You are on page 1of 10

Fitoterapia 132 (2019) 30–39

Contents lists available at ScienceDirect

Fitoterapia
journal homepage: www.elsevier.com/locate/fitote

Review

Polyphenols in the prevention and treatment of periodontal disease: A T


systematic review of in vivo, ex vivo and in vitro studies

Kübra Buntea, , Andreas Henselb,1, Thomas Beiklerc,1
a
Private Dental Clinic, Shanghai, China
b
University of Münster, Institute of Pharmaceutical Biology and Phytochemistry, Corrensstr. 48, 48149 Münster, Germany
c
University Medical Centre Hamburg-Eppendorf, Department of Periodontics, Preventive and Restorative Dentistry, Building O58, Martinistr. 52, 20246 Hamburg,
Germany

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

Keywords: Plant-derived polyphenols with antimicrobial and immunomodulatory characteristics appear to provide a
Plants variety of oral health benefits. Thus, the aim of the present study was to review the scientific literature to identify
Polyphenols these effects of polyphenols on periodontal pathogens and inflammation. A MEDLINE search from 1st January
Periodontal disease 2013 to 18th January 2018 was performed to identify studies reporting polyphenol-containing plant extracts.
Periodontitis
Reports regarding pure compounds and essential oils, as well as effects on bacteria that are not defined as
Antibacterial
Anti-inflammatory
periodontal pathogens, were excluded. Thirty-eight studies matched the selection criteria. Studies on im-
munomodulatory effects included in vitro, ex vivo, and in vivo studies (n = 23), whereas studies reporting anti-
bacterial effects against periodontal pathogens included only in vitro studies (n = 18). Three studies were in-
cluded in both groups. The antibacterial effects were characterised by inhibition of bacterial growth, adhesion to
oral cells, and enzymatic activity. Decreased secretion of pro-inflammatory and increased secretion of anti-
inflammatory cytokines were demonstrated. Higher attachment levels, lower inflammation, and bone loss were
reported by in vivo studies. Due to the high heterogeneity, it is difficult to draw clear conclusions for applic-
ability; nevertheless, polyphenols have great potential as antimicrobial and immunomodulatory substances in
the treatment and prevention of periodontal disease.

1. Introduction Non-surgical treatment of periodontitis by machine-driven or


manual instrumentation aims to reduce the intraoral bacterial load.
Periodontitis is a chronic inflammatory disease initiated and per- Moreover, adjunctive antimicrobial strategies such as local and sys-
petuated by an intraoral dysbiosis that leads to progressive destruction temic antibiotics or non-specific antimicrobials, e.g., chlorhexidine di-
of the periodontal ligament, connective tissue, and alveolar bone; and if gluconate, are used to augment the antimicrobial effects of mechanical
left untreated, results in tooth loss [1–3]. Although the majority of debridement [11]. However, numerous adverse effects, most im-
microorganisms colonizing the gingival sulcus at or below the gingival portantly an increase in bacterial resistance against antibiotics, high-
margin are compatible with periodontal health, a subset of species may light the need for the development of novel adjunctive strategies for the
cause or contribute to an intraoral dysbiosis, and subsequently to the treatment and prevention of this prevalent disease [12,13].
clinical signs of periodontal disease [4]. In this regard, approximately In this regard, plant-based traditional medicine or phytother-
15 to 20 bacterial species have been found to be closely associated with apeutics have become a focus of scientific and clinical interest [14].
periodontal disease [5]. Among these, Agregatibacter actinomycetemco- Polyphenols are natural products, mainly produced by higher plants as
mitans and the prominent species from the so-called “red complex” e.g. secondary metabolites with different functions ranging from defense to
Porphyromonas gingivalis appear to be the most virulent [6]. However, growth regulation and plant colour [15,16]. An exact classification and
not only bacterial invasion and virulence but also the host immune differentiation of these compounds is challenging due to their complex
response induced by this dysbiotic intraoral ecosystem plays a major structures; > 8000 identified compounds have been described to date,
role in the progression of periodontal disease [7–10]. and classification is mainly based on their respective chemical


Corresponding author at: DDS Dental Care, Office Building A, UG 05-07, No. 1799 Wuzhong Road, 201103 Shanghai, China.
E-mail addresses: kbrtncl@yahoo.com (K. Bunte), ahensel@uni-muenster.de (A. Hensel), t.beikler@uke.de (T. Beikler).
1
Co-author.

https://doi.org/10.1016/j.fitote.2018.11.012
Received 13 October 2018; Received in revised form 13 November 2018; Accepted 24 November 2018
Available online 27 November 2018
0367-326X/ © 2018 Elsevier B.V. All rights reserved.
K. Bunte et al. Fitoterapia 132 (2019) 30–39

Fig. 1. Classification of polyphenols and typical representative compounds.

improved endothelial function, reduced low-density lipoprotein (LDL)


plasma concentration, and cardiac infarct size have been reported
[29,30]. In addition to these health benefits, polyphenols have been
associated with improved wound healing [31,32], inhibition of the
adhesion of pathogens to host cells [33,34], and antibacterial effects
against gastrointestinal and cariogenic pathogens [35,36].
Such characteristics turn these substances into promising targets for
the development of adjunctive preventive and therapeutic strategies not
only but also for the management of periodontal disease. Therefore, the
aims of the present study were to review the literature to identify the
antibacterial effect of plant-derived polyphenols on periodontal pa-
thogens, as well as their modulatory effect on the host immune and
inflammatory responses.

2. Materials and methods

Fig. 2. Typical chemical structural features of flavonoids (A), anthocyanidins


2.1. Search strategy
(B), proanthocyanidins (C), cinnamic acid (D) and benzoic acid (E) derivatives.

An electronic database search was conducted using PubMed (access


composition [17]. Some authors have classified polyphenols into five date from 1st January 2013 to 18th January 2018). The search strategy
main groups: hydrolysable tannins, phenolic acids, polyphenolic terms were categorised into three groups: periodontitis OR periodontal
amides, flavonoids, and other, with their corresponding subgroups diseases AND polyphenols OR plant extracts OR herbal medicine OR
based on chemical composition (Fig. 1) [16,18]. Flavonoids, as the most plants AND anti-bacterial OR antiadhesive OR antivirulence OR anti-
abundant group of polyphenols, are further divided into seven sub- inflammatory OR antioxidant, respectively. Search results from all re-
groups; they share a common structure, which consists of two aromatic levant combinations of the above search terms were imported into a
rings (A and B) and three carbon atoms that bind ring A and B together, citation manager program, and all duplicates were automatically re-
creating an oxygenated heterocycle named ring C (Fig. 2) [19]. moved. All imported studies were screened by title and abstract.
Oxidative stress, described as an imbalance between the production Bibliographies of the retrieved articles were also screened for articles
and elimination of reactive oxygen/nitrogen species (ROS/RNS), results relevant to the search strategy.
in inflammation and associated health problems such as cardiovascular,
cerebrovascular, immunological, metabolic, and neurodegenerative 2.2. Inclusion criteria
diseases [17]. Due to their antioxidant effects, ameliorative or pre-
ventive effects of polyphenols on several inflammatory diseases have The effects were classified according to their antibacterial and im-
been reported by numerous studies [20–27]. Styshova et al., recently munomodulatory actions. Studies using polyphenol-containing plant
published anti-diabetic activities of polyphenols derived from Zostera extracts or fractions from flowers, seeds, roots, fruits, leaves, and
seagrasses [28], and the consumption of dietary flavonoids has also trunks, that have been analysed with respect to their effects on peri-
been related to a reduced risk of developing type II diabetes mellitus odontal pathogens were assigned to the antibacterial group.
[25]. Moreover, a recent systematic review indicated a reduced risk of Accordingly, studies reporting immunomodulatory effects on host cells
colorectal cancer in individuals with habitual dietary intake of flavo- or tissues treated with exotoxins from periodontal pathogens were al-
noid-rich foods [26]. Improved cardiac function, reduced diastolic located to the anti-inflammatory group. Thus, this review includes in
blood pressure, and more favourable cardiovascular risk factors, such as vitro, ex vivo, and in vivo studies that investigated the

31
K. Bunte et al. Fitoterapia 132 (2019) 30–39

immunomodulatory effects of plant-derived polyphenols and in vitro properties of proanthocyanidins from cranberry or cranberry juice ex-
studies that reported the antibacterial effects against periodontal pa- tract [70,72–74]. Two studies reported on the anti-bacterial and anti-
thogens. Only studies published in the English language over the last inflammatory effects of blueberry extract [45,54]. Individual studies
five years were taken into consideration. reported the effects of Epimedium species [38], Azadirachta indica leaves
[53], Limonium brasiliense [39], Ocimum sanctum [42], Rumex acetosa
2.3. Exclusion criteria (L.) [43], Murraya paniculata (L.) Jack [44], Vaccinium angustifolium Ait
[45], Glycyrrhiza uralensis [46], Phytolacca americana [47], Mammea
Compounds that were not directly extracted from plants, e.g., pure americana [52], Allium sativum (L.) [48], Dodonaea viscosa var. angu-
compounds purchased from a provider, and essential oils emanating stifolia [49], blackberry [50], Stemodia maritima (L.) [56], grape seed
from plants were omitted from the present study due to the differences [57], Salvia sclarea (L.) [58], Crataeus orientalis M Bieber [59], Termi-
in their pharmacological activity compared to crude plant extract. As it nalia chebula [61], Sophorae Flos [62], Magnolia officinalis (L.) [63],
is known that essential oils (syn. volatile oils) act unspecific against Polygoni Multiflori [65], Rhododendron ferrugineum (L.) [55], Os-
nearly all kinds of bacteria, mainly by destruction of cell membranes manthus fragrans [66], Angelica sinensis [67], Aristotelia chilensis Maqui
and no specificity or selectivity has been proven until now. The use of [69], and Geum urbanum (L.) root [64].
essential oil containing mouth wash products has already been re-
viewed and documented in the literature, therefore the present review 3.1. Effects on periodontal pathogens
excludes this compound group. Reviews and studies unrelated to peri-
odontal pathogens and tissues were also excluded from the present An overview of the antibacterial effects of the representative com-
study. Moreover, reports describing the application of plant extracts in pounds is depicted in Table 1. Fifteen studies reported on the effects
combination with routine pharmacological therapy, such as chlorhex- against P. gingivalis [38,39,41–44,46–53,55], four studies against Fu-
idine mouthrinses or antibiotics, and studies investigating the effects of sobacterium(F.) nucleatum [40,45,50,53], four studies against A. acti-
plant derived polyphenols on undefined oral multispecies biofilm were nomycetemcomitans [42,48,51,54], and two studies against Prevotella
excluded. (P.) intermedia [42,51]. Reduction or complete elimination of bacterial
enzymatic activity by direct inhibition of the synthesis of proteolytic
2.4. Data organisation enzymes and adhesins was reported by seven studies
[38,39,43,48–50,55].
Authors, year of publication, type of study, type of plant, tested Gingipains attribute to 85% of the total proteolytic activity of P.
compound from the plant extract, treatment duration, extract con- gingivalis and biochemically belong to the family of cysteine-protease,
centration, type of bacteria, type of cells and tissues, methods, and which means they are “trypsin-like” enzymes [77]. They are thiol de-
major outcomes of each study were noted in a standard document. The pendent and cleave carboxyl termini of amino acid chains to either
studies were categorised into two groups: studies reporting on anti- arginine or lysine [78]. There are two arginine-specific gingipains en-
bacterial effects on periodontal pathogens and immunomodulatory ef- coded by rgpA and rgpB genes and one lysine-specific gingipain encoded
fects on host cells and tissues. The group of studies reporting im- by kgp gene [79,80]. Acting alone or in combination, gingipains are
munomodulatory effects was further divided into three subgroups: a) in able to interfere with the host by invading and colonizing the host cells
vitro, b) ex vivo, and c) in vivo studies. The organised data were eval- [81], degrading antibacterial peptides such as defensins [82,83],
uated according to the Preferred Reporting Items for Systematic modulating the complement system by either activation or deactivation
Reviews and Meta-Analyses (PRISMA) guidelines (www.prisma- of its components [84], manipulating cytokine networks [85,86], and
statement.org) [37]. inactivating protease inhibitors [87–89]. An in vitro antiadhesive ac-
tivity of Rhododendron ferrugineum L., hydroalcoholic leaf extract
3. Results (1 mg/ml) against P. gingivalis was reported by Löhr et al. [55]. This was
induced by the inhibition of Arg-gingipain activity, and was accom-
A total of 1408 articles were identified following an online database panied by significantly reduced hemagglutination. Relative gene ex-
search and screening of reference lists of articles. The selection process pression of RgpA in P. gingivalis was also significantly downregulated by
of the search results is depicted in Fig. 3. After duplicate removal, 740 the extract, while typical inflammatory genes (e.g. cyclooxygenase-2
articles were further screened by title and abstract. A total of 601 ar- [COX-2], interleukin-1beta [IL-1β], interleukin[IL]-5, IL-8, and tumor
ticles were excluded in the first exclusion according to the exclusion necrosis factor alpha [TNF-α]) in KB cells -subline of the uniquitous
criteria − first exclusion criteria included studies that were older than KERATIN-forming tumor cell line Henrietta Lacks- were significantly
five years, unrelated to oral and periodontal pathogens, review articles, reduced. Interestingly, the Lys-gingipain activity was unaffected by the
and non-polyphenolic compounds − and 139 full-text studies were extract. The extract contained higher amounts of different flavonoids
reevaluated for eligibility. Following the second exclusion process − and 8.7% proanthocyanidin-type tannins. De Oliveira Caleare et al.,
second exclusion criteria included studies regarding non-periodontal also observed a significant reduction (80% inhibition at 100 μg/ml
pathogens, essential oils, and pure compounds −38 studies remained extract) in the proteolytic activity of Arg-gingipain and bacterial ad-
and were included in the analysis. 18 out of the 38 remaining studies hesion of P. gingivalis to KB cells for a standardised hydroalcoholic Li-
reported on the antibacterial effects against periodontal pathogens monium brasiliense rhizome extract [39]. The extract did not influence
[38–55] and 23 out of the 38 studies reported on anti-inflammatory the cell viability of P. gingivalis at concentrations that significantly in-
effects [41,54–74]. 5 out of 23 reported in vivo [56–60]; 7 out of 23 hibited the antiadhesive activity; and was characterised by the presence
reported ex vivo [61–67]; and 11 out of 23 reported in vitro of gallic acid, EGCG, and samarangenin.
[41,54,55,61,64,69–75] effects. Three of these studies were included in A pilot study demonstrated reduced Lys- and Arg-gingipain activity
both groups [41,54,55]. of P. gingivalis following exposure to Dodonaea viscosa var. angustifolia
Detailed data regarding the methods and results of each report are methanolic extract in sublethal concentrations [49]. Gonzalez et al.,
summarised in Tables 1–4. Among all the included studies, six reported tested blackberry extract (BBE), which typically contains hydrolysable
on the effects of green tea extract or its active component, epigalloca- and condensed tannins, at concentrations of 700 μg/ml and 1400 μg/ml
techin-3-O-gallate (EGCG) [40,51,60,70,75,76], and three studies re- to demonstrate its antibacterial effect [50]. Their results showed a 40%
ported on the effects of black tea extract or its active components, reduction in the metabolic activity of P. gingivalis and F. nucleatum
theaflavins [40,41,71], Both green tea and black tea are varietals of following a 24-h exposure to 700 μg/ml BBE, and an 84% reduction in
Camellia sinensis. Four studies highlighted the anti-inflammatory the metabolic activity of F. nucleatum following exposure to 1400 μg/ml

32
K. Bunte et al. Fitoterapia 132 (2019) 30–39

Fig. 3. Flowchart of the article search strategy, exclusion criteria, study selection, and data management process.

BBE. A more recent study with blueberry extract demonstrated an in- [60,68,70,75,90], of which EGCG is the active compound, and four
hibitory effect of 29.9 ± 12.3% on the growth of F. nucleatum at a studied cranberry proanthocyanidins (PACs) [71–74].
concentration of 500 μg/ml [45]. A reduced production of MMPs was reported by four studies
Antibacterial effects of green tea extract (unfermented leaves from [54,64,74,75]. In one study, A. actinomycetemcomitans lipopoly-
Camelia sinensis), characterised by the lead compound EGCG, a gal- saccharide(LPS)-induced matrix metalloproteinase(MMP)-3, −8,
loylated flavan-3-ol and a high amount of galloylated and unsubstituted and − 9 secretion from human gingival fibroblasts (HGFs) and macro-
B-type proanthocyanidins and were demonstrated by three studies phages was reduced following a 2-h pre-incubation with EGCG [75].
[40,41,51]. In one study, theaflavin from black tea extract (fermented MMP-9 reduction was found by Granica et al., following incubation
leaves from Camelia sinensis) was shown to inhibit the growth of P. with a Geum urbanum L., root extract [64]. Black tea theaflavins were
gingivalis and Arg- and Lys-gingipain activity [41]. In an earlier study, P. reported to inhibit interleukin secretion, especially IL-8, in oral epi-
gingivalis, P. intermedia, and A. actinomycetemcomitans were shown to be thelial cells [71]. In addition to the anti-inflammatory effect, thea-
sensitive to green tea extract, with the main compound being EGCG, flavins have also been shown to induce an enhanced host defense
with a minimum inhibitory concentration of 12.5 μg/ml, 12.5 μg/ml, against periodontal pathogens via an increase in human-beta-defensin
and 6.25 μg/ml, respectively [51]. Ben Lagha et al., conducted a com- (hBD)-1, −2, and − 4 production [71]. Gennaro et al., demonstrated
parative study on Camellia sinensis polyphenols [40] comparing black an increased level of osteoprotegerin in gingival tissues following
tea extract, green tea extract, and their bioactive components theaflavin treatment with green tea extract [60]. Six studies reported a reduction
and EGCG at 2 mg/ml, 1 mg/ml, and 0.5 mg/ml concentrations. Inter- in IL-8 levels [55,63,64,66,73,74], and nine studies reported a reduc-
estingly, the antibacterial activity of green tea, which contains a higher tion in interleukin-6 (IL-6) levels in periodontal tissues
concentration of EGCG as compared with black tea, could be correlated [55,58,65,66,70,72–74,76]. However, Tipton et al., also showed that
with the content of these proanthocyanidins. Moreover, their report incubation with cranberry proanthocyanidins (PACs) increased the se-
showed an inhibition of F. nucleatum growth, haemolytic activity, and cretion of IL-6 from HGFs [74]. In the same study, PACs were shown to
adhesion to oral cells. With the exception of two studies [40,53], all inhibit nuclear factor kappa-light-chain-enhancer of activated B cells
tested extracts (Table 1) were shown to inhibit the growth of at least (NF-κB) activation and MMP-3 secretion in HGFs. A reduction in
one periodontal pathogen in a dose-dependent manner. granulocyte colony-stimulating factor (G-CFS), growth-regulated pro-
tein alpha (GRO-α), IL-8, interferon gamma-induced protein 10 (IP-10),
monocyte chemoattractant protein 1 (MCP-1) [70], and IL-1ß stimu-
3.2. Effects on host inflammatory response
lated IL-6 production [72] was also reported; however, Bedran et al.,
found that cranberry PACs had no effect on IL-6 secretion [70].
A total of 23 studies were included in the assessment of host in-
Downregulation of COX-2 secretion was reported by two studies
flammatory responses following polyphenol exposure and bacterial
[61,69], iNOS downregulation by three studies [56,64,69], and in-
toxin stimulation in vivo, ex vivo, and in vitro (Tables 2–4). Five studies
hibition of receptor activator of NF-κB ligand(RANKL)-induced osteo-
reported in vivo [56–60,76], seven studies reported ex vivo [61–67], and
clast differentiation by eight studies [54,56,60–62,65,67,74]. A total of
11 studies reported in vitro [41,54,55,61,69–75] anti-inflammatory ef-
eight in vivo, in vitro, and ex vivo studies reported a reduction in TNF-α
fects. Five of these 23 studies analysed green tea extract

33
K. Bunte et al. Fitoterapia 132 (2019) 30–39

Table 1
Antibacterial effects of plant-derived polyphenols in vitro.
Study group Plant and/or active compound/s Periodontal pathogen/s Results

Ben Lagha et al., 2018 Highbush blueberry proanthocyanidins A. actinomycetemcomitans Reduction in bacterial growth
Kariu et al., 2017 Prenyl flavonoids from Epimedium species P. gingivalis Inhibition of Arg- and Lys-gingipain and bacterial
growth
Heyman et al., 2017 Polyphenols from Azadirachta indica leaves P. gingivalis Inhibition of P. gingivalis growth at 10 and 20 μM
F. nucleatum No effects on F. nucleatum
De Oliveira Caleare et al., Flavan-3-ols and proanthocyanidins from Limonium brasiliense P. gingivalis Inhibition of Arg-gingipain and bacterial adhesion
2017 No effects on bacterial vitality
Ben Lagha et al., 2017 EGCG from green tea and theaflavins from black tea F. nucleatum Inhibition of bacterial adhesion and F. nucleatum-
induced hemolysis
No effects on bacterial growth at antiadhesive
concentrations
Ben Lagha et al., 2017 Theaflavins from black tea P. gingivalis Inhibition of Arg- and Lys-gingipain and bacterial
adhesion
Enhanced tight junction integrity of gingival
keratinocytes
Mallikarjun et al., 2016 Ocimum sanctum A. actinomycetemcomitans Inhibition of A. actinomycetemcomitans growth similar
P. gingivalis to that of doxycycline
P. intermedia No significant effects on P. gingivalis or P. intermedia
Schmuch et al., 2015 Flavan-3-ols, oligomeric proanthocyanidins, and flavonoids P. gingivalis Inhibition of bacterial adhesion and P. gingivalis-
from Rumex acetosa (L.) induced hemagglutination
Inhibition of Arg-gingipain by gallolylated
proanthocyanidins
Rodanant et al., 2015 Coumarins and tetramethoxyflavone from Murraya paniculata P. gingivalis Inhibition of bacterial growth
(L.) Jack
Lohr et al., 2015 Rhododendron ferrugineum (L.) A P. gingivalis Inhibition of bacterial adhesion and Arg- and Lys-
gingipain
Ben Lagha et al., 2015 Phenolic acids, flavonols, anthocyanins, flavan-3-ols, and F. nucleatum Inhibition of bacterial growth
procyanidins from Vaccinium angustifolium Ait.
Villinski et al., 2014 Licoricidin and licorisoflavan from Glycyrrhiza uralensis P. gingivalis Inhibition of bacterial growth
Patra et al., 2014 Kaempferol from Phytolacca americana P. gingivalis 100% inhibition of bacterial growth at 1.8 mg/ml
Herrera et al., 2014 Mammea americana extract P. gingivalis 96% inhibition of bacterial growth at 500 μg/ml
Shetty et al., 2013 Allium sativum (L.) aqueous extract A. actinomycetemcomitans Inhibition of bacterial proteolytic enzymes and growth
P. gingivalis at 16.6 μl/ml
Patel et al., 2013 Dodonaea viscosa var. angustifolia P. gingivalis Reduction in Arg- and Lys-gingipain activity
Inhibition of total bacterial metabolic activity
Gonzalez et al., 2013 Blackberry P. gingivalis Reduction in metabolic activity of both pathogens
F. nucleatum
Araghizadeh et al., 2013 Green tea (Camelia sinensis) A. actinomycetemcomitans Inhibition of growth of all three pathogens
P. gingivalis
P. intermedia

levels [41,54–56,58,60,65,76], whereas one study reported an increase the antibacterial and anti-inflammatory effects. These effects can also
[64]. In addition to the reduction in pro-inflammatory cytokines, an be influenced by the exposure time to the substance, which ranged from
increased secretion of IL-10, an anti-inflammatory cytokine, was re- a minimum of 20 min [49] to a maximum of 7 days [44] for the in vitro
ported by three in vivo studies [56,57,60]. studies, and from 11 [56,59] to 90 days [60] for the in vivo studies.
Clearly, an extended treatment duration and the use of higher con-
centrations can cause an overestimation of positive antibacterial and/or
4. Discussion
anti-inflammatory effects. Nonetheless, it has also been reported in the
literature that exposure to high concentrations and larger molecules can
The aim of the present study was to review the current knowledge
lead to an increased inflammatory reaction in host cells [91,92]. In
about the antibacterial effects of plant-derived polyphenols on period-
addition to these limitations, the reviewed studies only tested plank-
ontal pathogens and their effects on the modulation of the host im-
tonic bacteria. The reaction of sessile bacteria to treatment with the
mune/inflammatory response. The pooled data from these reports in-
above-mentioned polyphenols was not analysed in any of these studies;
dicated beneficial antibacterial, antiadhesive, and anti-inflammatory
however, this needs to be considered for a proper evaluation. In this
characteristics. Moreover, a positive correlation between polyphenolic
regard, the influence of polyphenols on multispecies biofilms needs to
substance exposure and inhibition of bacterial growth, adhesion, and
be studied, since the virulence and susceptibility of bacteria, as well as
proteolytic activity, as well as a reduced host inflammatory response
the effectiveness of polyphenols, may vary immensely in the natural
and enhanced host defense mechanism could be derived from these
ecosystem, i.e., the biofilm. Moreover, the studies included in the pre-
studies.
sent review are characterised by specific periodontal pathogens and cell
However, generalised conclusions for routine treatment cannot be
culture systems, which contributes to heterogenous results, even when
drawn from this review. Methodology and substance heterogeneity
the same substance is being tested. Lastly, selected studies and language
among these studies do not allow the generation of a specific treatment
bias can create a restriction, since only reports in the English language
protocol or pharmaceutical approach. Despite the fact that the methods
published within the last five years were reviewed.
were well-explained in most studies, the extraction processes differed
Based on the data presented in the present review, flavonoids,
among studies. In this regard, a variety of solvents were used for ex-
especially flavan-3-ols and proanthocyanidins, appear to be the most
traction; some studies used ethanol [40–42,45,46,48,50,52–54],
promising candidates to be used in prevention or management of per-
whereas others used aqueous solvent [55], acetone [39,43], or me-
iodontal diseases. However, this does not mean that the consumption of
thanol [38,44,47,49]. Furthermore, the type of plants and extracts and
flavonoid-enriched foods reduces the risk of progression or prevents the
the concentrations applied were different, which can greatly influence

34
Table 2
In vivo inflammatory modulation effects of plant-derived polyphenols.
Study group Plant and/or active compound Methods Cells/tissues Results
K. Bunte et al.

Teixeira et al., Stemodia maritima L. extract 5 mg/kg body weight (bw) 1 h prior to placement of ligature and Maxillary alveolar bone of female Inhibition of alveolar bone loss
2017 once a day for the following 11 days until removal of the ligature Wister rats Decrease in TNF-α and cytokine-induced neutrophil chemoattractant
(CINC)-1 and increase in IL-10 levels in gingival tissues
Decrease in receptor activator of NF-κB (RANK), inducible nitric oxide
synthase (iNOS), IL-1β, and TNF-α mRNA expression
Ozden et al., 2017 Oligomeric proanthocyanidins 200 mg/kg bw by oral gavage, 4 groups: Mandibular molar region bone of Lower inflammatory cell number, higher connective tissue attachment
from grape seed A: Control group, B: two weeks prior to and six weeks after LPS male Sprague-Dawley rats level, and higher IL-10 and transforming growth factor beta (TGF-β)
ligature placement C: from ligation to two weeks after its removal, levels were measured in all groups, with group B having the highest IL-
D: two weeks from ligature removal 10 levels
Kostic et al., 2017 Rosmarinic acid from Salvia 200 mg/kg bw systemic administration by oral gavage twice a day Interdental papilla between the first Decrease in IL-1β, IL-6, and TNF-α levels
sclarea L. and second maxillary molars of Less inflammatory cells and more fibroblasts in the gingival tissue
Wistar rats
Hatipoglu et al., Crataeus orientalis M. Bieber 100 mg/kg bw systemic administration by oral gavage from the day Right mandibular first molars of rats Inhibition of periodontal inflammation and alveolar bone loss via
2015 of ligature placement throughout the following 11 days to ligature regulation of oxidative stress, and reduction in inflammatory cells and
removal osteoclasts in periodontal tissues
Gennaro et al., Green tea 7 mg/ml green tea ad libitum for 15, 30. 60 or 90 days after Hemi-maxilla of type I diabetic male Increase in osteoprotegerin (OPG) at 15, 60 and 90 days and increase in
2015 induction of diabetes at day 0 (n = 5 in each group) Wistar rats IL-10 at all times, increase in runt related transcription factor(RUNX)-2
and reduction in TNF-α and RANKL at 30, 60 and 90 days

35
Table 3
Ex vivo inflammatory modulation effects of plant-derived polyphenols.
Study group Plant and/or active compound Methods Cells/tissues Results

Lee et al., 2017 Terminalia chebula ethanolic extract (EETC) Medium containing dental plaque bacteria LPS (1 μg/ml) and/ Mouse-derived bone marrow Reduction in RANKL mRNA expression
or EETC (10 μg/ml) for 7 days macrophages (BMMs)
Kim et al., 2017 Sophorae Flos extract (SFE) BMMs were cultured with M-CSF (50 ng/ml) and RANKL Mouse-derived bone marrow Inhibition of RANKL-induced osteoclast differentiation via
(100 ng/ml) for 4 days to generate osteoclasts in the presence macrophages (BMMs) suppression of the NF-κB/ nuclear factor of activated T-cells,
or absence of SFE cytoplasmic 1(NFATc1) pathway
Walker et al., Magnolia officinalis (L.) extract Extract containing gum chewed by volunteers for 10 min, and Human oral epithelial cells from Reduction in IL-8 expression and oxidative stress
2016 collected cells were subsequently stimulated with LPS ex vivo 40 healthy adults
Granica et al., Pedunculagin, stachyurin, casuarynin, and gemin A, In the absence or presence of extracts/compounds at a final Human neutrophils derived from Gemin A inhibited NOS and MMP-9, IL-8, and IL-1β release,
2016 and ellagic acid derivatives from Geum urbanum L. concentration of 10 and 50 μg/ml/10 and 50 μM added 1 h blood of healthy adults but increased TNF-α secretion
root prior to stimulation with LPS (100 ng/ml)
Chin et al., 2016 2,3,5,4’-Tetrahydroxystilbene-2-O-beta-glucoside Pretreatment with 25 μM THSG followed by treatment with Human gingival fibroblasts from Suppression of NF-κB, TNF-α, IL-1β, and Il-6 release
(THSG) from Polygoni Multiflori 1 μg/ml P. gingivalis LPS for 3 h 40 healthy adults
Bin et al., 2015 Osmanthus fragrans ethanolic extract Cells were treated with increasing concentrations of extract Human periodontal ligament Inhibition of IL-6 at > 125 μg/ml and IL-8 > 62.5 μg/ml
(31.25, 62.5, 125, and 250 μg/ml) prior to P. gingivalis LPS cells obtained from 20 healthy
stimulation at a final concentration of 1 mg/ml adults
Kong et al., 2014 Angelica sinensis extract (ASE) BMMs were cultured in the presence of M-CSF (20 ng/ml) and Mouse-derived bone marrow Inhibition of RANKL-mediated osteoclast differentiation, and
RANKL (40 ng/ml) for 4 days in the presence or absence of ASE macrophages (BMMs) proto-oncogene c-Fos, protein c-Jun, and NFATc1 expression
Fitoterapia 132 (2019) 30–39
K. Bunte et al.

Table 4
Inflammatory modulation effects of plant-derived polyphenols in vitro.
Study group Plant and/or active compound Methods Cells/tissues Results

Ben Lagha et al., Highbush blueberry proanthocyanidins Cells were pretreated for 2 h with PACs (125–31.25 μg/ml) prior to Macrophage-like cells differentiated from U937 human Inhibition of NF-κB activation
2018 (PACs) stimulation with A. actinomycetemcomitans, followed by incubation monocytes Inhibition of IL-1β, IL-6, IL8, TNF-α, MMP-3,
for 24 h in the presence or absence of PACs and with or without and MMP-9 secretion
bacterial stimulation
Morin and Grenier EGCG from green tea and green tea 2-h stimulation with LPS followed by incubation with green tea 1:10 macrophage-like cells differentiated from U937 Reduction in MMP-3, −8, and − 9 secretion
2017 extract extract or EGCG human monocytes and primary human gingival by both green tea extract and EGCG at non-
fibroblast cell line HGF-1 cytotoxic concentrations
Lee et al., 2017 Terminalia chebula ethanolic extract Cells were exposed to serum-free medium containing LPS (1 μg/ RAW264.7 murine macrophage cells, immortalized Inhibition of prostaglandin E2 (PGE2), COX-
(EETC) ml) and/or EETC (10 mg/ml) for 24 h human oral keratinocytes (IHOK), immortalized human 2, and RANKL expression and osteoclast
gingival fibroblasts (IGF), and YD38 human gingival formation
epithelial cells
Cespedes et al., Quercetin, gallic acid, luteolin, and Cells were incubated in media with or without LPS (1.0 mg/ml), RAW264.7 murine macrophage cells Downregulation of iNOS and COX-2
2017 myricetin from Aristotelia chilensis and the samples (plant extracts, fractions, or pure com- pounds)
(Elaeocarpaceae), Maqui were subsequently applied and incubated for 12 h
Ben Lagha et al., Theaflavins from black tea Theaflavin preparation was applied at 31.25, 62.5, 125, and Macrophage-like cells differentiated from U937 human Reduction in IL-1β, TNF-α, IL-6, IL-8, MMP-
2017 250 mg/ml, and cells were subsequently stimulated with P. monocytes 3, -8, and -9

36
gingivalis and incubated for 4 h Inhibition of the NF-κB signaling pathway
Lombardo Bedran EGCG from green tea and A type- AC-PACs at 25 and 50 μg/ml, EGCG at 1 and 5 μg/ml Immortalized human gingival epithelial cell line OBA-9 Reduced secretion of GRO-α, G-CSF, IL-6, IL-
et al., 2015 cranberry proanthocyanidins (AC-PACs) and primary human gingival fibroblast cell line HGF-1 8, IP-10, and MCP-1 with both EGCG and
AC-PACs
Lombardo Bedran Theaflavin and theaflavin-3,3′-digallate 2-h pretreatment with black tea extract (100 and 200 μg/ml), Immortalized human oral epithelial cell line OBA-9 Reduction in IL-8 secretion with all tested
et al., 2015 from black tea and black tea extract theaflavin, theaflavin-3,3′-digallate, or EGCG (10 and 50 μg/ml) compounds
prior to A. actinomycetemcomitans LPS stimulation Increased hBD-1, hBD-2, and hBD-4
secretion following treatment with black tea
extract and theaflavin-3,3′-digallate
Lohr et al., 2015 Isoquercitrin and tannins from KB cells were incubated for 24 h prior to exposure to P. gingivalis KB cells derived from human oral epidermoid Inhibition of IL-1ß, IL-6, IL-8, and TNF-α
Rhododendron ferrugineum L. aqueous carcinoma expression
extract
Tipton et al., 2014 Proanthocyanidins (PACs) from cranberry Cells were incubated with IL-1β in the presence or absence of Smulow-Glickman (S-G) human gingival epithelial cells Inhibition of NF-κB activation, and activator
cranberry PACs for 6 days protein 1(AP-1)- and IL-1β-stimulated IL-6
expression
Tipton et al., 2013 Proanthocyanidins (PACs) from cranberry Cells were incubated with IL-17 in the presence or absence of Normal human gingival fibroblasts and Smulow- Reduction in IL-17-stimulated IL-6 and IL-8
cranberry PACs for 6 days Glickman (S-G) human gingival epithelial cells production
Tipton et al., 2013 Proanthocyanidins (PACs) from cranberry Aggressive periodontitis fibroblasts were incubated with PACs, Aggressive periodontitis and normal human gingival Increased IL-6 production in aggressive
with or without F. nucleatum or P. gingivalis LPS, for up to 6 days fibroblasts periodontitis HGFs
Reduction of IL-6 and MMP-3 production,
and inhibition of NF-κB in normal HGFs
Fitoterapia 132 (2019) 30–39
K. Bunte et al. Fitoterapia 132 (2019) 30–39

onset of periodontal disease. The hydrophilic compounds (e.g. flavonoid Funding


glycosides, proanthocyanidins, and anthocyanidins) are hardly ab-
sorbed following oral ingestion, unless they are transported by specific This research did not receive any specific grant from funding
carrier systems [93,94]. Most polyphenols are relatively hydrophilic agencies in the public, commercial, or not-for-profit sectors.
compounds due to the presence of polar hydroxyl groups in the aro-
matic parts of the molecules. Modification of these compounds (e.g. Acknowledgments
methylation of hydroxyl groups or prenylation) results in an increase in
lipophilicity, which leads to an increased cell membrane permeability The first author would like to acknowledge the contributions and
and a higher absorption rate. When polyphenols become bioavailable in comments by Dr. Ricarda Hess and Dr. Susanne Bierbaum from the
the respective pharmacokinetic compartment, a cascade of reactions is University of Dresden, Germany.
initiated that results in a decreased oxidation rate of lipids or proteins
within cells [95]; this process is mainly mediated by hydroxylation of References
the B-ring [15]. However, a high hydroxylation grade leads to higher
polarity of the molecules, and thus a reduced bioavailability in inflamed [1] W.J. Loesche, N.S. Grossman, Periodontal disease as a specific, albeit chronic, in-
tissues. For this reason, in vitro investigations of polyphenols do not fection: diagnosis and treatment, Clin. Microbiol. Rev. 14 (4) (2001) 727–752.
[2] I.L. Chapple, J.B. Matthews, The role of reactive oxygen and antioxidant species in
necessarily reflect the pharmacodynamic reality; which is why struc- periodontal tissue destruction, Periodontology 43 (2007) 160–232.
tural differences of polyphenols play a crucial role in their biological [3] A. Varela-Lopez, et al., Non-nutrient, naturally occurring phenolic compounds with
activities [16]. On the other hand, when used locally, e.g., as a mouth antioxidant activity for the prevention and treatment of periodontal diseases,
Antioxidants (Basel) 4 (3) (2015) 447–481.
rinse or gel, a favourable immunomodulatory and antibacterial effect [4] T. Beikler, T.F. Flemmig, Oral biofilm-associated diseases: trends and implications
can be achieved irrespective of tissue concentrations. Dabholkar et al., for quality of life, systemic health and expenditures, Periodontology 55 (1) (2011)
compared 0.12% chlorhexidine digluconate mouth rinse, a commercial 87–103.
[5] W.J. Peros, E.D. Savitt, The microbiology of periodontal disease, Clin. Microbiol.
mouth rinse containing extracts from Terminalia bellirica, Piper betle and
Newsl. 11 (7) (1989) 49–51.
Salvadora persica and another commercial mouth rinse containing Pu- [6] S.S. Socransky, Criteria for the infectious agents in dental caries and periodontal
nica granatum (pomegranate) and Camellia sinensis leaf extracts for their disease, J. Clin. Periodontol. 6 (7) (1979) 16–21.
[7] M.S. Tonetti, et al., Treatment of periodontitis and endothelial function, N. Engl. J.
antimicrobial effect against oral biofilm-forming bacteria in vitro. All
Med. 356 (9) (2007) 911–920.
three mouth rinses showed an antibacterial effect in varying con- [8] A. Cekici, et al., Inflammatory and immune pathways in the pathogenesis of peri-
centrations; however, there were statistically significant differences odontal disease, Periodontology 64 (1) (2014) 57–80.
between chlorhexidine digluconate and both herbal mouth rinses [12]. [9] N. Silva, et al., Host response mechanisms in periodontal diseases, J. Appl. Oral Sci.
23 (3) (2015) 329–355.
These findings indicate that chlorhexidine remains to be superior to the [10] S. Ji, Y.S. Choi, Y. Choi, Bacterial invasion and persistence: critical events in the
herbal mouth rinses, and thus may be considered the gold standard for pathogenesis of periodontitis? J. Periodontal Res. 50 (5) (2015) 570–585.
short-term use; however, a polyphenol-containing mouth rinse may be [11] J.W. Krayer, R.S. Leite, K.L. Kirkwood, Non-surgical chemotherapeutic treatment
strategies for the management of periodontal diseases, Dent. Clin. N. Am. 54 (1)
useful for long-term control of the microbial and inflammatory situa- (2010) 13–33.
tion avoiding the adverse effects of chlorhexidine digluconate. [12] C.S. Dabholkar, et al., Comparative evaluation of antimicrobial activity of pome-
Local delivery devices containing high concentrations of poly- granate-containing mouthwash against oral-biofilm forming organisms: an invitro
microbial study, J. Clin. Diagn. Res. 10 (3) (2016) ZC65–9.
phenols, especially flavonoids may be another way of implementing [13] H.D. Müller, et al., Cytotoxicity and antimicrobial activity of oral rinses in vitro,
these substances in the management or prevention of periodontitis. In Biomed. Res. Int. 2017 (2017).
this regard, a 4-week controlled, randomised, split-mouth, single-eva- [14] A. Scalbert, I.T. Johnson, M. Saltmarsh, Polyphenols: antioxidants and beyond, Am.
J. Clin. Nutr. 81 (1 Suppl) (2005) 215S–217S.
luator, masked study tested a green tea catechin gel for local applica- [15] S. Quideau, et al., Plant polyphenols: chemical properties, biological activities, and
tion in patients with chronic periodontitis, reporting a highly statisti- synthesis, Angew. Chem. Int. Ed. Engl. 50 (3) (2011) 586–621.
cally significant reduction (p < .001) in pocket-probing depth and [16] R. Tsao, Chemistry and biochemistry of dietary polyphenols, Nutrients 2 (12)
(2010) 1231–1246.
inflammation as compared with baseline [96]. When incorporated into
[17] T. Hussain, et al., Oxidative stress and inflammation: what polyphenols can do for
a controlled delivery device, one could further envision a clinically and Us? Oxidative Med. Cell. Longev. 2016 (2016) 7432797.
cost-effective local therapy. Moreover, preventive measures and addi- [18] G.P.P. Lima, F. Vianello, C.R. Corrêa, R.A. da Silva Campos, M.G. Borguini,
tional supportive therapy are beneficial and can be more efficient in Polyphenols in fruits and vegetables and its effect on human health, Food Nutr. Sci.
2014 (5) (2014) 1065–1082.
terms of cost savings than mechanical debridement alone [97]. It is [19] C. Manach, et al., Polyphenols: food sources and bioavailability, Am. J. Clin. Nutr.
therefore necessary to identify substances that are not yet deployed and 79 (5) (2004) 727–747.
could serve the adult dentate population to facilitate substantial ag- [20] N.S. Dhalla, R.M. Temsah, T. Netticadan, Role of oxidative stress in cardiovascular
diseases, J. Hypertens. 18 (6) (2000) 655–673.
gregate improvements in periodontal health at an acceptable cost. [21] A.C. Maritim, R.A. Sanders, J.B. Watkins 3rd, Diabetes, oxidative stress, and anti-
oxidants: a review, J. Biochem. Mol. Toxicol. 17 (1) (2003) 24–38.
[22] N. Khansari, Y. Shakiba, M. Mahmoudi, Chronic inflammation and oxidative stress
as a major cause of age-related diseases and cancer, Recent Patents Inflamm.
5. Conclusions Allergy Drug Discov. 3 (1) (2009) 73–80.
[23] K. Ganesan, B. Xu, A critical review on polyphenols and health benefits of black
Despite the variety of substances and concentrations applied and the soybeans, Nutrients (2017) 9(5).
[24] X.M. Liu, et al., Dietary total flavonoids intake and risk of mortality from all causes
heterogeneous methodology used in the reviewed studies, the anti- and cardiovascular disease in the general population: A systematic review and
bacterial and anti-inflammatory effects of plant-derived polyphenols meta-analysis of cohort studies, Mol. Nutr. Food Res. (2017) 61(6).
are readily identifiable. The positive findings regarding the inhibition of [25] Y.J. Liu, et al., Dietary flavonoids intake and risk of type 2 diabetes: a meta-analysis
of prospective cohort studies, Clin. Nutr. 33 (1) (2014) 59–63.
periodontal pathogen activities and the reduction in the host in-
[26] X. He, L.M. Sun, Dietary intake of flavonoid subclasses and risk of colorectal cancer:
flammatory and immune responses following the use of polyphenols evidence from population studies, Oncotarget 7 (18) (2016) 26617–26627.
warrant further studies aiming to identify the substances that can pre- [27] G. Grosso, et al., A comprehensive meta-analysis on dietary flavonoid and lignan
vent tissue breakdown or suppress periodontal pathogens and reverse intake and cancer risk: level of evidence and limitations, Mol. Nutr. Food Res.
(2017) 61(4).
the intraoral dysbiosis. [28] O.N. Styshova, et al., Main constituents of polyphenol complex from seagrasses of
the genus Zostera, their antidiabetic properties and mechanisms of action, Exp.
Ther. Med. 13 (5) (2017) 1651–1659.
[29] R. Mattera, et al., Effects of polyphenols on oxidative stress-mediated injury in
Declarations of interest cardiomyocytes, Nutrients (2017) 9(5).
[30] M. Vitale, et al., Polyphenol intake and cardiovascular risk factors in a population
None. with type 2 diabetes: The TOSCA.IT study, Clin. Nutr. 36 (6) (2016) 1686–1692.

37
K. Bunte et al. Fitoterapia 132 (2019) 30–39

[31] H.C. Tsai, et al., Citrus polyphenol for oral wound healing in oral ulcers and peri- (2015) e0134784.
odontal diseases, J. Formos. Med. Assoc. 115 (2) (2016) 100–107. [61] J. Lee, et al., Use of ethanol extracts of Terminalia chebula to prevent periodontal
[32] M. Gomez-Florit, M. Monjo, J.M. Ramis, Identification of quercitrin as a potential disease induced by dental plaque bacteria, BMC Complement. Altern. Med. 17 (1)
therapeutic agent for periodontal applications, J. Periodontol. 85 (7) (2014) (2017) 113.
966–974. [62] J.M. Kim, et al., Sophorae Flos extract inhibits RANKL-induced osteoclast differ-
[33] A. Furiga, et al., In vitro anti-bacterial and anti-adherence effects of natural poly- entiation by suppressing the NF-kappaB/NFATc1 pathway in mouse bone marrow
phenolic compounds on oral bacteria, J. Appl. Microbiol. 105 (5) (2008) cells, BMC Complement. Altern. Med. 17 (1) (2017) 164.
1470–1476. [63] J. Walker, et al., Magnolia officinalis L. fortified gum improves resistance of oral
[34] G. Lohr, et al., Polyphenols from Myrothamnus flabellifolia Welw. inhibit in vitro epithelial cells against inflammation, Am. J. Chin. Med. 44 (6) (2016) 1167–1185.
adhesion of Porphyromonas gingivalis and exert anti-inflammatory cytoprotective [64] S. Granica, et al., Effects of Geum urbanum L. root extracts and its constituents on
effects in KB cells, J. Clin. Periodontol. 38 (5) (2011) 457–469. polymorphonuclear leucocytes functions. Significance in periodontal diseases, J.
[35] C. Stoicov, R. Saffari, J.M. Houghton, Green tea inhibits Helicobacter growth in vivo Ethnopharmacol. 188 (2016) 1–12.
and in vitro, Int. J. Antimicrob. Agents 33 (5) (2009) 473–478. [65] Y.T. Chin, et al., 2,3,5,4'-Tetrahydroxystilbene-2-O-beta-glucoside isolated from
[36] S. Archana, J. Abraham, Comparative Analysis of Antimicrobial Activity of Leaf polygoni multiflori ameliorates the development of periodontitis, Mediat. Inflamm.
Extracts From Fresh Green Tea and Black Tea on Pathogens, Vol. 1 (2011), pp. 2016 (2016) 6953459.
149–152. [66] H. Bin, C. Huangqin, S. Longquan, The ethanol extract of Osmanthus fragrans at-
[37] D. Moher, et al., Preferred reporting items for systematic reviews and meta-ana- tenuates Porphyromonas gingivalis lipopolysaccharide-stimulated inflammatory
lyses: the PRISMA statement, Int. J. Surg. 8 (5) (2010) 336–341. effect through the nuclear factor erythroid 2-related factor-mediated antioxidant
[38] T. Kariu, et al., Inhibition of gingipains and Porphyromonas gingivalis growth and signalling pathway, Arch. Oral Biol. 60 (7) (2015) 1030–1038.
biofilm formation by prenyl flavonoids, J. Periodontal Res. 52 (1) (2017) 89–96. [67] L. Kong, et al., Angelica sinensis extract inhibits RANKL-mediated osteoclastogen-
[39] A. de Oliveira Caleare, et al., Flavan-3-ols and proanthocyanidins from Limonium esis by down-regulated the expression of NFATc1 in mouse bone marrow cells, BMC
brasiliense inhibit the adhesion of Porphyromonas gingivalis to epithelial host cells Complement. Altern. Med. 14 (2014) 481.
by interaction with gingipains, Fitoterapia 118 (2017) 87–93. [68] M.-P. Morin, D. Grenier, Regulation of matrix metalloproteinase secretion by green
[40] A. Ben Lagha, B. Haas, D. Grenier, Tea polyphenols inhibit the growth and virulence tea catechins in a three-dimensional co-culture model of macrophages and gingival
properties of Fusobacterium nucleatum, Sci. Rep. 7 (2017) 44815. fibroblasts, Arch. Oral Biol. 75 (2017) 89–99.
[41] A. Ben Lagha, D. Grenier, Black tea theaflavins attenuate Porphyromonas gingivalis [69] C.L. Cespedes, et al., The chilean superfruit black-berry Aristotelia chilensis
virulence properties, modulate gingival keratinocyte tight junction integrity and (Elaeocarpaceae), Maqui as mediator in inflammation-associated disorders, Food
exert anti-inflammatory activity, J. Periodontal Res. 52 (3) (2017) 458–470. Chem. Toxicol. 108 (Pt B) (2017) 438–450.
[42] S. Mallikarjun, et al., Antimicrobial efficacy of Tulsi leaf (Ocimum sanctum) extract [70] T.B. Lombardo Bedran, D. Palomari Spolidorio, D. Grenier, Green tea polyphenol
on periodontal pathogens: an in vitro study, J. Indian Soc. Periodontol. 20 (2) epigallocatechin-3-gallate and cranberry proanthocyanidins act in synergy with
(2016) 145–150. cathelicidin (LL-37) to reduce the LPS-induced inflammatory response in a three-
[43] J. Schmuch, et al., Extract from Rumex acetosa L. for prophylaxis of periodontitis: dimensional co-culture model of gingival epithelial cells and fibroblasts, Arch. Oral
inhibition of bacterial in vitro adhesion and of gingipains of Porphyromonas gin- Biol. 60 (6) (2015) 845–853.
givalis by epicatechin-3-O-(4beta– > 8)-epicatechin-3-O-gallate (procyanidin-B2- [71] T.B. Lombardo Bedran, et al., Black tea extract and its theaflavin derivatives inhibit
Di-gallate), PLoS One 10 (3) (2015) (p. e0120130). the growth of periodontopathogens and modulate interleukin-8 and beta-defensin
[44] P. Rodanant, et al., Coumarins and flavonoid from Murraya paniculata (L.) Jack: secretion in oral epithelial cells, PLoS One 10 (11) (2015) e0143158.
antibacterial and anti-inflammation activity, Pak. J. Pharm. Sci. 28 (6) (2015) [72] D.A. Tipton, T.B. Carter, M. Dabbous, Inhibition of interleukin 1beta-stimulated
1947–1951. interleukin-6 production by cranberry components in human gingival epithelial
[45] A. Ben Lagha, et al., Wild blueberry (Vaccinium angustifolium Ait.) polyphenols cells: effects on nuclear factor kappaB and activator protein 1 activation pathways,
target fusobacterium nucleatum and the host inflammatory response: potential in- J. Periodontal Res. 49 (4) (2014) 437–447.
novative molecules for treating periodontal diseases, J. Agric. Food Chem. 63 (31) [73] D.A. Tipton, et al., Inhibition of interleukin-17-stimulated interleukin-6 and -8
(2015) 6999–7008. production by cranberry components in human gingival fibroblasts and epithelial
[46] J.R. Villinski, et al., Pyrano-isoflavans from Glycyrrhiza uralensis with antibacterial cells, J. Periodontal Res. 48 (5) (2013) 638–646.
activity against Streptococcus mutans and Porphyromonas gingivalis, J. Nat. Prod. [74] D.A. Tipton, J.P. Babu, M. Dabbous, Effects of cranberry components on human
77 (3) (2014) 521–526. aggressive periodontitis gingival fibroblasts, J. Periodontal Res. 48 (4) (2013)
[47] J.K. Patra, et al., Antibacterial effect of crude extract and metabolites of Phytolacca 433–442.
americana on pathogens responsible for periodontal inflammatory diseases and [75] M.P. Morin, D. Grenier, Regulation of matrix metalloproteinase secretion by green
dental caries, BMC Complement. Altern. Med. 14 (2014) 343. tea catechins in a three-dimensional co-culture model of macrophages and gingival
[48] S. Shetty, et al., An in-vitro evaluation of the efficacy of garlic extract as an anti- fibroblasts, Arch. Oral Biol. 75 (2017) 89–99.
microbial agent on periodontal pathogens: a microbiological study, Ayu 34 (4) [76] Y. Cai, et al., Green tea epigallocatechin-3-gallate alleviates Porphyromonas gin-
(2013) 445–451. givalis-induced periodontitis in mice, Int. Immunopharmacol. 29 (2) (2015)
[49] M. Patel, R. Naidoo, F.J. Owotade, Inhibitory effect of Dodonaea viscosa var. an- 839–845.
gustifolia on the virulence properties of the oral pathogens Streptococcus mutans [77] E. Palm, H. Khalaf, T. Bengtsson, Suppression of inflammatory responses of human
and Porphyromonas gingivalis, Evid. Based Complement. Alternat. Med. 2013 gingival fibroblasts by gingipains from Porphyromonas gingivalis, Mol Oral
(2013) 624089. Microbiol 30 (1) (2015) 74–85.
[50] O.A. Gonzalez, et al., Antibacterial effects of blackberry extract target period- [78] R.J. Lamont, H.F. Jenkinson, Life below the gum line: pathogenic mechanisms of
ontopathogens, J. Periodontal Res. 48 (1) (2013) 80–86. Porphyromonas gingivalis, Microbiol. Mol. Biol. Rev. 62 (4) (1998) 1244–1263.
[51] A. Araghizadeh, J. Kohanteb, M.M. Fani, Inhibitory activity of green tea (Camellia [79] I. Olsen, J. Potempa, Strategies for the inhibition of gingipains for the potential
sinensis) extract on some clinically isolated cariogenic and periodontopathic bac- treatment of periodontitis and associated systemic diseases, J. Oral Microbiol. 6
teria, Med. Princ. Pract. 22 (4) (2013) 368–372. (2014).
[52] A. Herrera Herrera, et al., Susceptibility of Porphyromonas gingivalis and [80] T. Beikler, et al., Sequence variations in rgpA and rgpB of Porphyromonas gingivalis
Streptococcus mutans to antibacterial effect from Mammea americana, Adv. in periodontitis, J. Periodontal Res. 40 (3) (2005) 193–198.
Pharmacol. Sci. 2014 (2014) 384815. [81] G. Hajishengallis, Porphyromonas gingivalis-host interactions: open war or in-
[53] L. Heyman, et al., Combined antioxidant effects of Neem extract, bacteria, red blood telligent guerilla tactics? Microbes Infect. 11 (6–7) (2009) 637–645.
cells and Lysozyme: possible relation to periodontal disease, BMC Complement. [82] M.D. Carlisle, R.N. Srikantha, K.A. Brogden, Degradation of human alpha- and beta-
Altern. Med. 17 (1) (2017) 399. defensins by culture supernatants of Porphyromonas gingivalis strain 381, J. Innate
[54] A. Ben Lagha, G. LeBel, D. Grenier, Dual action of highbush blueberry proantho- Immun. 1 (2) (2009) 118–122.
cyanidins on Aggregatibacter actinomycetemcomitans and the host inflammatory [83] S. Joly, et al., Human beta-defensins 2 and 3 demonstrate strain-selective activity
response, BMC Complement. Altern. Med. 18 (1) (2018) 10. against oral microorganisms, J. Clin. Microbiol. 42 (3) (2004) 1024–1029.
[55] G. Lohr, T. Beikler, A. Hensel, Inhibition of in vitro adhesion and virulence of [84] G. Hajishengallis, et al., Complement involvement in periodontitis: molecular me-
Porphyromonas gingivalis by aqueous extract and polysaccharides from chanisms and rational therapeutic approaches, Adv. Exp. Med. Biol. 865 (2015)
Rhododendron ferrugineum L. A new way for prophylaxis of periodontitis? 57–74.
Fitoterapia 107 (2015) 105–113. [85] P.G. Stathopoulou, et al., The host cytokine response to Porphyromonas gingivalis is
[56] A.H. Teixeira, et al., Stemodia maritima L. extract decreases inflammation, oxidative modified by gingipains, Oral Microbiol. Immunol. 24 (1) (2009) 11–17.
stress, and alveolar bone loss in an experimental periodontitis rat model, Front. [86] C.C. Calkins, et al., Inactivation of tumor necrosis factor-alpha by proteinases
Physiol. 8 (2017) 988. (gingipains) from the periodontal pathogen, Porphyromonas gingivalis.
[57] F.O. Ozden, et al., Effects of grape seed extract on periodontal disease: an experi- Implications of immune evasion, J. Biol. Chem. 273 (12) (1998) 6611–6614.
mental study in rats, J. Appl. Oral Sci. 25 (2) (2017) 121–129. [87] M. Benedyk, et al., Gingipains: critical factors in the development of aspiration
[58] M. Kostic, et al., Anti-inflammatory effect of the Salvia sclarea L. ethanolic extract pneumonia caused by Porphyromonas gingivalis, J. Innate Immun. 8 (2) (2016)
on lipopolysaccharide-induced periodontitis in rats, J. Ethnopharmacol. 199 (2017) 185–198.
52–59. [88] M.A. Gorman, et al., Structure of the lysine specific protease Kgp from
[59] M. Hatipoglu, et al., The effectiveness of Crataegus orientalis M Bieber. (Hawthorn) Porphyromonas gingivalis, a target for improved oral health, Protein Sci. 24 (1)
extract administration in preventing alveolar bone loss in rats with experimental (2015) 162–166.
periodontitis, PLoS One 10 (6) (2015) (p. e0128134). [89] J.W. Smalley, T. Olczak, Heme acquisition mechanisms of Porphyromonas gingi-
[60] G. Gennaro, et al., Green tea modulates cytokine expression in the periodontium valis - strategies used in a polymicrobial community in a heme-limited host en-
and attenuates alveolar bone resorption in type 1 diabetic rats, PLoS One 10 (8) vironment, Mol Oral Microbiol 32 (1) (2017) 1–23.

38
K. Bunte et al. Fitoterapia 132 (2019) 30–39

[90] Y. Hosokawa, et al., (−)-Epigallocatechin-3-gallate inhibits CC chemokine ligand [94] M. D'Archivio, et al., Bioavailability of the polyphenols: status and controversies,
11 production in human gingival fibroblasts, Cell. Physiol. Biochem. 31 (6) (2013) Int. J. Mol. Sci. 11 (4) (2010) 1321–1342.
960–967. [95] S.V. Verstraeten, C.G. Fraga, P.I. Oteiza, Interactions of flavan-3-ols and procyani-
[91] T.K. Mao, et al., Effect of cocoa flavanols and their related oligomers on the se- dins with membranes: mechanisms and the physiological relevance, Food Funct. 6
cretion of interleukin-5 in peripheral blood mononuclear cells, J. Med. Food 5 (1) (1) (2015) 32–41.
(2002) 17–22. [96] V.K. Chava, B.D. Vedula, Thermo-reversible green tea catechin gel for local appli-
[92] I. Andujar, et al., Cocoa polyphenols and their potential benefits for human health, cation in chronic periodontitis: a 4-week clinical trial, J. Periodontol. 84 (9) (2013)
Oxidative Med. Cell. Longev. 2012 (2012) 906252. 1290–1296.
[93] C. Manach, et al., Bioavailability and bioefficacy of polyphenols in humans. I. [97] A.A. Antczak-Bouckoms, M.C. Weinstein, Cost-effectiveness analysis of periodontal
Review of 97 bioavailability studies, Am. J. Clin. Nutr. 81 (1 Suppl) (2005) disease control, J. Dent. Res. 66 (11) (1987) 1630–1635.
230s–242s.

39

You might also like