You are on page 1of 9

Archives of Oral Biology 105 (2019) 72–80

Contents lists available at ScienceDirect

Archives of Oral Biology


journal homepage: www.elsevier.com/locate/archoralbio

Host modulation therapy using anti-inflammatory and antioxidant agents in T


periodontitis: A review to a clinical translation

Benso Sulijayaa,b,d, Naoki Takahashib,c, Kazuhisa Yamazakia,
a
Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata,
Japan
b
Division of Periodontology, Department of Oral Biological Science, Niigata University Faculty of Dentistry, Niigata, Japan
c
Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
d
Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia

A R T I C LE I N FO A B S T R A C T

Keywords: Objective: To highlight the shifting paradigm of periodontitis, describe mechanism of periodontal bone de-
Host modulation struction, and propose an updated host modulation therapy (HMT) strategy. To add further clinical relevance,
Periodontitis related studies investigating the efficacy of several HMT agents in periodontitis will be discussed.
Anti-inflammation Design: Literature searches were conducted from articles published in PubMed using keywords “periodontal
Antioxidant
disease AND periodontitis AND host modulation therapy AND anti-inflammatory AND antioxidant”, and then the
Therapy
findings were comprehensively summarized and elaborated.
Result: Accumulating evidence indicates that periodontitis is no longer defined solely as a pathogen-induced
disease; rather, it is now recognized as a consequence of uncontrolled immune response and oxidative stress
leading to periodontal tissue damage. Although periodontopathic bacteria initiate the disease, inflammation and
oxidative stress were reported to be the main causes for the severity of tissue destruction. Thus, since the concept
of periodontitis has shifted, our approach to its management needs to be adjusted to accommodate the latest
paradigm. Nowadays, the modulation of inflammation and oxidative stress is considered a target of HMT. HMT
agents, such as probiotics, anti-inflammatory drugs, anti-chemokines, lipid mediators, and bio-active fatty acids,
have been extensively investigated for their remarkable functions in modulating the immune response and
providing antioxidant effects.
Conclusion: Findings from in vitro, in vivo, and human studies frequently demonstrate positive association by the
administration of HMT in periodontitis. HMT strategy targeted on anti-inflammatory and antioxidant in peri-
odontitis might serve as an excellent therapeutic approach to reach the level of clinical benefit.

1. Introduction 2004; Sanz et al., 2017). Inflammation might also be caused by the
alteration in bacterial composition, dysbiosis, and oxidative stress
The concept that dental plaque is necessary but not sufficient to conditions (Herrero et al., 2018; Shi et al., 2015; Wang et al., 2014).
cause periodontitis progression has led to a shift in strategy for peri- During pathogenesis, pro-inflammatory cytokines and reactive
odontal therapy from anti-infective approaches to host modulation oxygen species (ROS) may act as double-edged swords; they are re-
(Bartold & Van Dyke, 2017; Van Dyke, 2008a). Besides the bacterial quired not only to keep bacteria outside the tissues but also the ex-
challenge, several studies have revealed that uncontrolled production aggerated response of these molecules can damage the tissue (Li et al.,
of pro-inflammatory cytokines lead to major pathogenicity and tissue 2004; Schieber & Chandel, 2014; Tomofuji et al., 2009). Thus, in-
destruction (Bartold & Van Dyke, 2013, 2017). The number of bacteria vestigations into the significance of clinical results and unwanted side
does not always correlate with the severity of this destruction in clinical effects of using host modulation therapy (HMT) agents in periodontitis
studies; however, the contribution from the initiating bacteria, in- management have been reported (Elkhouli, 2011; Preshaw et al., 2004;
cluding their putative factors that trigger the host’s immune response, is Preshaw, 2018; Reddy, Geurs, & Gunsolley, 2003). In this review, we
indisputable (Cekici, Kantarci, Hasturk, & Van Dyke, 2014; Marsh, highlighted the mechanism of periodontal bone destruction by


Corresponding author at: Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of
Medical and Dental Sciences 5274 Gakkocho 2-ban-cho, Chuo-ku, Niigata 951-8514, Japan.
E-mail address: kaz@dent.niigata-u.ac.jp (K. Yamazaki).

https://doi.org/10.1016/j.archoralbio.2019.07.002
Received 13 May 2019; Received in revised form 26 June 2019; Accepted 2 July 2019
0003-9969/ © 2019 Elsevier Ltd. All rights reserved.
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

inflammation and ROS, and the management strategies attempting to tissue (Guyodo et al., 2012). Considering this finding, it might be as-
modulate the immune response. Moreover, based on the relevant sumed that bacteria and their products are simply the initiators; they do
findings and our investigative results, we outline the current concept of not even have a chance to reach the underlying connective tissue where
HMT. Finally, the clinical objective of this review is to guide clinicians the destruction occurs.
and researchers towards better insight into the plausible mechanisms of Related to the bacteria, dental plaque contributes only 20% of the
inflammatory damage, a clear understanding of which will allow for risk of developing periodontitis which leads to the assumption that
targeted HMT and will improve periodontal therapy outcomes. ‘periodontal pathogens’ appear as a result of the disease rather than a
cause (Bartold & Van Dyke, 2017; Marsh, 2003). In brief, for period-
2. Material and methods ontitis, there is convincing longitudinal data confirming that bacteria
are necessary but not sufficient for disease to develop; the uncontrolled
Literature searches were conducted by 2 reviewers (BS and NT), inflammation and immune response are responsible for bone resorption
independently. Articles published in PubMed within the past 10 years (Kornman et al., 1991; Socransky & Haffajee, 1992; Van Dyke, 2008a,
with full text written in English available were included. Keywords used 2008b).
were “periodontal disease AND periodontitis AND host modulation Another current hypothesis has been introduced with great interest,
therapy AND anti-inflammatory AND antioxidant”. Paper selection and known as the polymicrobial synergy and dysbiosis theory. This model
data extraction were performed by aforementioned reviewers. Also, stresses that the host response is initially triggered by bacteria with
additional articles were manually searched and included from citations pathogenic potential; then, accessory bacteria assist in the colonization
of the selected papers. We focused on the articles describing: 1) pa- and metabolic activities until later when they are outgrown by the over-
thogenesis of periodontitis and the conceptual shifting, 2) inflammation activated pathobionts leading to destructive inflammation (Darveau,
and oxidative stress in periodontitis, 3) HMT development and its Hajishengallis, & Curtis, 2012; Hajishengallis & Lamont, 2012;
agents, and 4) plausible HMT approach mechanism. Based on article Hajishengallis, Darveau, & Curtis, 2012; Hajishengallis, 2015; Lamont
abstracts, irrelevant topics, and non-English written articles were ex- & Hajishengallis, 2015). Most likely there is no specific series of bac-
cluded. Further evidence and findings reported were discussed in the terial species that exclusively act as such; there may be individual dif-
form of narrative review. ferences in biofilm composition and individual variation on how the
host responses to these bacterial communities. Thus, period-
3. Results ontopathogenic bacteria encompass a much wider range of bacterial
species than hitherto assumed. Moreover, host susceptibility is influ-
3.1. The current concept of the pathogenesis of periodontitis enced by various factors, such as genetic, epigenetic, and life style
factors (smoking, stress, and diet), as well as the presence of systemic
Bacteria play an undeniable role in the etiology of periodontal disease (e.g. diabetes and aging) (Hajishengallis & Lamont, 2012;
disease, including gingivitis and periodontitis (Socransky & Haffajee, Lamont & Hajishengallis, 2015; Rinuchitra, Govindarajan, Thangavelu,
1992). In fact, almost all bacterial species harbor pathogenic properties & Jayaraman, 2012). In summary, periodontitis is no longer defined as
when they are allowed to expand and when they take over the normal a solely bacteria-caused disease, rather it is now seen as an uncontrolled
microbiota. In 1965, the nonspecific plaque hypothesis was described immune response-related disease caused by dysbiosis; therefore, its
as the amount of the entire dental-plaque microbiota correlating with management should include the modulation of host immunity, which
disease status (Löe, Theilade, & Jensen, 1965). This hypothesis might be can lead to the restoration of tissue homeostasis.
acceptable in gingivitis; however, in periodontitis, further interaction
between bacterial by-products and the immune system are somehow 3.2. Inflammation and oxidative stress in periodontitis
absent from this hypothesis. Another hypothesis in the late 1970s de-
scribed how the presence or increase of specific pathogenic bacteria in Biological processes including inflammation and oxidative stress at
sub-gingiva and their toxic products were associated with periodontitis the level of cellular state may further contribute to the clinical re-
(Theilade, 1986). Moreover, the microbial shift hypothesis, known as levance of the onset and development of periodontitis (Bullon, 2014).
dysbiosis, reinforced the previous hypothesis, which confirms that the Inflammation and oxidative stress conditions occurring in periodontitis
change from predominantly Gram-positive aerobic bacteria to groups of and their role in the mechanism of alveolar bone destruction has been
Gram-negative anaerobes (pathogenic flora) is consistent with disease explored by many authors (D’Aiuto et al., 2010; Liu, Lerner, & Teng,
(Berezow & Darveau, 2011). In the 1990s, these hypotheses were ad- 2010; Schieber & Chandel, 2014; Tomofuji et al., 2009). Periodontal
justed to become the ecological plaque hypothesis, in which the en- tissue becomes inflamed after the bacterial components and metabolic
vironmental changes in the gingival sulcus mediate the growth of products of bacteria trigger the junctional epithelium cells to produce
Gram-negative and proteolytic species of bacteria, resulting in further cytokines, thus stimulating neurons to produce neuropeptides that
inflammation and tissue destruction (Marsh, 2004). In fact, pathogen- cause vasodilatation of peripheral blood vessels (Gaffen &
esis is initiated by the oral microbial biofilm; the inflammation reacts to Hajishengallis, 2008). Following this event, an increased number of
the biofilm and drives the destruction of the periodontium (Sima & polymorphonuclear neutrophils (PMNs), macrophages, lymphocytes,
Glogauer, 2014; Van Dyke, 2008b) as well as a change in microbes plasma cells, and mast cells are activated and accumulated in the
(Marsh, 2004). Thus, it is assumed that periodontitis is more likely a connective tissue (Cekici et al., 2014). In the host response to these
non-resolving inflammation that is ineffective in eliminating the in- bacterial components and metabolites, the innate immune response is
itiating pathogens and maintains constant inflammatory conditions dominated by the phagocytic activities of macrophages and neu-
(Gaffen & Hajishengallis, 2008). trophils, whereas plasma cells, T lymphocytes, and B lymphocytes
In addition to this hypothesis, an in vivo study has shown that (IgG1 and IgG3 subclasses) are dominant in the transition to the
Porphyromonas gingivalis (P. gingivalis), one of the red complex bacteria, adaptive immune stage (Janeway & Medzhitov, 2002; Medzhitov &
failed to induce periodontal tissue breakdown and alveolar bone de- Janeway, 1997). Extracted T-cells from periodontitis lesions exhibit a
struction in germ-free mice (Hajishengallis et al., 2011). However, in reduced response to stimuli; however, this can be restored with peri-
specific pathogen-free mice, the inoculation of P. gingivalis triggered odontal therapy (Petit et al., 2001). Later, activation of the tumor ne-
compositional changes to oral bacteria, thus resulting in alveolar bone crosis factor (TNF) family and their receptors activates B-cell produc-
loss (Hajishengallis et al., 2011). Also, surprising evidence revealed that tion, which marks the maturation of the antibody response (Cekici
in advanced periodontitis lesions, the invasion of P. gingivalis reached et al., 2014). Hence, both the innate and adaptive immune systems are
only the epithelial cells and did not extend to the underlying connective coordinately activated in the inflammatory response to

73
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

periodontopathic bacteria. also cause tissue damage via oxidation processes (Chiu, Saigh,
In immune cells within the oral epithelium, bacterial lipopoly- McCulloch, & Glogauer, 2017). An in vitro study using human gingival
saccharide (LPS) binds to toll-like receptors 2 and 4 (TLR2 and TLR4) fibroblast challenged with P. gingivalis LPS demonstrated higher level of
(Hatakeyama et al., 2003; Pihlstrom, Michalowicz, & Johnson, 2005) ROS followed with mitochondrial dysfunction by a reduction in mi-
and activates the myeloid differentiation primary response protein tochondrial protein expression, mass, and membrane potential (Bullon
MYD88 as well as the toll/interleukin-1 (IL-1) receptor adaptor protein, et al., 2011). On the other word, the excessive ROS production affects to
resulting in the production of TNF-α, IL-6 and IL-12 (Cook, Pisetsky, & mitochondrial dysfunction thereby promoting cell apoptosis and com-
Schwartz, 2004; Kaisho & Akira, 2002; Watters, Kenny, & O’Neill, plex signaling pathways (Dirkx, Schwenk, Glatz, Luiken, & van Eys,
2007). In addition, the nuclear factor κB (NF-κB) complex has been 2011). These findings suggest that mitochondria status is also a critical
investigated as a key transcription factor that regulates immune re- point in the initiation of inflammation and oxidative stress at the level
sponses to bacteria and viruses, inflammation, developmental aspects, of cell.
and cell proliferation (Smale, 2011) through the TLR pathway The activation of matrix metalloproteinases, involved in tissue,
(Baldwin, 1996; Hanada & Yoshimura, 2002). In brief, the recognition plasma, and bacterial proteinase activity, also reportedly increased by
of bacteria by innate immune cells results in the production of in- oxidative stress (Murphy & Reiner, 2002; Vandooren, Van Den Steen, &
flammatory cytokines including TNF-α, IL-1β, and IL-6 (Garlet, 2010). Opdenakker, 2013). Clinical investigations have found that patients
Thus, TNF-α is also associated with bone resorption by promoting the with hyperactive ROS responses are more susceptible to periodontitis
production of RANKL and secretion of matrix metalloproteinases de- than those with normal responses (Aboodi et al., 2011; Johnstone, Koh,
grading periodontal tissue (Garlet, Martins, Fonseca, Ferreira, & Silva, Goldberg, & Glogauer, 2007). Moreover, the inhibition of antioxidant
2004; Graves & Cochran, 2003; Graves, Fine, Teng, Van Dyke, & production, by down-regulation of the nuclear factor erythroid 2-re-
Hajishengallis, 2008; Hadler-Olsen, Fadnes, Sylte, Uhlin-Hansen, & lated factor 2 (NRF2) pathway in oral polymorphonuclear neutrophils,
Winberg, 2011). TNF-α induces immune cell migration by increasing is associated with severe periodontitis (Sima et al., 2016). Hence, the
the adhesion of neutrophils and stimulating the production of cytokines NRF2 and NF-κB pathways are implicated to be linked in the patho-
IL-6 (Garlet et al., 2007; Graves et al., 2008; Steeve, Marc, Sandrine, genesis of bone destruction (Wardyn, Ponsford, & Sanderson, 2015);
Dominique, & Yannick, 2004) and IL-1β (Kindle, Rothe, Kriss, Osdoby, NF-κB stimulates an increase in mitochondrial activity and the ex-
& Collin-Osdoby, 2006; Mohammed, Ferzandi, & Hamilton, 2010; pression of NADPH oxidase, which are both main sources of en-
Rajput & Ware, 2015). The mechanism of periodontal tissue destruction dogenous free radical ROS (Mauro et al., 2011). In addition, knocked-
mediated by inflammation through the TLR-mediated NF-κB pathway is down NRF2 in macrophages reduces TNF-α, inducible nitric oxide
thus well established. synthase (iNOS), cyclooxygenase 2 (COX2), and IL-1β production in
Besides, inflammasome, a multiprotein complex located in the cy- response to LPS (Lin et al., 2008). Therefore, apart from the in-
toplasm of the cell, is responsible for the maturation of pro-in- flammation, periodontitis is partly triggered by over-production of ROS
flammatory cytokines such as IL-1β and IL-18, also for the activation of through NRF2, which leads to oxidative stress and vice versa. Regulation
inflammatory cell death called pyroptosis (Olsen & Yilmaz, 2016). of ROS is important to maintain homeostasis.
Likewise, IL-1β mediated by inflammasome activity has been reported In the sense of gaining the homeostasis, a supporting data have been
to associate with the initiation, onset, and progression of periodontitis interestingly showed the involvement of autophagy in the pathogenesis
(Aoki‐Nonaka et al., 2019; Lukens et al., 2014; Netea, van de Veerdonk, of periodontitis. Peripheral blood mononuclear cells from periodontitis
van der Meer, Dinarello, & Joosten, 2014; Shibata, 2018). Pathogen- patients demonstrated higher expression of autophagy gene that posi-
associated molecular patterns (PAMPs) are recognized by TLRs and tively correlated with high levels of mitochondrial ROS production
stimulate inflammasome-driven inflammation throughout the upregu- (Bullon et al., 2012). Additionally, in human gingival fibroblast cells
lation of nucleotide-binding domain-like receptor protein 3 (NLRP3) in treated with P. gingivalis LPS, the protein level and gene expression of
an NF-κB-dependent manner, thereby cleaving pro-IL-1β into an active autophagy-related protein 12 (ATG12) was increased (Bullon et al.,
form of IL-1β with the help of caspase-1 (Olsen & Yilmaz, 2016; 2012). A reduction of mitochondrial ROS reduces autophagy activity,
Yamaguchi, Kurita-Ochiai, Kobayashi, Suzuki, & Ando, 2017). Clinical while inhibition of autophagy increases cell apoptosis (Bullon, 2014).
study has been reported the higher levels of inflammasome components Altogether, this might suggest the protective role of autophagy in per-
(e.g. NLRP3 and absent in melanoma 2 (AIM2)) in the gingival tissues iodontitis (e.g. eliminate metabolic debris).
from chronic periodontitis patients than healthy controls (Park et al., These processes can lead to changes in the local environment. In the
2014). Further, an in vitro study using THP-1 cells challenged with live dysbiosis theory, the influence of individual species in a polymicrobial
P. gingivalis demonstrated higher secretion of IL-1β followed with the community could alter host-microbial interactions leading to destruc-
activation of some inflammasomes (NRLP3 and AIM2) via the re- tive inflammation (Hajishengallis et al., 2012). In brief, non-pathogenic
cognition of TLR2/4 and AIM2-cytosolic bacterial double-stranded DNA bacteria known as accessory pathogens could further exacerbate in-
(dsDNA) (Park et al., 2014). These findings suggest that period- flammation by producing nutritional products from the end products of
ontopathic bacteria can modify innate immunity by affecting in- tissue destruction (e.g. degraded collagen peptides and heme-containing
flammasome activity; however, this in vitro study where cells are compounds), that help the other bacteria to flourish (Hajishengallis,
brought in contact with whole/live bacteria might be not realistic based 2014). Moving forward, it is worth noting that each factor contributes
on the evidence found that bacteria were not found in the deeper tis- to this complex mechanism wherein synergistic effects from various
sues. factors may occur and lead to greater destruction.
Oxidative stress is performed by the cells not only to produce energy
needed for physiological activity but also to destroy bacteria and their 3.3. Host modulation therapy
products; this is known as defense mechanism by the cells (Bullon,
2014). The excessive endogenous production of reactive oxygen species As uncontrolled products of the immune system and oxidative stress
(ROS) is likely to contribute to periodontal tissue breakdown, as their both drive the breakdown of periodontal tissue, we propose a mod-
levels are increased in lesions (Aboodi, Goldberg, & Glogauer, 2011; ification to HMT therapeutic strategies which was mainly addressed to
Battino, Bullon, Wilson, & Newman, 1999). In brief, when exposed to modulate the level of inflammation (Bartold & Dyke, 2017). We have
the pathogen and its components, PMNs activate oxidative mechanisms observed that three major components are involved in the pathogenesis
that kill phagocytosed bacteria and in doing so, produce ROS including of periodontitis, including periodontal pathogen bacteria, immune re-
superoxide radicals, hydrogen peroxide, hydroxyl radicals, and hypo- sponse, and periodontal tissue destruction induced by ROS.
chlorous acid. Beside killing pathogenic bacteria, those ROS molecules Consequently, the possible way to modulate bacteria is by anti-bacterial

74
B. Sulijaya, et al.

Table 1
Studies regarding the association between periodontitis and the use of HMT agents.
Category Author (year) Study design Treatment Main outcome

Probiotics Morales et al. (2018) (Morales et al., 2018) RCT Lactobacillus rhamnosus SP1 Lactobacillus rhamnosus SP1 administration showed similar clinical and microbiological
improvements compared to baseline
Iwasaki et al. (2016) (Iwasaki et al., 2016) RCT Heat-killed Lactobacillus plantarum L-137 Significant greater pocket depth reduction was found in test group compared to control.
(HK L-137) Daily consumption of HK L-137 decreased periodontal pocket depth in patients undergoing
SPT
Laleman et al. (2015) (Laleman et al., 2015) RCT Streptococcus oralis KJ3, Streptococcus No differences were detected of the adjunctive use of streptococci containing tablet after
uberis KJ2, Streptococcus rattus JH145 SRP
Teughels, Durukan, Ozcelik, Pauwels, Quirynen, Cenk Haytac RCT Lactobacillus reuteri Oral administration of Lactobacillus reuteri significantly enhanced the pocket depth
(2013) (Teughels, Durukan, Ozcelik, Pauwels, Quirynen, Haytac reduction, attachment gain and the reduction of P. gingivalis
et al., 2013)
Vicario et al. (2013) (Vicario et al., 2013) RCT Lactobacillus reuteri Prodentis Lactobacillus reuteri Prodentis improved the short-term clinical outcomes (plaque index,
bleeding on probing and probing depth) in patients with mild-moderate chronic
periodontitis
Mayanagi et al. (2009) (Mayanagi et al., 2009) RCT Lactobacillus salivarius WB21 Oral administration of Lactobacillus salivarius WB21 reduced the amount of
periodontopathic bacteria
Anti-chemokine Glowacki et al. (2013) (Glowacki et al., 2013) In vivo Anti-CCL22 Application anti-CCL22 in ligature-induced periodontitis in beagle dogs reduced the
inflammation and alveolar bone loss
Lipid mediators Hasturk et al. (2007) (Hasturk et al., 2007) In vivo and in Resolvin E1 In periodontitis rabbit model, Resolvin E1 significantly reduced bone loss and systemic

75
vitro CRP and IL-1β levels
Lee et al. (2016) (Lee et al., 2016) In vivo Resolvin E1 Resolvin E1 prevented bone loss in ligature induced periodontitis, reduced osteoclast
density, suppressed inflammation-related genes
Gao et al. (2013) (Gao et al., 2013) In vivo and in Resolvin E1 Resolvin E1 modulates osteoclast differentiation and bone remodeling by inducing OPG
vitro thus restore the favorable RANKL/OPG ratio
Fredman et al. (2011) (Fredman et al., 2011) In vitro Resolvin E1 Resolvin E1 (0.1-100nM) rescues the impaired phagocytic activity of localized aggressive
periodontitis-macrophages
Yamada et al. (2018) (Yamada et al., 2018) In vivo and in HYA HYA significantly improved the epithelial barrier junction on the gingival epithelial cell
vitro
Sulijaya et al. (2018) (Sulijaya et al., 2018) In vitro KetoC KetoC suppressed inflammation induced by P. gingivalis LPS through NF-κB by binding to G
protein-coupled receptor 120
Yang et al. (2017) (Yang et al., 2017) In vitro KetoC KetoC reduced pro-inflammatory cytokines via NF-κB and mitogen-activated protein
kinase/activator protein-1
Matsuda et al. (2018) (Matsuda et al., 2018) In vivo and In Resveratrol Resveratrol significantly suppressed bone loss and the expression of osteoclastogenesis-
vitro related gene in periodontal tissue
Furumoto et al. (2016) (Furumoto et al., 2016) In vitro KetoC KetoC increased antioxidant genes by upregulating the NRF2-ARE pathway
Ashour et al. (2016) (Ashour et al., 2016) RCT Cytoflavin The use of Cytoflavin showed positive impact (reduction of Russell periodontal index
45.3% and Green-Vermillion index by 39.8%)
Tamaki et al. (2014) (Tamaki et al., 2014) In vitro Resveratrol Resveratrol relieved alveolar bone resorption and activated the Sirt1/AMPK and the
NRF2/antioxidant pathways in inflamed gingival tissue.

RCT: Randomized Clinical Trial.


Archives of Oral Biology 105 (2019) 72–80
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

drugs, probiotic intake and mechanical debridement. Host modulation periodontitis rabbit model using ligatures inoculated with P. gingivalis,
therapy based on anti-cytokine/chemokine and antioxidant agents are resolvin E1 not only suppressed bone loss but also restored the systemic
used to modulate the over-production of the immune system (Freire & C-reactive protein (CRP) and IL-1β levels (Hasturk et al., 2007). Re-
Dyke, 2013; Preshaw, 2018; Yanine et al., 2013; Yen et al., 2008). In solvin E1 also recovered the impaired phagocytosis activity of macro-
addition, the anti-inflammatory drugs and gingival epithelial barrier phages collected from humans with localized aggressive periodontitis
junction improver are used to modulate the destruction of the tissue (Fredman et al., 2011). Furthermore, Chemokine-like receptor 1
(Sulijaya et al., 2018; Yamada et al., 2018). On top of that, the sus- (ChemR23) and BLT1 (a leukotriene receptor) have been investigated
ceptibility of the host is affecting the outcome of those approaches. as receptors of resolvin E1 that mediate its inflammation-related action
As a non-invasive therapeutic approach for periodontitis, host on monocytes, macrophages, dendritic cells, osteoblasts, and osteo-
modulation therapy has been investigated and explored in the last three clasts (Arita, Bianchini et al., 2005; Arita, Yoshida et al., 2005; Arita
decades (Preshaw, 2018). It has focused on several approaches as pre- et al., 2007; El Kebir, Gjorstrup, & Filep, 2012; Freire, Dalli, Serhan, &
sented in Table 1. Interestingly, many studies have tried to investigate Van Dyke, 2017). ChemR23 acts as a chemoattractant and as an adi-
the benefit of using anti-inflammatory and antioxidant-targeted drugs pokine (Bondue, Wittamer, & Parmentier, 2011), whereas BLT1 is
(Ashour et al., 2016; Fredman et al., 2011; Furumoto et al., 2016; Gao highly associated with leukotriene B4 (LTB4), a pro-inflammatory
et al., 2013; Glowacki et al., 2013; Hasturk et al., 2007; Iwasaki et al., mediator (Arita et al., 2007). Therefore, it is proposed that via binding
2016; Laleman et al., 2015; Lee et al., 2016; Matsuda, Minagawa, Okui, to ChemR23, resolvin E1 induces phagocytosis of apoptotic PMNs and
& Yamazaki, 2018; Mayanagi et al., 2009; Morales et al., 2018; Sulijaya inhibits inflammation. Also, by binding to BLT1, resolvin E1 competes
et al., 2018; Tamaki et al., 2014; Teughels, Durukan, Ozcelik, Pauwels, with LTB4 thus attenuating the inflammatory signal. Finally, with no
Quirynen, Cenk Haytac, 2013; Vicario, Santos, Violant, Nart, & Giner, unwanted side effects having been reported, these consistent results
2013; Yamada et al., 2018; Yang et al., 2017). In addition, nutrition has bring about the possibility of this compound to be applied in humans.
been reviewed to possess beneficial modulation effects in adjunct to Hence, to the best of our knowledge, no clinical studies on humans have
periodontal therapy in periodontitis management (Sulijaya et al., evaluated the effect of this compound on periodontitis.
2019). In the following paragraphs, several approaches including anti- Resveratrol, an inhibitor of bone resorption-related factors, has been
chemokine, lipid mediator, bone resorption-related factor inhibitor, and shown to have cardioprotective and anti-inflammatory effects
bio-active fatty acid are discussed. (Minagawa et al., 2015), antioxidant properties (Orihuela-Campos
Chemokines are chemotactic cytokines that attract phagocytic cells et al., 2015), and the capacity to inhibit osteoclast differentiation
to the challenged site once the cells have migrated from the blood (Matsuda et al., 2018). In a ligature-induced periodontitis rat model,
vessel. In addition, chemokines provide messages that influence other the administration of Resveratrol induced production of Sirt1/AMPK
biological processes including proliferation, apoptosis, tumor metas- and NFR2 proteins, thus inhibiting the NF-κB/mitogen-activated pro-
tasis, and host defense (Zlotnik & Yoshie, 2000). CCL22 has been as- tein kinase (MAPK) pathway, resulting in the improvement of systemic
sociated with the reduction of periodontitis severity (Glowacki et al., inflammation and oxidative stress (Tamaki et al., 2014). In human
2013). The blockade of CCL22 leads to higher bone resorption, in- gingival epithelial cells, Resveratrol suppresses IL-1β, IL-8, and mono-
flammatory cell counts, and increased levels of TNF-α and RANKL. cyte chemoattractant-1 (MCP-1) in a Sirt1-independent manner
Moreover, it has been shown that anti-CCL22 treatment resulted in a (Minagawa et al., 2015). Aside from that, it promotes heme oxygenase 1
significant decrease in the number of Treg cells (CD4+FOXp3+) and (HO-1), an antioxidant enzyme, through the upregulation of NRF2
inflammatory bone loss through CCR4 by inhibiting the early migration pathway. Thus, mediated by carbon monoxide (CO), HO-1 can suppress
of Tregs (Araujo-Pires et al., 2015), whereas the migration of in- oxidative stress by down-regulating iNOS, an oxidant gene (Datta,
flammatory cells is highly observed in the disease lesion site (Garlet Koukouritaki, Hopp, & Lianos, 1999; Thirunavukkarasu et al., 2007).
et al., 2006). This result suggests that the inhibition of CCR4-mediated Summarizing these findings, it is accepted that Resveratrol has multi-
migrating inflammatory cells by anti-CCL22 treatment might be bene- functional and beneficial effects as an HMT targeted to inflammatory
ficial to attenuate periodontitis due to its ability to prevent in- genes, osteoclast differentiation, and oxidant-related genes.
flammatory cell migration and suppress the inflammatory cytokines Several studies have been performed to investigate the effect of
and osteoclastogenic mediators. In other words, this chemokine re- polyunsaturated fatty acids (PUFAs) on periodontitis. Omega-3 (ω-3),
ceptor seems to have potential as a target for HMT to reduce in- one of the PUFAs, showed anti-inflammatory activity by reducing the
flammatory and bone-resorptive processes. Regarding lipid mediators, synthesis of IL-1β and TNF-α in in vitro studies (Caughey, Mantzioris,
resolvins have been reported to be induced and produced endogenously Gibson, Cleland, & James, 1996; Meydani et al., 1991). In clinical
during the resolution phase of inflammation. They are synthesized from studies, supplementation with omega-3 (ω-3) for 21 days failed to re-
Omega-3 fatty acids especially docosahexaenoic acid for D-series and duce levels of gingivitis (Campan, 1997). In contrast, a clinical trial
eicosapentaenoic acid for E-series resolvins. Both of these series have study using ω-3 as an adjunctive therapy to scaling and root planing
shown anti-inflammatory and pro-resolution functions (Freire & Van (SRP) in periodontitis subjects, showed significant reduction on gin-
Dyke, 2013). The D-series resolvins (resolvin D2) have been shown to gival index, probing depth, and gain in clinical attachment level com-
suppress inflammation by reducing platelet-leukocyte adhesion and the pared to control (Deore et al., 2014). Furthermore, several studies
combination with aspirin accelerates pro-resolution activities (Serhan & found positive improvement on periodontal parameters by using a
Savill, 2005). combination of PUFAs with aspirin (El-Sharkawy et al., 2010; Naqvi
Previous studies have also investigated the role of resolvin E1 in et al., 2014). However, prolonged bleeding time that may be a result
inhibiting osteoclast differentiation, increasing bone formation through from impaired blood vessels or decreased number of thrombocytes was
osteoprotegerin (OPG) production (Gao et al., 2013), enhancing the reported as a systemic adverse effect of ω-3 dietary supplementation
phagocytic activity of apoptotic PMN, and reducing ROS production. In (Dyerberg & Bang, 1979). Several investigations later disproved the
addition, restoring bone remodeling (as indicated by the OPG:RANKL association between prolonged bleeding time and high dietary doses of
ratio) brings inflammation back to homeostasis (Balta, Loos, & Nicu, ω-3 (Eritsland, Arnesen, Seljeflot, & Høstmark, 1995; Wachira, Larson,
2017). Local treatment with resolvin E1 solution on ligature-induced & Harris, 2014). Therefore, the adjunctive use of ω-3 in periodontal
periodontitis in a rat model showed suppression of inflammatory cell therapy affects hemostasis without increasing the risk of bleeding.
infiltration as well as inflammation-related genes (Lee et al., 2016). Recently, another PUFA generated by Lactobacillus plantarum has
Resolvin E1 also reduced PMN infiltration by about 50% (Arita, been widely investigated due to its potential effects. The PUFAs KetoC
Bianchini et al., 2005; Arita, Yoshida et al., 2005) and down-regulated and HYA have been shown to have beneficial effects in the context of
leukocyte rolling by about 40% (Dona et al., 2008). In an experimental inflammatory disease, especially for periodontitis. KetoC was shown to

76
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

have an anti-inflammatory effect on TNF-α production through the NF- e.g. anti-inflammatory and anti-oxidative properties. However, limited
κB/G protein-coupled receptor-120 pathway by treating macrophages studies have investigated HMT in humans and the possible side effects
with LPS derived from P. gingivalis (Sulijaya et al., 2018); moreover, it of HMT agents need to be further clarified. In this sense, future clinical
was also reported that this suppression effect was mediated by MAPK/ investigations are required to support the remarkable role of HMT
activator protein-1 (Yang et al., 2017). KetoC also showed some po- agents in the management of periodontitis.
tential antioxidant properties by up-regulating the HO-1 and NQO1
genes through the NRF2 pathway in HepG2 cells (Furumoto et al., 5. Contributors
2016), macrophages (Sulijaya et al. unpublished data), and epithelial
cells (Yokoji et al. unpublished data). Regarding osteoclastogenesis, All authors have read and approved the final article.
KetoC has no significant effect on osteoclast differentiation-related
genes, e.g. RANKL and OPG (Sulijaya et al. unpublished data). Mean- Acknowledgements
while, HYA is reported to have a protective role in epithelial cells by
inducing E-cadherin (E-cad), a gingival epithelial barrier junction This work was financially supported in part by Japan Society for the
component (Yamada et al., 2018). Bringing these results together, it Promotion of Science (JSPS) Grants-in-Aid for Scientific Research
may be assumed that gut metabolites suppress inflammatory cytokines (KAKENHI) (Grant numbers 15H02578 (to K.Y.) and 16H05554 (to T.
and induce oxidant genes in addition to improving the epithelial barrier N.)), located in Japan.
junction, thus resolving inflammation and oxidative stress.
Concerning the disadvantages of HMT agents, several studies have References
been performed. For example, even though consumption of oral pro-
biotics is generally safe (Chatterjee, Bhattacharya, & Kandwal, 2011), Aboodi, G. M., Goldberg, M. B., & Glogauer, M. (2011). Refractory periodontitis popu-
cases of probiotics-related bacteremia, Lactobacillus endocarditis, and lation characterized by a hyperactive oral neutrophil phenotype. Journal of
Periodontology, 82(5), 726–733. https://doi.org/10.1902/jop.2010.100508.
secondary liver abscesses have been reported from Lactobacillus rham- Alani, A., & Seymour, R. (2014). Systemic medication and the inflammatory cascade.
nosus (Meurman & Stamatova, 2007). Targeted inhibition of pros- Periodontology 2000, 64(1), 198–210. https://doi.org/10.1111/j.1600-0757.2012.
taglandins and leukotrienes by corticosteroids has shown a variety of 00454.x.
Aoki‐Nonaka, Y., Tabeta, K., Yokoji, M., Matsugishi, A., Matsuda, Y., Takahashi, N., ...
side effects including osteoporosis, peptic ulceration, inhibition of Yamazaki, K. (2019). The AmyI‐1‐18 peptide derived from rice inhibits alveolar bone
adrenal cortex function, hypertension, and impaired wound healing resorption via suppression of inflammatory cytokine production induced by lipopo-
(Alani & Seymour, 2014). Bisphosphonates, which are often used to lysaccharide and interleukin‐1β in mice. Journal of Periodontology, 1–10. https://doi.
org/10.1002/jper.18-0630.
treat osteoporosis and other bone-resorptive related diseases, have been
Araujo-Pires, A. C., Vieira, A. E., Francisconi, C. F., Biguetti, C. C., Glowacki, A.,
shown to potentially induce osteonecrosis (Otomo-Corgel, 2012). As Yoshizawa, S., ... Garlet, G. P. (2015). IL-4/CCL22/CCR4 axis controls regulatory T-
mentioned earlier, PUFAs, and especially ω-3, have been reported to cell migration that suppresses inflammatory bone loss in murine experimental peri-
odontitis. Journal of Bone and Mineral Research, 30(3), 400–410. https://doi.org/10.
increase bleeding time, but this side effect has since been disproven
1002/jbmr.2376.
(Eritsland et al., 1995; Wachira et al., 2014). Arita, M., Ohira, T., Sun, Y.-P., Elangovan, S., Chiang, N., & Serhan, C. N. (2007). Resolvin
Furthermore, the use of low dose doxycycline (SDD; 20 mg, sub anti- E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate
microbial dose) twice a day for a minimum of 3 months has been in- inflammation. The Journal of Immunology, 178(6), 3912–3917. https://doi.org/10.
4049/jimmunol.178.6.3912.
dicated to be safe and can improve clinical outcomes; however, anti- Arita, M., Yoshida, M., Hong, S., Tjonahen, E., Glickman, J. N., Petasis, N. A., ... Serhan, C.
biotic resistance has gained global concern recently and therefore the N. (2005). Resolvin E1, an endogenous lipid mediator derived from omega-3 eico-
use of antibiotics should be limited (Caton et al., 2000; Jepsen & sapentaenoic acid, protects against 2,4,6-trinitrobenzene sulfonic acid-induced co-
litis. Proceedings of the National Academy of Sciences, 102(21), 7671–7676. https://
Jepsen, 2016; Preshaw et al., 2004; Sgolastra, Petrucci, Gatto, doi.org/10.1073/pnas.0409271102.
Giannoni, & Monaco, 2011; Sulijaya, Takahashi, Yamazaki, & Arita, M., Bianchini, F., Aliberti, J., Sher, A., Chiang, N., Hong, S., ... Serhan, C. N. (2005).
Yamazaki, 2019). With respect to this, resolvin E1 might be potentially Stereochemical assignment, antiinflammatory properties, and receptor for the
omega-3 lipid mediator resolvin E1. The Journal of Experimental Medicine, 201(5),
beneficial for periodontal therapy. No clinical studies have yet been 713–722. https://doi.org/10.1084/jem.20042031.
performed to analyze the efficacy of gut metabolites (KetoC and HYA) Ashour, A. Z., Belov, V. G., Parfyonov, U. A., Parfyonov, S. A., Ershov, E. V., Tuchin, I. A.,
in periodontitis subjects; so far, investigations are still progressing in in ... Talikova, E. V. (2016). [The effectiveness of the combined use of energomonitor
antioxidant and cognitive psychotherapy in the treatment of generalized period-
vitro and in vivo studies. However, the prospect of these agents as food ontitis in elderly patients]. Stomatologiia, 95, 14–17. https://doi.org/10.17116/
supplements is expected to be valuable without giving any harmful side stomat201695214-17.
effects. Baldwin, A. S. (1996). THE NF-κB AND IκB PROTEINS: New discoveries and insights.
Annual Review of Immunology, 14(1), 649–681. https://doi.org/10.1146/annurev.
immunol.14.1.649.
4. Conclusions Balta, M. G., Loos, B. G., & Nicu, E. A. (2017). Emerging concepts in the resolution of
periodontal inflammation: A role for resolvin E1. Frontiers in Immunology, 8. https://
In summary, the host system itself is perhaps the best system for doi.org/10.3389/fimmu.2017.01682.
Bartold, P. M., & Van Dyke, T. E. (2013). Periodontitis: A host-mediated disruption of
coming to a non-destructive equilibrium with the biofilm; not by taking microbial homeostasis. Unlearning learned concepts. Periodontology 2000, 62(1),
drugs or the likes, but fortifying the host immune response by natural 203–217. https://doi.org/10.1111/j.1600-0757.2012.00450.x.
ways, which is improvement of immune fitness by a healthy life style Bartold, P. M., & Van Dyke, T. E. (2017). Host modulation : Controlling the in fl ammation
to control the infection. Periodontology 2000, 75, 317–329. https://doi.org/10.1111/
(e.g. not smoking, sufficient dietary precursors of resolving, anti-oxi- prd.12169.
dant vitamins, maintaining proper body weight, etc.). Furthermore, Battino, M., Bullon, P., Wilson, M., & Newman, H. (1999). Oxidative injury and in-
increasing evidence supports the substantive effectiveness of HMT flammatory periodontal diseases: The challenge of anti-oxidants to free radicals and
reactive oxygen species. Critical Reviews in Oral Biology & Medicine, 10(4), 458–476.
agents in periodontitis through in vitro, in vivo, and human studies. https://doi.org/10.1177/10454411990100040301.
Though HMT in combination with periodontal therapy improves peri- Berezow, A. B., & Darveau, R. P. (2011). Microbial shift and periodontitis. Periodontology
odontal parameters, systemic effects caused by HMT agent should 2000, 55(1), 36–47. https://doi.org/10.1111/j.1600-0757.2010.00350.x.
Bondue, B., Wittamer, V., & Parmentier, M. (2011). Chemerin and its receptors in leu-
continue to be monitored especially in immunocompromised patients. kocyte trafficking, inflammation and metabolism. Cytokine & Growth Factor Reviews,
A better understanding of how HMT agents work, and their targets will 22(5–6), 331–338. https://doi.org/10.1016/j.cytogfr.2011.11.004.
be beneficial in periodontal therapy. Rationally, to treat periodontitis, a Bullon, P. (2014). New theories and their clinical relevance to the onset and development of
periodontal diseases. Studies on periodontal disease, oxidative stress in applied basic re-
chronic inflammatory disease in response to commensals and patho-
search and clinical practice227–249. https://doi.org/10.1007/978-1-4614-9557-4_16.
bionts, the inflammation and oxidative stress induced tissue destruction Bullon, P., Cordero, M. D., Quiles, J. L., Morillo, J. M., Ramirez-Tortosa, M. D. C., &
could be managed in part by modulating the host immune response Battino, M. (2011). Mitochondrial dysfunction promoted by Porphyromonas gingi-
with adjunctive use of a bioactive compound with protective functions, valis lipopolysaccharide as a possible link between cardiovascular disease and

77
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

periodontitis. Free Radical Biology & Medicine, 50(10), 1336–1343. https://doi.org/ 154405910808700908.
10.1016/j.freeradbiomed.2011.02.018. Gao, L., Faibish, D., Fredman, G., Herrera, B. S., Chiang, N., Serhan, C. N., ... Gyurko, R.
Bullon, P., Cordero, M. D., Quiles, J. L., Ramirez-Tortosa, M. D. C., Gonzalez-Alonso, A., (2013). Resolvin E1 and chemokine-like receptor 1 mediate bone preservation. The
Alfonsi, S., ... Battino, M. (2012). Autophagy in periodontitis patients and gingival Journal of Immunology, 190(2), 689–694. https://doi.org/10.4049/jimmunol.
fibroblasts: Unraveling the link between chronic diseases and inflammation. BMC 1103688.
Medicine, 10, 122. https://doi.org/10.1186/1741-7015-10-122. Garlet, G. P. (2010). Critical reviews in oral biology & medicine: Destructive and pro-
Campan, P. (1997). Pilot study on n-3 polyunsaturated fatty acids in the treatment of tective roles of cytokines in periodontitis: A re-appraisal from host defense and tissue
human experimental gingivitis. Journal of Clinical Periodontology, 24(12), 907–913. destruction viewpoints. Journal of Dental Research, 89, 1349–1363. https://doi.org/
https://doi.org/10.1111/j.1600-051X.1997.tb01210.x. 10.1177/0022034510376402.
Caton, J. G., Ciancio, S. G., Blieden, T. M., Bradshaw, M., Crout, R. J., Hefti, A. F., ... Garlet, G. P., Cardoso, C. R. B., Campanelli, A. P., Ferreira, B. R., Avila-Campos, M. J.,
Walker, C. (2000). Treatment with subantimicrobial dose doxycycline improves the Cunha, F. Q., & Silva, J. S. (2007). The dual role of p55 tumour necrosis factor-??
efficacy of scaling and root planing in patients with adult periodontitis. Journal of receptor in Actinobacillus actinomycetemcomitans-induced experimental period-
Periodontology, 71(4), 521–532. https://doi.org/10.1902/jop.2000.71.4.521. ontitis: Host protection and tissue destruction. Clinical and Experimental Immunology,
Caughey, G. E., Mantzioris, E., Gibson, R. A., Cleland, L. G., & James, M. J. (1996). The 147(1), 128–138. https://doi.org/10.1111/j.1365-2249.2006.03260.x.
effect on human tumor necrosis factor α and interleukin 1β production of diets en- Garlet, G. P., Cardoso, C. R., Silva, T. A., Ferreira, B. R., Ávila-Campos, M. J., Cunha, F. Q.,
riched in n-3 fatty acids from vegetable oil or fish oil. The American Journal of Clinical & Silva, J. S. (2006). Cytokine pattern determines the progression of experimental
Nutrition, 63(1), 116–122. https://doi.org/10.1093/ajcn/63.1.116. periodontal disease induced by Actinobacillus actinomycetemcomitans through the
Cekici, A., Kantarci, A., Hasturk, H., & Van Dyke, T. (2014). Inflammatory and inmune modulation of MMPs, RANKL, and their physiological inhibitors. Oral Microbiology
pathways in the pathogenesis of periodontal disease. Periodontology 2000, 64(1), and Immunology, 21(1), 12–20. https://doi.org/10.1111/j.1399-302X.2005.00245.x.
57–80. Garlet, G. P., Martins, W., Fonseca, B. A. L., Ferreira, B. R., & Silva, J. S. (2004). Matrix
Chatterjee, A., Bhattacharya, H., & Kandwal, A. (2011). Probiotics in periodontal health metalloproteinases, their physiological inhibitors and osteoclast factors are differ-
and disease. Journal of Indian Society of Periodontology, 15(1), 23. https://doi.org/10. entially regulated by the cytokine profile in human periodontal disease. Journal of
4103/0972-124X.82260. Clinical Periodontology, 31(8), 671–679. https://doi.org/10.1111/j.1600-051X.2004.
Chiu, A. V., Saigh, M. A., McCulloch, C. A., & Glogauer, M. (2017). The role of NrF2 in the 00545.x.
regulation of periodontal health and disease. Journal of Dental Research, 96, 975–983. Glowacki, A. J., Yoshizawa, S., Jhunjhunwala, S., Vieira, A. E., Garlet, G. P., Sfeir, C., &
https://doi.org/10.1177/0022034517715007. Little, S. R. (2013). Prevention of inflammation-mediated bone loss in murine and
Cook, D. N., Pisetsky, D. S., & Schwartz, D. A. (2004). Toll-like receptors in the patho- canine periodontal disease via recruitment of regulatory lymphocytes. Proceedings of
genesis of human disease. Nature Immunology, 5, 975–979. https://doi.org/10.1038/ the National Academy of Sciences, 110(46), 18525–18530. https://doi.org/10.1073/
ni1116. pnas.1302829110.
D’Aiuto, F., Nibali, L., Parkar, M., Patel, K., Suvan, J., & Donos, N. (2010). Oxidative Graves, D. T., & Cochran, D. (2003). The contribution of Interleukin-1 and tumor necrosis
stress, systemic inflammation, and severe periodontitis. Journal of Dental Research, factor to periodontal tissue destruction. Journal of Periodontology, 74(3), 391–401.
89(11), 1241–1246. https://doi.org/10.1177/0022034510375830. https://doi.org/10.1902/jop.2003.74.3.391.
Darveau, R. P., Hajishengallis, G., & Curtis, M. A. (2012). Porphyromonas gingivalis as a Graves, D. T., Fine, D., Teng, Y. T. A., Van Dyke, T. E., & Hajishengallis, G. (2008). The
potential community activist for disease. Journal of Dental Research, 91(9), 816–820. use of rodent models to investigate host-bacteria interactions related to periodontal
https://doi.org/10.1177/0022034512453589. diseases. Journal of Clinical Periodontology, 35(2), 89–105. https://doi.org/10.1111/j.
Datta, P. K., Koukouritaki, S. B., Hopp, K. A., & Lianos, E. A. (1999). Heme oxygenase-1 1600-051X.2007.01172.x.
induction attenuates inducible nitric oxide synthase expression and proteinuria in Guyodo, H., Meuric, V., Le Pottier, L., Martin, B., Faili, A., Pers, J. O., & Bonnaure-Mallet,
glomerulonephritis. Journal of the American Society of Nephrology : JASN, 10(12), M. (2012). Colocalization of Porphyromonas gingivalis with CD4+ T cells in peri-
2540–2550. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/10589693. odontal disease. FEMS Immunology and Medical Microbiology, 64(2), 175–183. https://
Deore, G. D., Gurav, A. N., Patil, R., Shete, A. R., Naiktari, R. S., & Inamdar, S. P. (2014). doi.org/10.1111/j.1574-695X.2011.00877.x.
Herbal anti-inflammatory immunomodulators as host modulators in chronic peri- Hadler-Olsen, E., Fadnes, B., Sylte, I., Uhlin-Hansen, L., & Winberg, J. O. (2011).
odontitis patients: A randomised, double-blind, placebo-controlled, clinical trial. Regulation of matrix metalloproteinase activity in health and disease. The FEBS
Journal of Periodontal & Implant Science, 44(2), 71–78. https://doi.org/10.5051/jpis. Journal, 278(1), 28–45. https://doi.org/10.1111/j.1742-4658.2010.07920.x.
2014.44.2.71. Hajishengallis, G. (2014). The inflammophilic character of the periodontitis-associated
Dirkx, E., Schwenk, R. W., Glatz, J. F. C., Luiken, J. J. F. P., & van Eys, G. J. J. M. (2011). microbiota. Molecular Oral Microbiology, 29(6), 248–257. https://doi.org/10.1111/
High fat diet induced diabetic cardiomyopathy. Prostaglandins, Leukotrienes, and omi.12065.
Essential Fatty Acids, 85(5), 219–225. https://doi.org/10.1016/j.plefa.2011.04.018. Hajishengallis, G., & Lamont, R. J. (2012). Beyond the red complex and into more
Dona, M., Fredman, G., Schwab, J. M., Chiang, N., Arita, M., Goodarzi, A., ... Serhan, C. N. complexity: The polymicrobial synergy and dysbiosis (PSD) model of periodontal
(2008). Resolvin El, an EPA-derived mediator in whole blood, selectively counter- disease etiology. Molecular Oral Microbiology, 27(6), 409–419. https://doi.org/10.
regulates leukocytes and platelets. Blood, 112(3), 848–855. https://doi.org/10.1182/ 1111/j.2041-1014.2012.00663.x.
blood-2007-11-122598. Hajishengallis, G. (2015). Periodontitis: From microbial immune subversion to systemic
Dyerberg, J., & Bang, H. O. (1979). Hæmostatic function and platelet polyunsaturated inflammation. Nature Reviews Immunology, 15, 30–44. https://doi.org/10.1038/
fatty acids in eskimos. Lancet (London, England), 314(8140), 433–435. https://doi. nri3785.
org/10.1016/S0140-6736(79)91490-9. Hajishengallis, G., Darveau, R. P., & Curtis, M. A. (2012). The keystone-pathogen hy-
El-Sharkawy, H., Aboelsaad, N., Eliwa, M., Darweesh, M., Alshahat, M., Kantarci, A., ... pothesis. Nature Reviews Microbiology, 10, 717–725. https://doi.org/10.1038/
Van Dyke, T. E. (2010). Adjunctive treatment of chronic periodontitis with daily nrmicro2873.
dietary supplementation with Omega-3 fatty acids and low-dose aspirin. Journal of Hajishengallis, G., Liang, S., Payne, M. A., Hashim, A., Jotwani, R., Eskan, M. A., ... Curtis,
Periodontology, 81(11), 1635–1643. https://doi.org/10.1902/jop.2010.090628. M. A. (2011). Low-abundance biofilm species orchestrates inflammatory periodontal
El Kebir, D., Gjorstrup, P., & Filep, J. G. (2012). Resolvin E1 promotes phagocytosis- disease through the commensal microbiota and complement. Cell Host & Microbe,
induced neutrophil apoptosis and accelerates resolution of pulmonary inflammation. 10(5), 497–506. https://doi.org/10.1016/j.chom.2011.10.006.
Proceedings of the National Academy of Sciences, 109(37), 14983–14988. https://doi. Hanada, T., & Yoshimura, A. (2002). Regulation of cytokine signaling and inflammation.
org/10.1073/pnas.1206641109. Cytokine & Growth Factor Reviews, 13, 413–421. https://doi.org/10.1016/S1359-
Elkhouli, A. M. (2011). The efficacy of host response modulation therapy (omega-3 plus 6101(02)00026-6.
low-dose aspirin) as an adjunctive treatment of chronic periodontitis (Clinical and Hasturk, H., Kantarci, A., Goguet-Surmenian, E., Blackwood, A., Andry, C., Serhan, C. N.,
biochemical study). Journal of Periodontal Research, 46(2), 261–268. https://doi.org/ & Van Dyke, T. E. (2007). Resolvin E1 regulates inflammation at the cellular and
10.1111/j.1600-0765.2010.01336.x. tissue level and restores tissue homeostasis in vivo. The Journal of Immunology,
Eritsland, J., Arnesen, H., Seljeflot, I., & Høstmark, A. T. (1995). Long-term metabolic 179(10), 7021–7029. https://doi.org/10.4049/jimmunol.179.10.7021.
effects of n-3 polyunsaturated fatty acids in patients with coronary artery disease. The Hatakeyama, J., Tamai, R., Sugiyama, A., Akashi, S., Sugawara, S., & Takada, H. (2003).
American Journal of Clinical Nutrition, 61(4), 831–836. https://doi.org/10.1093/ajcn/ Contrasting responses of human gingival and periodontal ligament fibroblasts to
61.4.831. bacterial cell-surface components through the CD14/toll-like receptor system. Oral
Fredman, G., Oh, S. F., Ayilavarapu, S., Hasturk, H., Serhan, C. N., & van Dyke, T. E. Microbiology and Immunology, 18(1), 14–23. https://doi.org/10.1034/j.1399-302X.
(2011). Impaired phagocytosis in localized aggressive periodontitis: Rescue by re- 2003.180103.x.
solvin E1. PloS One, 6(9), https://doi.org/10.1371/journal.pone.0024422. Herrero, E. R., Fernandes, S., Verspecht, T., Ugarte-Berzal, E., Boon, N., Proost, P., ...
Freire, M. O., Dalli, J., Serhan, C. N., & Van Dyke, T. E. (2017). Neutrophil resolvin E1 Teughels, W. (2018). Dysbiotic biofilms deregulate the periodontal inflammatory
receptor expression and function in type 2 diabetes. The Journal of Immunology, response. Journal of Dental Research, 97(5), 547–555. https://doi.org/10.1177/
198(2), 718–728. https://doi.org/10.4049/jimmunol.1601543. 0022034517752675.
Freire, M. O., & Van Dyke, T. E. (2013). Natural resolution of inflammation. Iwasaki, K., Maeda, K., Hidaka, K., Nemoto, K., Hirose, Y., & Deguchi, S. (2016). Daily
Periodontology 2000, 63(1), 149–164. https://doi.org/10.1111/prd.12034. intake of heat-killed Lactobacillus plantarum L-137 decreases the probing depth in
Furumoto, H., Nanthirudjanar, T., Kume, T., Izumi, Y., Park, S. B., Kitamura, N., ... patients undergoing supportive periodontal therapy. Oral Health & Preventive
Sugawara, T. (2016). 10-Oxo-trans-11-octadecenoic acid generated from linoleic acid Dentistry, 14(3), 207–214. https://doi.org/10.3290/j.ohpd.a36099.
by a gut lactic acid bacterium Lactobacillus plantarum is cytoprotective against Janeway, C. A., & Medzhitov, R. (2002). Innate immune recognition. Annual Review of
oxidative stress. Toxicology and Applied Pharmacology, 296, 1–9. https://doi.org/10. Immunology, 20(1), 197–216. https://doi.org/10.1146/annurev.immunol.20.083001.
1016/j.taap.2016.02.012. 084359.
Gaffen, S. L., & Hajishengallis, G. (2008). A new inflammatory cytokine on the block: Re- Jepsen, K., & Jepsen, S. (2016). Antibiotics/antimicrobials: Systemic and local adminis-
thinking periodontal disease and the Th1/Th2 paradigm in the context of Th17 cells tration in the therapy of mild to moderately advanced periodontitis. Periodontology
and IL-17. Journal of Dental Research, 87, 817–828. https://doi.org/10.1177/ 2000, 71(1), 82–112. https://doi.org/10.1111/prd.12121.

78
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

Johnstone, A. M., Koh, A., Goldberg, M. B., & Glogauer, M. (2007). A hyperactive neu- Netea, M. G., van de Veerdonk, F. L., van der Meer, J. W. M., Dinarello, C. A., & Joosten,
trophil phenotype in patients with refractory periodontitis. Journal of Periodontology, L. A. B. (2014). Inflammasome-independent regulation of IL-1-Family cytokines.
78(9), 1788–1794. https://doi.org/10.1902/jop.2007.070107. Annual Review of Immunology, 33(1), 49–77. https://doi.org/10.1146/annurev-
Kaisho, T., & Akira, S. (2002). Toll-like receptors as adjuvant receptors. Biochimica et immunol-032414-112306.
Biophysica Acta - Molecular Cell Research, 1589, 1–13. https://doi.org/10.1016/ Olsen, I., & Yilmaz, Ö. (2016). Modulation of inflammasome activity by Porphyromonas
S0167-4889(01)00182-3. gingivalis in periodontitis and associated systemic diseases. Journal of Oral
Kindle, L., Rothe, L., Kriss, M., Osdoby, P., & Collin-Osdoby, P. (2006). Human micro- Microbiology, 8(1), 30385. https://doi.org/10.3402/jom.v8.30385.
vascular endothelial cell activation by IL-1 and TNF-α stimulates the adhesion and Orihuela-Campos, R. C., Tamaki, N., Mukai, R., Fukui, M., Miki, K., Terao, J., & Ito, H.-O.
transendothelial migration of circulating human CD14+monocytes that develop with (2015). Biological impacts of resveratrol, quercetin, and &i&N&/i&-acetylcysteine on
RANKL into functional osteoclasts. Journal of Bone and Mineral Research, 21(2), oxidative stress in human gingival fibroblasts. Journal of Clinical Biochemistry and
193–206. https://doi.org/10.1359/JBMR.051027. Nutrition, 56(3), 220–227. https://doi.org/10.3164/jcbn.14-129.
Kornman, K. S., Newman, M. G., Alvarado, R., Flemmig, T. F., Nachnani, S., & Tumbusch, Otomo-Corgel, J. (2012). Osteoporosis and osteopenia: Implications for periodontal and
J. (1991). Clinical and microbiological patterns of adults with periodontitis. Journal implant therapy. Periodontology 2000, 59(1), 111–139. https://doi.org/10.1111/j.
of Periodontology, 62(10), 634–642. https://doi.org/10.1902/jop.1991.62.10.634. 1600-0757.2011.00435.x.
Laleman, I., Yilmaz, E., Ozcelik, O., Haytac, C., Pauwels, M., Herrero, E. R., ... Teughels, Park, E., Na, H. S., Song, Y.-R., Shin, S. Y., Kim, Y.-M., & Chung, J. (2014). Activation of
W. (2015). The effect of a streptococci containing probiotic in periodontal therapy: A NLRP3 and AIM2 inflammasomes by Porphyromonas gingivalis infection. Infection
randomized controlled trial. Journal of Clinical Periodontology, 42(11), 1032–1041. and Immunity, 82(1), 112–123. https://doi.org/10.1128/iai.00862-13.
https://doi.org/10.1111/jcpe.12464. Petit, M. D. A., Hovenkamp, E., Hamann, D., Roos, M. T. L., Van Der Velden, U., Miedema,
Lamont, R. J., & Hajishengallis, G. (2015). Polymicrobial synergy and dysbiosis in in- F., & Loos, B. G. (2001). Phenotypical and functional analysis of T cells in period-
flammatory disease. Trends in Molecular Medicine, 21, 172–183. https://doi.org/10. ontitis. Journal of Periodontal Research, 36(4), 214–220. https://doi.org/10.1034/j.
1016/j.molmed.2014.11.004. 1600-0765.2001.036004214.x.
Lee, C.-T., Teles, R., Kantarci, A., Chen, T., McCafferty, J., Starr, J. R., ... Van Dyke, T. E. Pihlstrom, B. L., Michalowicz, B. S., & Johnson, N. W. (2005). Periodontal diseases.
(2016). Resolvin E1 reverses experimental periodontitis and dysbiosis. The Journal of Lancet, 366(9499), 1809–1820. https://doi.org/10.1016/S0140-6736(05)67728-8.
Immunology, 197(7), 2796–2806. https://doi.org/10.4049/jimmunol.1600859. Preshaw, P. M. (2018). Host modulation therapy with anti-inflammatory agents.
Li, N., Alam, J., Venkatesan, M. I., Eiguren-Fernandez, A., Schmitz, D., Di Stefano, E., ... Periodontology 2000, 76, 131–149. https://doi.org/10.1111/prd.12148.
Nel, A. E. (2004). Nrf2 is a key transcription factor that regulates antioxidant defense Preshaw, P. M., Hefti, A. F., Jepsen, S., Etienne, D., Walker, C., & Bradshaw, M. H. (2004).
in macrophages and epithelial cells: Protecting against the proinflammatory and Subantimicrobial dose doxycycline as adjunctive treatment for periodontitis: A re-
oxidizing effects of diesel exhaust chemicals. The Journal of Immunology, 173(5), view. Journal of Clinical Periodontology, 31, 697–707. https://doi.org/10.1111/j.
3467–3481. https://doi.org/10.4049/jimmunol.173.5.3467. 1600-051X.2004.00558.x.
Lin, W., Wu, R. T., Wu, T., Khor, T. O., Wang, H., & Kong, A. N. (2008). Sulforaphane Rajput, A., & Ware, C. F. (2015). Tumor necrosis factor signaling pathways. Encyclopedia
suppressed LPS-induced inflammation in mouse peritoneal macrophages through of Cell Biology, 3, 353–363. https://doi.org/10.1016/B978-0-12-394447-4.30048-7.
Nrf2 dependent pathway. Biochemical Pharmacology, 76(8), 967–973. https://doi. Reddy, M. S., Geurs, N. C., & Gunsolley, J. C. (2003). Periodontal host modulation with
org/10.1016/j.bcp.2008.07.036. Antiproteinase, anti-inflammatory, and bone-sparing Agents.A systematic review.
Liu, Y. C. G., Lerner, U. H., & Teng, Y. T. A. (2010). Cytokine responses against period- Annals of Periodontology, 8(1), 12–37. https://doi.org/10.1902/annals.2003.8.1.12.
ontal infection: Protective and destructive roles. Periodontology 2000, 52(1), Rinuchitra, D., Govindarajan, J., Thangavelu, S., & Jayaraman, A. (2012). Kikuchi-
163–206. https://doi.org/10.1111/j.1600-0757.2009.00321.x. Fujimoto’s disease and scrub typhus: A rare association. Brunei International Medical
Löe, H., Theilade, E., & Jensen, S. B. (1965). Experimental gingivitis in man. Journal of Journal, 8(5), 271–274. https://doi.org/10.1080/20002297.2017.1340085.
Periodontology, 36(3), 177–187. https://doi.org/10.1902/jop.1965.36.3.177. Sanz, M., Beighton, D., Curtis, M. A., Cury, J. A., Dige, I., Dommisch, H., ... Zaura, E.
Lukens, J. R., Gross, J. M., Calabrese, C., Iwakura, Y., Lamkanfi, M., Vogel, P., & (2017). Role of microbial biofilms in the maintenance of oral health and in the de-
Kanneganti, T.-D. (2014). Critical role for inflammasome-independent IL-1 produc- velopment of dental caries and periodontal diseases. Consensus report of group 1 of
tion in osteomyelitis. Proceedings of the National Academy of Sciences, 111(3), the Joint EFP/ORCA workshop on the boundaries between caries and periodontal
1066–1071. https://doi.org/10.1073/pnas.1318688111. disease. Journal of Clinical Periodontology, 44, S5–S11. https://doi.org/10.1111/jcpe.
Marsh, P. D. (2003). Are dental diseases examples of ecological catastrophes? 12682.
Microbiology, 149(2), 279–294. https://doi.org/10.1099/mic.0.26082-0. Schieber, M., & Chandel, N. S. (2014). ROS function in redoz signaling and oxidative
Marsh, P. D. (2004). Dental plaque as a microbial biofilm. Caries Research, 38(3), stress. Current Biology, 24(10), R453–R462. https://doi.org/10.1016/j.cub.2014.03.
204–211. https://doi.org/10.1159/000077756. 034.ROS.
Matsuda, Y., Minagawa, T., Okui, T., & Yamazaki, K. (2018). Resveratrol suppresses the Serhan, C. N., & Savill, J. (2005). Resolution of inflammation: The beginning programs
alveolar bone resorption induced by artificial trauma from occlusion in mice. Oral the end. Nature Immunology, 6(12), 1191–1197. https://doi.org/10.1038/ni1276.
Diseases, 24(3), 412–421. https://doi.org/10.1111/odi.12785. Sgolastra, F., Petrucci, A., Gatto, R., Giannoni, M., & Monaco, A. (2011). Long-term ef-
Mauro, C., Leow, S. C., Anso, E., Rocha, S., Thotakura, A. K., Tornatore, L., ... Franzoso, G. ficacy of subantimicrobial-dose doxycycline as an adjunctive treatment to scaling and
(2011). NF-κB controls energy homeostasis and metabolic adaptation by upregulating root planing: A systematic review and meta-analysis. Journal of Periodontology,
mitochondrial respiration. Nature Cell Biology, 13(10), 1272–1279. https://doi.org/ 82(11), 1570–1581. https://doi.org/10.1902/jop.2011.110026.
10.1038/ncb2324. Shi, B., Chang, M., Martin, J., Mitreva, M., Lux, R., Klokkevold, P., ... Li, H. (2015).
Mayanagi, G., Kimura, M., Nakaya, S., Hirata, H., Sakamoto, M., Benno, Y., & Shimauchi, Dynamic changes in the subgingival microbiome and their potential for diagnosis and
H. (2009). Probiotic effects of orally administered Lactobacillus salivarius WB21- prognosis of periodontitis. MBio, 6(1), https://doi.org/10.1128/mbio.01926-14.
containing tablets on periodontopathic bacteria: A double-blinded, placebo-con- Shibata, K. (2018). Historical aspects of studies on roles of the inflammasome in the
trolled, randomized clinical trial. Journal of Clinical Periodontology, 36(6), 506–513. pathogenesis of periodontal diseases. Molecular Oral Microbiology, 33(3), 203–211.
https://doi.org/10.1111/j.1600-051X.2009.01392.x. https://doi.org/10.1111/omi.12217.
Medzhitov, R., & Janeway, C. A. (1997). Innate immunity: The virtues of a nonclonal Sima, C., Aboodi, G. M., Lakschevitz, F. S., Sun, C., Goldberg, M. B., & Glogauer, M.
system of recognition. Cell, 91, 295–298. https://doi.org/10.1016/S0092-8674(00) (2016). Nuclear factor erythroid 2-Related factor 2 down-regulation in oral neu-
80412-2. trophils is associated with periodontal oxidative damage and severe chronic peri-
Meurman, J. H., & Stamatova, I. (2007). Probiotics: Contributions to oral health. Oral odontitis. The American Journal of Pathology, 186(6), 1417–1426. https://doi.org/10.
Diseases, 13, 443–451. https://doi.org/10.1111/j.1601-0825.2007.01386.x. 1016/j.ajpath.2016.01.013.
Meydani, S. N., Endres, S., Woods, M. M., Goldin, B. R., Soo, C., Morrill-Labrode, A., ... Sima, C., & Glogauer, M. (2014). Neutrophil dysfunction and host susceptibility to peri-
Gorbach, S. L. (1991). Oral (n-3) fatty acid supplementation suppresses cytokine odontal inflammation: Current state of knowledge. Current Oral Health Reports,
production and lymphocyte proliferation: Comparison between young and older 95–103. https://doi.org/10.1007/s40496-014-0015-x.
women. The Journal of Nutrition, 121(4), 547–555. https://doi.org/10.1093/jn/121. Smale, S. T. (2011). Hierarchies of NF-kappaB target-gene regulation. Nature Immunology,
4.547. 12, 689–694.
Minagawa, T., Okui, T., Takahashi, N., Nakajima, T., Tabeta, K., Murakami, S., ... Socransky, S. S., & Haffajee, A. D. (1992). The bacterial etiology of destructive period-
Yamazaki, K. (2015). Resveratrol suppresses the inflammatory responses of human ontal disease: Current concepts. Journal of Periodontology, 63(4s), 322–331. https://
gingival epithelial cells in a SIRT1 independent manner. Journal of Periodontal doi.org/10.1902/jop.1992.63.4s.322.
Research, 50(5), 586–593. https://doi.org/10.1111/jre.12238. Steeve, K. T., Marc, P., Sandrine, T., Dominique, H., & Yannick, F. (2004). IL-6, RANKL,
Mohammed, S., Ferzandi, L., & Hamilton, K. (2010). Metaphor no more: A 15-year review TNF-alpha/IL-1: Interrelations in bone resorption pathophysiology. Cytokine &
of the team mental model construct. Journal of Management, 36(4), 876–910. https:// Growth Factor Reviews, 15, 49–60. https://doi.org/10.1016/j.cytogfr.2003.10.005.
doi.org/10.1177/0149206309356804. Sulijaya, B., Takahashi, N., Yamada, M., Yokoji, M., Sato, K., Aoki-Nonaka, Y., ...
Morales, A., Gandolfo, A., Bravo, J., Carvajal, P., Silva, N., Godoy, C., ... Gamonal, J. Yamazaki, K. (2018). The anti-inflammatory effect of 10-oxo-trans-11-octadecenoic
(2018). Microbiological and clinical effects of probiotics and antibiotics on non- acid (KetoC) on RAW 264.7 cells stimulated with Porphyromonas gingivalis lipopo-
surgical treatment of chronic periodontitis: A randomized placebo- controlled trial lysaccharide. Journal of Periodontal Research, 53(5), 777–784. https://doi.org/10.
with 9-month follow-up. Journal of Applied Oral Science, 26(0), https://doi.org/10. 1111/jre.12564.
1590/1678-7757-2017-0075. Sulijaya, B., Takahashi, N., Yamazaki, K., & Yamazaki, K. (2019). Nutrition as adjunct
Murphy, K. M., & Reiner, S. L. (2002). The lineage decisions of helper T cells. Nature therapy in periodontal disease management. Current Oral Health Reports, 6(2), 61–69.
Reviews Immunology, 2, 933–944. https://doi.org/10.1038/nri954. https://doi.org/10.1007/s40496-019-0216-4.
Naqvi, A. Z., Hasturk, H., Mu, L., Phillips, R. S., Davis, R. B., Halem, S., ... Mukamal, K. J. Tamaki, N., Cristina Orihuela-Campos, R., Inagaki, Y., Fukui, M., Nagata, T., & Ito, H. O.
(2014). Docosahexaenoic acid and periodontitis in adults: A randomized controlled (2014). Resveratrol improves oxidative stress and prevents the progression of peri-
trial. Journal of Dental Research, 93(8), 767–773. https://doi.org/10.1177/ odontitis via the activation of the Sirt1/AMPK and the Nrf2/antioxidant defense
0022034514541125. pathways in a rat periodontitis model. Free Radical Biology & Medicine, 75, 222–229.

79
B. Sulijaya, et al. Archives of Oral Biology 105 (2019) 72–80

https://doi.org/10.1016/j.freeradbiomed.2014.07.034. but do not increase the risk of bleeding: Clinical observations and mechanistic in-
Teughels, W., Durukan, A., Ozcelik, O., Pauwels, M., Quirynen, M., & Cenk Haytac, M. sights. The British Journal of Nutrition, 111(9), 1652–1662. https://doi.org/10.1017/
(2013). Clinical and microbiological effects of Lactobacillus reuteri probiotics in the S000711451300425X.
treatment of chronic periodontitis: A randomized placebo-controlled study Clinical Wang, H., Zhou, H., Duan, X., Jotwani, R., Vuddaraju, H., Liang, S., ... Lamont, R. J.
and microbiological effects of Lactobacillus reuteri probiotics in the treatment of (2014). Porphyromonas gingivalis-induced reactive oxygen species activate JAK2
chronic. Journal of Clinical Periodontology, 40, 1025–1035. https://doi.org/10.1111/ and regulate production of inflammatory cytokines through c-Jun. Infection and
jcpe.12155. Immunity, 82(10), 4118–4126. https://doi.org/10.1128/IAI.02000-14.
Teughels, W., Durukan, A., Ozcelik, O., Pauwels, M., Quirynen, M., & Haytac, M. C. Wardyn, J. D., Ponsford, A. H., & Sanderson, C. M. (2015). Dissecting molecular cross-talk
(2013). Clinical and microbiological effects of Lactobacillus reuteri probiotics in the between Nrf2 and NF-κB response pathways. Biochemical Society Transactions, 43(4),
treatment of chronic periodontitis: A randomized placebo-controlled study. Journal of 621–626. https://doi.org/10.1042/BST20150014.
Clinical Periodontology, 40(11), 1025–1035. https://doi.org/10.1111/jcpe.12155. Watters, T. M., Kenny, E. F., & O’Neill, L. A. J. (2007). Structure, function and regulation
Theilade, E. (1986). The non‐specific theory in microbial etiology of inflammatory peri- of the Toll/IL-1 receptor adaptor proteins. Immunology and Cell Biology, 85, 411–419.
odontal diseases. Journal of Clinical Periodontology, 13(10), 905–911. https://doi.org/ https://doi.org/10.1038/sj.icb.7100095.
10.1111/j.1600-051X.1986.tb01425.x. Yamada, M., Takahashi, N., Matsuda, Y., Sato, K., Yokoji, M., Sulijaya, B., ... Yamazaki, K.
Thirunavukkarasu, M., Penumathsa, S. V., Koneru, S., Juhasz, B., Zhan, L., Otani, H., ... (2018). A bacterial metabolite ameliorates periodontal pathogen-induced gingival
Maulik, N. (2007). Resveratrol alleviates cardiac dysfunction in streptozotocin-in- epithelial barrier disruption via GPR40 signaling. Scientific Reports, 8(1), https://doi.
duced diabetes: Role of nitric oxide, thioredoxin, and heme oxygenase. Free Radical org/10.1038/s41598-018-27408-y.
Biology & Medicine, 43(5), 720–729. https://doi.org/10.1016/j.freeradbiomed.2007. Yamaguchi, Y., Kurita-Ochiai, T., Kobayashi, R., Suzuki, T., & Ando, T. (2017). Regulation
05.004. of the NLRP3 inflammasome in Porphyromonas gingivalis-accelerated periodontal
Tomofuji, T., Irie, K., Sanbe, T., Azuma, T., Ekuni, D., Tamaki, N., ... Morita, M. (2009). disease. Inflammation Research, 66(1), 59–65. https://doi.org/10.1007/s00011-016-
Periodontitis and increase in circulating oxidative stress. The Japanese Dental Science 0992-4.
Review, 45, 46–51. https://doi.org/10.1016/j.jdsr.2008.12.002. Yang, H. E., Li, Y., Nishimura, A., Jheng, H. F., Yuliana, A., Kitano-Ohue, R., ... Goto, T.
Van Dyke, T. E. (2008a). Inflammation and periodontal diseases: A reappraisal. Journal of (2017). Synthesized enone fatty acids resembling metabolites from gut microbiota
Periodontology, 79(8s), 1501–1502. https://doi.org/10.1902/jop.2008.080279. suppress macrophage-mediated inflammation in adipocytes. Molecular Nutrition &
Van Dyke, T. E. (2008b). The management of inflammation in periodontal disease. Food Research, 61(10), 1–13. https://doi.org/10.1002/mnfr.201700064.
Journal of Periodontology, 79(8s), 1601–1608. https://doi.org/10.1902/jop.2008. Yanine, N., Araya, I., Brignardello-Petersen, R., Carrasco-Labra, A., González, A.,
080173. Preciado, A., ... Martin, C. (2013). Effects of probiotics in periodontal diseases: A
Vandooren, J., Van Den Steen, P. E., & Opdenakker, G. (2013). Biochemistry and mole- systematic review. Clinical Oral Investigations, 17(7), 1627–1634. https://doi.org/10.
cular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9): The next 1007/s00784-013-0990-7.
decade. Critical Reviews in Biochemistry and Molecular Biology, 48, 222–272. https:// Yen, C. A., Damoulis, P. D., Stark, P. C., Hibberd, P. L., Singh, M., & Papas, A. S. (2008).
doi.org/10.3109/10409238.2013.770819. The effect of a selective Cyclooxygenase-2 inhibitor (Celecoxib) on chronic period-
Vicario, M., Santos, A., Violant, D., Nart, J., & Giner, L. (2013). Clinical changes in ontitis. Journal of Periodontology, 79(1), 104–113. https://doi.org/10.1902/jop.2008.
periodontal subjects with the probiotic Lactobacillus reuteri Prodentis: A preliminary 070271.
randomized clinical trial. Acta Odontologica Scandinavica, 71(3–4), 813–819. https:// Zlotnik, A., & Yoshie, O. (2000). Chemokines: A new classification system and their role
doi.org/10.3109/00016357.2012.734404. in immunity. Immunity, 12(2), 121–127. https://doi.org/10.1016/S1074-7613(00)
Wachira, J. K., Larson, M. K., & Harris, W. S. (2014). N-3 Fatty acids affect haemostasis 80165-X.

80

You might also like