxidative stress in cultured neural stem/progenitor cells via PI3 kinase/Akt pathway. Biomed. Res. 2010, 31, 45–52. [Google Scholar] [CrossRef]
  101. Lu, S.H.; Hsu, W.L.; Chen, T.H.; Chou, T.C. Activation of Nrf2/HO-1signaling pathway involves the anti-inflammatory activity of magnolol in Porphyromonas gingivalis lipopolysaccharide-stimulated mouse RAW 264.7 macrophages. Int. Immunopharmacol. 2015, 29, 770–778. [Google Scholar] [CrossRef] [PubMed]
  102. Bhattarai, G.; Poudel, S.B.; Kook, S.H.; Lee, J.C. Resveratrol prevents alveolar bone loss in an experimental rat model of periodontitis. Acta Biomater. 2016, 29, 398–408. [Google Scholar] [CrossRef] [PubMed]
  103. Ma, Y.; Qian, Y.; Chen, Y.; Ruan, X.; Peng, X.; Sun, Y.; Zhang, J.; Luo, J.; Zhou, S.; Deng, C. Resveratrol modulates the inflammatory response in hPDLSCs via the NRF2/HO-1 and NF-kappaB pathways and promotes osteogenic differentiation. J. Periodontal Res. 2023, 59, 162–173. [Google Scholar] [CrossRef] [PubMed]
  104. Ohno, T.; Takemura, G.; Murata, I.; Kagawa, T.; Akao, S.; Minatoguchi, S.; Fujiwara, T.; Fujiwara, H. Water extract of the root of Lindera strychnifolia slows down the progression of diabetic nephropathy in db/db mice. Life Sci. 2005, 77, 1391–1403. [Google Scholar] [CrossRef] [PubMed]
  105. Noda, Y.; Mori, A. Antioxidant activities of uyaku (lindera strychnifolia) leaf extract: A natural extract used in traditional medicine. J. Clin. Biochem. Nutr. 2007, 41, 139–145. [Google Scholar] [CrossRef]
  106. Jeong, G.S.; Lee, D.S.; Li, B.; Kim, J.J.; Kim, E.C.; Kim, Y.C. Anti-inflammatory effects of lindenenyl acetate via heme oxygenase-1 and AMPK in human periodontal ligament cells. Eur. J. Pharmacol. 2011, 670, 295–303. [Google Scholar] [CrossRef]
  107. Yoshimura, A.; Hara, Y.; Kaneko, T.; Kato, I. Secretion of IL-1 beta, TNF-alpha, IL-8 and IL-1ra by human polymorphonuclear leukocytes in response to lipopolysaccharides from periodontopathic bacteria. J. Periodontal Res. 1997, 32, 279–286. [Google Scholar] [CrossRef]
  108. Ko, K.M.; Ip, S.P.; Poon, M.K.; Wu, S.S.; Che, C.T.; Ng, K.H.; Kong, Y.C. Effect of a lignan-enriched fructus schisandrae extract on hepatic glutathione status in rats: Protection against carbon tetrachloride toxicity. Planta Med. 1995, 61, 134–137. [Google Scholar] [CrossRef]
  109. Wang, X.; Wang, X.; Yao, H.; Shen, C.; Geng, K.; Xie, H. A comprehensive review on Schisandrin and its pharmacological features. Naunyn Schmiedebergs Arch. Pharmacol. 2024, 397, 783–794. [Google Scholar] [CrossRef]
  110. Park, S.Y.; Park, D.J.; Kim, Y.H.; Kim, Y.; Kim, S.G.; Shon, K.J.; Choi, Y.W.; Lee, S.J. Upregulation of heme oxygenase-1 via PI3K/Akt and Nrf-2 signaling pathways mediates the anti-inflammatory activity of Schisandrin in Porphyromonas gingivalis LPS-stimulated macrophages. Immunol. Lett. 2011, 139, 93–101. [Google Scholar] [CrossRef]
  111. Moon, H.I.; Chung, I.M.; Seo, S.H.; Kang, E.Y. Protective effects of 3’-deoxy-4-O-methylepisappanol from Caesalpinia sappan against glutamate-induced neurotoxicity in primary cultured rat cortical cells. Phytother. Res. 2010, 24, 463–465. [Google Scholar] [CrossRef] [PubMed]
  112. Washiyama, M.; Sasaki, Y.; Hosokawa, T.; Nagumo, S. Anti-inflammatory constituents of Sappan Lignum. Biol. Pharm. Bull. 2009, 32, 941–944. [Google Scholar] [CrossRef] [PubMed]
  113. Jeong, G.S.; Lee, D.S.; Li, B.; Lee, H.J.; Kim, E.C.; Kim, Y.C. Effects of sappanchalcone on the cytoprotection and anti-inflammation via heme oxygenase-1 in human pulp and periodontal ligament cells. Eur. J. Pharmacol. 2010, 644, 230–237. [Google Scholar] [CrossRef] [PubMed]
  114. Zhao, Y.; Quan, Y.; Lei, T.; Fan, L.; Ge, X.; Hu, S. The Role of Inflammasome NLPR3 in the Development and Therapy of Periodontitis. Int. J. Med. Sci. 2022, 19, 1603–1614. [Google Scholar] [CrossRef] [PubMed]
  115. Li, X.; Men, X.; Ji, L.; Chen, X.; He, S.; Zhang, P.; Chen, S. NLRP3-mediated periodontal ligament cell pyroptosis promotes root resorption. J. Clin. Periodontol. 2024, 51, 474–486. [Google Scholar] [CrossRef]
  116. Toth, F.; Cseh, E.K.; Vecsei, L. Natural Molecules and Neuroprotection: Kynurenic Acid, Pantethine and alpha-Lipoic Acid. Int. J. Mol. Sci. 2021, 22, 403. [Google Scholar] [CrossRef]
  117. Campagna, R.; Vignini, A. NAD(+) Homeostasis and NAD(+)-Consuming Enzymes: Implications for Vascular Health. Antioxidants 2023, 12, 376. [Google Scholar] [CrossRef]
  118. Gao, Y.; Guo, X.; Zhou, Y.; Du, J.; Lu, C.; Zhang, L.; Sun, S.; Wang, S.; Li, Y. Kynurenic acid inhibits macrophage pyroptosis by suppressing ROS production via activation of the NRF2 pathway. Mol. Med. Rep. 2023, 28, 211. [Google Scholar] [CrossRef]
  119. Liao, S.; Tang, Y.; Chu, C.; Lu, W.; Baligen, B.; Man, Y.; Qu, Y. Application of green tea extracts epigallocatechin-3-gallate in dental materials: Recent progress and perspectives. J. Biomed. Mater. Res. Part A 2020, 108, 2395–2408. [Google Scholar] [CrossRef]
  120. Fan, Q.; Zhou, X.H.; Wang, T.F.; Zeng, F.J.; Liu, X.; Gu, Y.; Chen, B.; Yang, J.; Pang, Z.Y.; Liu, J.G.; et al. Effects of epigallocatechin-3-gallate on oxidative stress, inflammation, and bone loss in a rat periodontitis model. J. Dent. Sci. 2023, 18, 1567–1575. [Google Scholar] [CrossRef]
  121. Akhoond-Ali, Z.; Rahimi, A.; Ghorbani, A.; Forouzanfar, F.; Hosseinian, S.; Ghazavi, H.; Vafaee, F. Silibinin effects on cognitive disorders: Hope or treatment? Avicenna J. Phytomed. 2023, 13, 597–614. [Google Scholar] [CrossRef] [PubMed]
  122. Li, X.; Zhou, R.; Han, Y.; Zeng, J.; Shi, L.; Mao, Y.; Sun, X.; Ji, Y.; Zhang, X.; Chen, Y.; et al. Silibinin Attenuates Experimental Periodontitis by Downregulation of Inflammation and Oxidative Stress. Oxidative Med. Cell. Longev. 2023, 2023, 5617800. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, L.; Du, H.; Chen, P. Chlorogenic acid inhibits the proliferation of human lung cancer A549 cell lines by targeting annexin A2 in vitro and in vivo. Biomed. Pharmacother. 2020, 131, 110673. [Google Scholar] [CrossRef] [PubMed]
  124. El-Khadragy, M.F.; Al-Megrin, W.A.; Alomar, S.; Alkhuriji, A.F.; Metwally, D.M.; Mahgoub, S.; Amin, H.K.; Habotta, O.A.; Abdel Moneim, A.E.; Albeltagy, R.S. Chlorogenic acid abates male reproductive dysfunction in arsenic-exposed mice via attenuation of testicular oxido-inflammatory stress and apoptotic responses. Chem. Biol. Interact. 2021, 333, 109333. [Google Scholar] [CrossRef] [PubMed]
  125. Huang, J.; Xie, M.; He, L.; Song, X.; Cao, T. Chlorogenic acid: A review on its mechanisms of anti-inflammation, disease treatment, and related delivery systems. Front. Pharmacol. 2023, 14, 1218015. [Google Scholar] [CrossRef]
  126. Huang, X.; Liu, Y.; Shen, H.; Fu, T.; Guo, Y.; Qiu, S. Chlorogenic acid attenuates inflammation in LPS-induced Human gingival fibroblasts via CysLT1R/Nrf2/NLRP3 signaling. Int. Immunopharmacol. 2022, 107, 108706. [Google Scholar] [CrossRef]
  127. Song, J.; Kim, B.; Kim, O.; Yang, Y.; Liu, D.; Fu, W.; Ma, G.; Kim, Y.; Kim, O. Effect of Coffee on Lipopolysaccharide-Induced Immortalized Human Oral Keratinocytes. Foods 2022, 11, 2199. [Google Scholar] [CrossRef]
  128. Tsalamandris, S.; Antonopoulos, A.S.; Oikonomou, E.; Papamikroulis, G.A.; Vogiatzi, G.; Papaioannou, S.; Deftereos, S.; Tousoulis, D. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur. Cardiol. 2019, 14, 50–59. [Google Scholar] [CrossRef]
  129. Tossetta, G.; Piani, F.; Borghi, C.; Marzioni, D. Role of CD93 in Health and Disease. Cells 2023, 12, 1778. [Google Scholar] [CrossRef]
  130. Piani, F.; Tossetta, G.; Cara-Fuentes, G.; Agnoletti, D.; Marzioni, D.; Borghi, C. Diagnostic and Prognostic Role of CD93 in Cardiovascular Disease: A Systematic Review. Biomolecules 2023, 13, 910. [Google Scholar] [CrossRef]
  131. Jimenez, M.; Hu, F.B.; Marino, M.; Li, Y.; Joshipura, K.J. Type 2 diabetes mellitus and 20 year incidence of periodontitis and tooth loss. Diabetes Res. Clin. Pract. 2012, 98, 494–500. [Google Scholar] [CrossRef] [PubMed]
  132. Bullon, P.; Newman, H.N.; Battino, M. Obesity, diabetes mellitus, atherosclerosis and chronic periodontitis: A shared pathology via oxidative stress and mitochondrial dysfunction? Periodontol. 2000 2014, 64, 139–153. [Google Scholar] [CrossRef] [PubMed]
  133. Li, X.; Sun, X.; Zhang, X.; Mao, Y.; Ji, Y.; Shi, L.; Cai, W.; Wang, P.; Wu, G.; Gan, X.; et al. Enhanced Oxidative Damage and Nrf2 Downregulation Contribute to the Aggravation of Periodontitis by Diabetes Mellitus. Oxidative Med. Cell. Longev. 2018, 2018, 9421019. [Google Scholar] [CrossRef] [PubMed]
  134. Vincent, R.R.; Appukuttan, D.; Victor, D.J.; Balasundaram, A. Oxidative stress in chronic periodontitis patients with type II diabetes mellitus. Eur. J. Dent. 2018, 12, 225–231. [Google Scholar] [CrossRef]
  135. Chen, J.; Wang, Q.; Li, R.; Li, Z.; Jiang, Q.; Yan, F.; Ye, J. The role of sirtuins in the regulatin of oxidative stress during the progress and therapy of type 2 diabetes mellitus. Life Sci. 2023, 333, 122187. [Google Scholar] [CrossRef]
  136. Zhu, C.; Zhao, Y.; Wu, X.; Qiang, C.; Liu, J.; Shi, J.; Gou, J.; Pei, D.; Li, A. The therapeutic role of baicalein in combating experimental periodontitis with diabetes via Nrf2 antioxidant signaling pathway. J. Periodontal Res. 2020, 55, 381–391. [Google Scholar] [CrossRef]
  137. Zhao, T.; Tang, H.; Xie, L.; Zheng, Y.; Ma, Z.; Sun, Q.; Li, X. Scutellaria baicalensis Georgi. (Lamiaceae): A review of its traditional uses, botany, phytochemistry, pharmacology and toxicology. J. Pharm. Pharmacol. 2019, 71, 1353–1369. [Google Scholar] [CrossRef]
  138. Liu, M.; Chen, R.; Xu, Y.; Zheng, J.; Wang, M.; Wang, P. Exosomal miR-141-3p from PDLSCs Alleviates High Glucose-Induced Senescence of PDLSCs by Activating the KEAP1-NRF2 Signaling Pathway. Stem Cells Int. 2023, 2023, 7136819. [Google Scholar] [CrossRef]
  139. Yuan, H.; Xu, Y.; Luo, Y.; Wang, N.X.; Xiao, J.H. Role of Nrf2 in cell senescence regulation. Mol. Cell. Biochem. 2021, 476, 247–259. [Google Scholar] [CrossRef]
  140. Giacco, F.; Brownlee, M. Oxidative stress and diabetic complications. Circ. Res. 2010, 107, 1058–1070. [Google Scholar] [CrossRef]
  141. Akram, Z.; Alqahtani, F.; Alqahtani, M.; Al-Kheraif, A.A.; Javed, F. Levels of advanced glycation end products in gingival crevicular fluid of chronic periodontitis patients with and without type-2 diabetes mellitus. J. Periodontol. 2020, 91, 396–402. [Google Scholar] [CrossRef] [PubMed]
  142. Moldogazieva, N.T.; Mokhosoev, I.M.; Mel’nikova, T.I.; Porozov, Y.B.; Terentiev, A.A. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxidative Med. Cell. Longev. 2019, 2019, 3085756. [Google Scholar] [CrossRef] [PubMed]
  143. Thomas, J.T.; Joseph, B.; Sorsa, T.; Mauramo, M.; Anil, S.; Waltimo, T. Expression of advanced glycation end products and their receptors in diabetic periodontitis patients. Oral Dis. 2023, 30, 2784–2796. [Google Scholar] [CrossRef] [PubMed]
  144. Dong, C.; Hu, X.; Tripathi, A.S. A brief review of vitamin D as a potential target for the regulation of blood glucose and inflammation in diabetes-associated periodontitis. Mol. Cell. Biochem. 2022, 477, 2257–2268. [Google Scholar] [CrossRef]
  145. Liu, C.M.; Chen, S.H.; Liao, Y.W.; Yu, C.H.; Yu, C.C.; Hsieh, P.L. Magnolol ameliorates the accumulation of reactive oxidative stress and inflammation in diabetic periodontitis. J. Formos. Med. Assoc. 2021, 120, 1452–1458. [Google Scholar] [CrossRef]
Figure 1. Pathogenesis of periodontitis. If gingivitis remains unresolved, there is a persistence of pathogens and inflammation that leads to different grades of periodontitis. CAL = clinical attachment loss; PD = probing depth.
Figure 1. Pathogenesis of periodontitis. If gingivitis remains unresolved, there is a persistence of pathogens and inflammation that leads to different grades of periodontitis. CAL = clinical attachment loss; PD = probing depth.
Antioxidants 13 01270 g001
Figure 2. NRF2/KEAP1 signaling regulation. Under the basal condition, NRF2 is bound to the KEAP1/CUL3/RBX1 E3-Ub ligase complex that induces NRF2 proteasomal degradation. Under oxidant stimuli, ROS oxidate the cysteine residues of KEAP1, causing a conformational change that inhibits NRF2 ubiquitination/degradation. Since NRF2 avoids proteasomal degradation, it can migrate into the nucleus and bind ARE regions present in the upstream regulatory region (promoter) of several antioxidant genes, causing their transcription. ARE, antioxidant response element; Cul3, Cullin 3; E3, Ubiquitin ligase 3; KEAP1, Kelch Like ECH Associated Protein 1; NRF2, Nuclear Factor Erythroid 2-Related Factor 2; RBX1, RING box protein 1; Ub, Ubiquitin.
Figure 2. NRF2/KEAP1 signaling regulation. Under the basal condition, NRF2 is bound to the KEAP1/CUL3/RBX1 E3-Ub ligase complex that induces NRF2 proteasomal degradation. Under oxidant stimuli, ROS oxidate the cysteine residues of KEAP1, causing a conformational change that inhibits NRF2 ubiquitination/degradation. Since NRF2 avoids proteasomal degradation, it can migrate into the nucleus and bind ARE regions present in the upstream regulatory region (promoter) of several antioxidant genes, causing their transcription. ARE, antioxidant response element; Cul3, Cullin 3; E3, Ubiquitin ligase 3; KEAP1, Kelch Like ECH Associated Protein 1; NRF2, Nuclear Factor Erythroid 2-Related Factor 2; RBX1, RING box protein 1; Ub, Ubiquitin.
Antioxidants 13 01270 g002
Figure 3. Molecular structures of dehydrocostus lactone and hesperetin. The chemical structures of the compounds illustrated in this table have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).
Figure 3. Molecular structures of dehydrocostus lactone and hesperetin. The chemical structures of the compounds illustrated in this table have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).
Antioxidants 13 01270 g003
Figure 4. Molecular structures of baicalein and magnolol. The chemical structures of the compounds illustrated in this figure have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).
Figure 4. Molecular structures of baicalein and magnolol. The chemical structures of the compounds illustrated in this figure have been taken from ChemSpider free database (https://www.chemspider.com (accessed on 30 September 2024)).
Antioxidants 13 01270 g004
Figure 5. Modulation of NRF2/KEAP1 signaling by phytotherapeutics in periodontitis. Phytotherapeutics (in light blue) increase NRF2 expression, favoring its nuclear translocation and binding to the ARE regions present in the promoter of antioxidant genes (NQO1, CAT, HO-1, SOD, etc.), inducing their transcription. The increased expression of HO-1 inhibits NF-κB activation, thus reducing inflammatory cytokines production (in green). The activation of NRF2 also increases GSH levels, reducing ROS levels and DNA damage (in blue). Moreover, the activation of NRF2 reduces cell senescence, osteoclast differentiation, and alveolar bone loss while favoring osteoblast differentiation, osteogenesis, and periodontal tissue repair (in red). ARE, antioxidant response element; CAT, catalase; GSH, glutathione; HO-1, Heme-oxygenase 1; IL, interleukin; NQO1, NAD(P)H:quinone oxidoreductase; NRF2, Nuclear Factor Erythroid 2-Related Factor 2; SOD, superoxide dismutase; TNF-α, Tumor Necrosis Factor-α.
Figure 5. Modulation of NRF2/KEAP1 signaling by phytotherapeutics in periodontitis. Phytotherapeutics (in light blue) increase NRF2 expression, favoring its nuclear translocation and binding to the ARE regions present in the promoter of antioxidant genes (NQO1, CAT, HO-1, SOD, etc.), inducing their transcription. The increased expression of HO-1 inhibits NF-κB activation, thus reducing inflammatory cytokines production (in green). The activation of NRF2 also increases GSH levels, reducing ROS levels and DNA damage (in blue). Moreover, the activation of NRF2 reduces cell senescence, osteoclast differentiation, and alveolar bone loss while favoring osteoblast differentiation, osteogenesis, and periodontal tissue repair (in red). ARE, antioxidant response element; CAT, catalase; GSH, glutathione; HO-1, Heme-oxygenase 1; IL, interleukin; NQO1, NAD(P)H:quinone oxidoreductase; NRF2, Nuclear Factor Erythroid 2-Related Factor 2; SOD, superoxide dismutase; TNF-α, Tumor Necrosis Factor-α.
Antioxidants 13 01270 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tossetta, G.; Fantone, S.; Togni, L.; Santarelli, A.; Olivieri, F.; Marzioni, D.; Rippo, M.R. Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants 2024, 13, 1270. https://doi.org/10.3390/antiox13101270

AMA Style

Tossetta G, Fantone S, Togni L, Santarelli A, Olivieri F, Marzioni D, Rippo MR. Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants. 2024; 13(10):1270. https://doi.org/10.3390/antiox13101270

Chicago/Turabian Style

Tossetta, Giovanni, Sonia Fantone, Lucrezia Togni, Andrea Santarelli, Fabiola Olivieri, Daniela Marzioni, and Maria Rita Rippo. 2024. "Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis" Antioxidants 13, no. 10: 1270. https://doi.org/10.3390/antiox13101270

APA Style

Tossetta, G., Fantone, S., Togni, L., Santarelli, A., Olivieri, F., Marzioni, D., & Rippo, M. R. (2024). Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants, 13(10), 1270. https://doi.org/10.3390/antiox13101270

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