You are on page 1of 11

| |

Received: 8 February 2019    Revised: 28 October 2019    Accepted: 25 November 2019

DOI: 10.1111/jre.12722

ORIGINAL ARTICLE

The therapeutic role of baicalein in combating experimental


periodontitis with diabetes via Nrf2 antioxidant signaling
pathway

Chunhui Zhu1,2 | Ying Zhao2 | Xiaoyan Wu2 | Cui Qiang1 | Jin Liu1,2 | Jianfeng Shi1 |


Jianzhong Gou2 | Dandan Pei1  | Ang Li1,2

1
Key Laboratory of Shaanxi Province for
Craniofacial Precision Medicine Research, Abstract
College of Stomatology, Xi'an Jiaotong Background and objective: Oxidative stress has been suggested as an impor-
University, Xi'an, China
2 tant pathogenic factor contributing to chronic periodontitis with diabetes mellitus
Department of Periodontology, College
of Stomatology, Xi’an Jiaotong University, (CPDM). Previous studies have revealed the potential therapeutic properties of bai-
Xi'an, China
calein (BCI) in oxidative stress-related diseases; however, the antioxidant effects of
Correspondence BCI on therapy for individual with CPDM remain largely unexplored. Nuclear fac-
Ang Li, Key Laboratory of Shaanxi Province
tor erythroid 2-related factor 2 (Nrf2) plays a critical role in cellular defence against
for Craniofacial Precision Medicine
Research, College of Stomatology, Xi'an oxidative stress. In this study, we aim to determine whether BCI prevents diabetes-
Jiaotong University, Xi'an, China.
related periodontal tissue destruction by regulating Nrf2 signaling pathway.
Email: drliang@mail.xjtu.edu.cn
Material and methods: Human gingival epithelial cells (hGECs) were challenged with
Funding information
high glucose (HG, 25 mmol/L) and/or lipopolysaccharide (LPS, 20 µg/mL). Reactive
Fundamental Research Funds for the
Central Universities, Grant/Award Number: oxygen species (ROS) were detected by fluorescence-activated cell sorting. The
xjj2017025 and xjj2017097; National
changes of antioxidant-related genes, including Nrf2, catalase (Cat), glutamate-
Natural Science Foundation of China, Grant/
Award Number: 81901019 and 81870798 cysteine ligase catalytic subunit (Gclc), superoxide dismutase 1 (Sod1), and superoxide
dismutase 2 (Sod2), were quantified by real-time PCR. The localization of phospho-
Nrf2 (pNrf2, S40) in the nucleus was detected by immunofluorescence staining and
laser scanning confocal microscope (LSCM). PNrf2 and total form of Nrf2 were de-
termined using western blot. The above indicators together with mitochondrial mem-
brane potential (MMP) were further investigated in hGECs pre-treated with different
concentrations of BCI (0.01, 0.1, or 0.5 µg/mL) before stimulated with HG plus LPS
(GP). Finally, the role of BCI in activating Nrf2 signaling pathway and relieving the
alveolar bone absorption was examined in the CPDM model of Sprague Dawley rats.
CPDM rats were oral gavaged with BCI (50, 100, or 200 mg/kg daily). The pNrf2 was
detected by immunohistochemistry, and the alveolar bone absorption was examined
by microcomputed tomography.
Results: Our results showed that ROS were significantly increased in both groups
of HG and LPS, with the strongest generation in the GP group. In terms of ROS-
related gene expression, we found that the mRNA levels of Nrf2, Cat, Gclc, Sod1,
and Sod2 were significantly decreased in HG and LPS groups. In consistent with the
strongest induction of ROS in GP group, the gene expression in GP group was further

Dandan Pei and Ang Li have contributed equally to the work.

J Periodont Res. 2020;55:381–391. wileyonlinelibrary.com/journal/jre © 2019 John Wiley & Sons A/S.     381 |
Published by John Wiley & Sons Ltd
|
382       ZHU et al.

decreased as compared to those of HG and LPS groups. Also, the expression of pNrf2
exhibited the same trend with the expression of those antioxidant genes. However,
the generation of ROS and the loss of mitochondrial membrane potential induced by
GP were abolished by pre-treatment with different concentrations of BCI (0.01, 0.1,
or 0.5 µg/mL). Interestingly, we observed that BCI promoted the nucleus transloca-
tion of pNrf2, as well as the gene expression levels of pNrf2 and its target genes
(Cat, Gclc, Sod1, and Sod2). Finally, in the CPDM animal model, we found that BCI (at
concentrations: 50, 100, and 200 mg/kg) markedly increased the number of pNrf2-
positive cells in periodontal tissue and mitigated the alveolar bone loss.
Conclusions: Our data revealed a potential role for clinic application of BCI under
CPDM conditions, suggesting a new therapeutic drug for CPDM patients.

KEYWORDS

adjunctive periodontal therapy, antioxidants, diabetes, oxidative stress

1 |  I NTRO D U C TI O N activated Nrf2 to nucleus up-regulates the expression of phase 2


enzyme genes, including catalase (Cat), glutamate-cysteine ligase
1
Periodontitis is a ubiquitous and irreversible inflammatory disease. catalytic subunit (Gclc), and superoxide dismutase 1 and 2 (Sod1 and
Some systemic diseases, such as diabetes mellitus (DM), have been Sod2). Phosphorylation of Nrf2 at serine40 has been confirmed to
proved to be closely associated with periodontitis. 2 The risk of peri- play a key mechanistic role in resisting oxidative stress.16 It has been
odontitis was reported approximately three times higher in patients proved that BCI possesses the ability to activate Nrf2 signaling path-
3
with DM than in those without DM. Even if the patients underwent way in oxidative stress-related diseases, including vitiligo, neurode-
the same periodontal treatment, DM patients with poor glycaemic generation, and cardiomyopathy.17-19
control could not achieve the same curative effect as non-diabet- Therefore, in the present study, we hypothesized that BCI ex-
ics.4 Numerous studies have been implemented to reveal the un- erts its protective function against diabetes-related periodontal
derling mechanisms contributing to chronic periodontitis with DM tissue damage by activating the Nrf2 antioxidant system. First, we
(CPDM), in which oxidative stress has been suggested as an import- established the cellular CPDM model using human gingival epithe-
ant pathogenic factor in this composite disease. 5,6 Clinically, the lial cells (hGECs) and characterized this model by determining the
immunoglobulin G antibodies to periodontitis-related pathogens ROS production, the expression levels of pNrf2 protein and Nrf2-
were positively correlated with the levels of serum reactive oxygen downstream genes (Cat, Gclc, Sod1, and Sod2). Based on this model,
species (ROS) metabolites.7 Moreover, high-glucose exposure could we investigated the effects of BCI, including mitochondrial mem-
induce higher gingival oxidative stress, which enhanced alveolar brane potential (MMP), the phosphorylation and translocation of
bone resorption in CPDM.6 These studies indicated that restricting Nrf2, as well as the expression levels of Nrf2-downstream genes.
the ROS level to a certain range could be beneficial for periodontal Particularly, we observed that BCI delivered by oral gavage into rat
health. Therefore, effective antioxidants were believed to have the CPDM model could halt the alveolar bone absorption. Taken to-
potential function to ameliorate CPDM by relieving ROS-induced gether, our findings might provide a novel potential drug for patients
oxidative stress.8 with CPDM.
Baicalein (BCI) is a natural polyphenolic flavonoid component
extracted from Scutellaria baicalensis Georgi. Diverse pharmaco-
logical properties of BCI were addressed, such as antioxidant, an- 2 | M ATE R I A L A N D M E TH O DS
ti-inflammatory, anti-bacterial, and immunomodulating effects.9-12
Jiang et al suggested that the healthy benefits of BCI are mainly at- 2.1 | Study design
tributed to its antioxidant properties.13 Nevertheless, the effect on
protecting periodontal tissue of BCI through its antioxidant effect 2.1.1 | In vitro experiments
has not yet been elucidated. Our previous study found that BCI was
beneficial for periodontal bone regeneration, whereas the underly- For dose-response studies, hGECs were treated with glucose
ing mechanisms remained largely unknown.14 One of the primary or Porphyromonas gingivalis lipopolysaccharide (P gingivalis LPS;
cellular antioxidant pathways against ROS is mediated by nuclear Invivogen) at the indicated concentration, respectively (glucose: 5.5,
factor erythroid 2-related factor 2 (Nrf2).15 The translocation of 12.5, and 25  mmol/L; LPS: 2.5, 5, 10, and 20  µg/mL). The typical
ZHU et al. |
      383

TA B L E 1   Specific primer sequences used for real-time PCR analysis

Abbreviations GenBank number Product size (bp) Primer sequence (5′-3′)

Cat NM_001752.3 145 Forward: TGAGGTTGAACAGATAGC


Reverse: CACAGGTATATGAAGATAATTG
Gclc NM_001197115 90 Forward: GCAAGGCCCAGAACAGCACG
Reverse: TCCCTCATCCATCTGGCAACTGT
Sod1 NM_000454.4 109 Forward: CCGATGTGTCTATTGAAGATTCTG
Reverse: TTTCCACCTTTGCCCAAGTC
Sod2 NM_001322820.1 164 Forward: TGGTGGTCATATCAATCATAGC
Reverse: AACCTGAGCCTTGGACAC
Nrf2 NM_001313904.1 215 Forward: TGCCCCTGGAAGTGTCAAACA
Reverse: CAACAGGGAGGTTAATGATTT
β-actin NM_001101.3 85 Forward: TGTTACAGGAAGTCCCTTGCCATC
Reverse: CTGTGTGGACTTGGGAGAGGAC

pro-inflammatory mediators, including tumor necrosis factor-alpha 2.2 | Cell culture


(Tnf-α), interleukin 1β (Il-1β), Interleukin 6 (Il-6), and interleukin 8
(Il-8), were detected to determine the optimum concentrations of Human gingival epithelial cells were purchased from Hua Tuo
glucose and LPS to simulate CPDM. After the CPDM model was es- Biotechnology Co., Ltd. and incubated in Dulbecco's Modified
tablished in hGECs, cells were divided into four groups: 5.5 mmol/L Eagle's Medium (Gibco Life Technologies) supplemented with
glucose (Con group); 25 mmol/L glucose (HG group); 5.5 mmol/L glu- 10% fetal bovine serum (Gibco Life Technologies) and antibiotics
cose + 20 µg/mL LPS (LPS group); and 25 mmol/L glucose + 20 µg/ (penicillin 100  U/mL and streptomycin 100  µg/mL; Invitrogen Life
mL LPS (GP group). The oxidative stress level of hGECs in different Technologies). Cells were cultured at 37°C in a humidified atmos-
conditions was assessed by ROS production, Nrf2-related antioxi- phere with 95% air, 5% carbon dioxide. Culture medium was replaced
dant genes expression (Cat, Gclc, Sod1, and Sod2), cellular pNrf2 lo- every three days. The cells were passaged when they reached ~ 80%
calization, and Nrf2 protein expression. Then, cells were incubated confluence using 0.25% trypsin-EDTA (Gibco Life Technologies).
with various concentrations of BCI (0.01, 0.1, 0.5, 1, and 10 µg/mL)
for 24 hours in order to investigate the effect of BCI on the viability
of the hGECs. Cells were pre-treated with BCI (0.01, 0.1, 0.5 µg/mL) 2.3 | Cell viability assay
for 1 hour before stimulating with GP. Once the optimum concentra-
tions of BCI were determined, cells were divided into the following Human gingival epithelial cells were seeded in 96-well plates at a
groups: Con group; GP group; BCI + GP groups (exposure of cells to density of 5  ×  103 cells per well. After 48  hours, cell vitality and
GP before pre-incubated BCI for 1 hour with a range of concentra- proliferation were assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-
tions of 0.01, 0.1, 0.5 µg/mL). Antioxidant capacity of BCI was deter- diphenyltetrazolium bromide (MTT) (Sigma-Aldrich) according to
mined by MMP, ROS production, Nrf2-related antioxidant genes, and the manufacturer's protocol. 20 The optical density (OD) at 450 nm
pNrf2 protein expression. was measured using a microplate reader (Bio-Rad™ Model 680).
Experimental values were normalized to those of the untreated cells.

2.1.2 | In vivo experiment


2.4 | Pro-inflammatory and Nrf2-related
Sprague Dawley rats were divided randomly into four groups antioxidative genes expression
(N = 10 per group): normal control (Con group); ligature and P gin-
givalis 33 277 infection at the maxillary second molars (PD group); The expression of pro-inflammatory and Nrf2-related antioxidant
intraperitoneally injected with 35 mg/kg streptozotocin (DM group); genes was analyzed by real-time PCR as described previously.21
1  month after confirmation of diabetes, periodontitis was induced Briefly, hGECs were plated on 6-well plates (2.5  ×  105 cells per
for another 1 month (CPDM group). The animal model was verified well). After 48 hours, the total RNA was extracted using TRIzol rea-
by characterizing the alveolar bone absorption, fasting blood glu- gent (Invitrogen Life Technologies) and its concentration was deter-
cose, and insulin sensitivity index. Then, the effects of BCI on acti- mined by the NanoDrop 2000c spectrophotometer (Thermo Fisher
vating Nrf2 and inhibiting alveolar bone resorption in CPDM animal Scientific). Equal amounts of 2 μg total RNA were reverse-transcribed
model were assessed at three different doses (50, 100, or 200 mg/ using a First Strand cDNA Synthesis Kit (Fermentas) according to the
kg/d), via oral gavage for 1 month. manufacturer's instruction. Gene expression was analyzed by an iQ5
|
384       ZHU et al.

kit (BIO-RAD) with SYBR® Premix Ex Taq™ II enzyme (TaKaRa) and with corresponding primary antibodies. The following primary anti-
normalized to the expression of β-actin. The sequencing primers of bodies were used: rabbit anti-pNrf2 antibody (diluted 1:500; Abcam,
Tnf-α, Il-1β, Il-6, Il-8, and β-actin were listed in Table S1. In addition, the cat. no. 76026), rabbit anti-Nrf2 antibody (diluted 1:500; Abcam,
sequencing primers of human Cat, Gclc, Sod1, Sod2, Nrf2, and β-actin cat. no. 31163) and rabbit anti-lamin B (diluted 1:1000; Abcam, cat.
were listed in Table 1. no. 32535). Membranes were incubated at room temperature for
2  hours with HRP-conjugated anti-rabbit IgG (diluted 1:3000; Cell
Signaling Technology, cat. no. 7074) or anti-mouse IgG secondary
2.5 | ROS generation antibodies (diluted 1:3000; Cell Signaling Technology, cat. no. 7076).
Then, immunoreactive bands were detected using the ECL detec-
The ROS production was quantified using 2,7-dichlorodihydrofluo- tion kit (Millipore). The band intensity was quantitated with Image
rescein diacetate (DCFH-DA, Sigma-Aldrich) by fluorescence-acti- J software.
vated cell sorting (FACS). HGECs were plated in 6-well plates at a
density of 1.5 × 106 for 24 hours. Cells were incubated for 48 hours
with the corresponding treatments and loaded with 10  µmol/L 2.8 | MMP
DCFH-DA for another 1  hour at 37°C. Cells were collected and
analyzed by FACS according to the manufacturer's instructions. 22 5,5′,6,6′-tetrachloro-1,1′,3,3′ tetraethylbenzimidazolylcarbocya-
Intracellular DCF fluorescence was detected at wavelengths of nine iodide (JC-1) kit (Beyotime Biotechnology Co., Ltd.) combined
488 nm for excitation and 525 nm for emission using fluorescence with Mitochondrial Membrane Potential Assay Kit (Beyotime
microplate reader (Thermo Fischer Scientific) and quantified. Biotechnology Co., Ltd.) were employed to measure mitochondrial
depolarization in hGECs according to instruction of manufacturer. 25
Briefly, hGECs were seeded in laser confocal dishes at a density of
2.6 | Cell localization of pNrf2 8  ×  10 4 cells per well and they were exposed to different stimu-
lation after 24  hours incubation. JC-1 accumulates in depolarized
Cellular localization of pNrf2 was examined by laser scanning confo- mitochondria as J-monomers emitting green fluorescence and ac-
cal microscopy (LSCM) as described previously. 23 Briefly, cells were cumulates in healthy mitochondria to form J-aggregates emitting
4
cultured in 20-mm glass-bottom dishes at a density of 5 × 10 cells red fluorescence. HGECs were incubated with JC-1 dye (10 µmol/L
per dish. After treating for 48 hours, cells were fixed in paraformal- final) for 20 min at 37°C in dark. The results of the assay were ob-
dehyde for 10 minutes at room temperature, washed and incubated tained by LSCM (J-aggregates: excitation/emission  =  525/590 nm;
with an anti-pNrf2 antibody (1:50 dilution; Abcam, cat. no. 76026) JC-1 monomers: excitation/emission  =  490/530  nm). The ratio of
for 12 hours at 4°C. Then, cells were incubated with goat anti-rabbit JC-1 red/green fluorescence was calculated and normalized to the
Cy3 antibody (1:1000 dilution; Abcam, cat. no. 6939) for 2 hours at Con group.
room temperature. Cells were counterstained with 4′, 6′-diamidino-
2-phenylindole-dihydrochloride (DAPI) (1:1000 dilution; Sigma, cat.
no. 9542) for visualizing the nucleus. Cells were mounted on slides 2.9 | Animals and experimental treatments
using 90% glycerol in PBS as mounting medium, and images were ob-
tained using LSCM (Olympus FV3000, Japan). To analyze the locali- The experimental protocol was approved by Xi'an Jiaotong University
zation of each interesting protein in cells, different images obtained according to the guidelines of the Care and Use of Laboratory
from the same area were merged using EZ-C1 software (EZ-C1; Animals of the Chinese Council on Animal Research and Care. The
Nikon) and measured using NIS-Elements AR 3.2 software (Nikon animal studies were conducted strictly following the University of
Instruments). Fluorescence intensity of pNrf2 was analyzed using Xi'an Jiaotong University Guidelines for Animals in Research.
ImageJ software. Seventy Sprague Dawley (male, 6-8  weeks old, 180-220  g)
rats were obtained from the animal research center, Xi'an Jiaotong
University. Rats were kept in a temperature-controlled (24°C) and
2.7 | Protein expression of Nrf2 humidity-controlled (50%) room on a 12 hours light/dark cycle in a
specific pathogen-free facility with free access to food and water.
Protein expression of Nrf2 and pNrf2 was analyzed by western blot Rats were fed with high-sugar and high-fat diet for 1 month before
as described previously. 24 Briefly, protein extracts were performed grouping. The rats were divided randomly into the following groups
using the protein extraction kit (Jiancheng Bioengineering Institute) (N = 10 per group):
according to the manufacturer's instruction. The total concentration
of protein in each sample was determined by the BCA protein assay Con group: normal control.
kit (Thermo Scientific). Equal amounts of protein (25 μg) were sepa- PD group: rats were induced using a ligature and P gingivalis
rated on 10% SDS-PAGE (w/v) Bis-Tris gels, transferred to polyvi- 33 277 infection at the maxillary second molars as previously
nylidene fluoride membranes, and then incubated overnight at 4°C described. 26
ZHU et al. |
      385

DM group: rats were intraperitoneally injected with 35  mg/kg Sections were then incubated with streptavidin biotin conjugated
streptozotocin (STZ; dissolved in 1% sodium citrate buffer, anti-rabbit IgG. Diaminobenzidine chromogenic reagent kit (Boster,
pH  =  4.5; Sigma-Aldrich) to induce type 2 DM. Three days Wuhan, China) was used to visualize the specific staining, following
following STZ administration, rats with fasting blood glucose by counterstaining with hematoxylin. The images were captured by
concentrations ≥16.7 mmol/L and decreased insulin sensitivity microscope (Olympus BX51, Japan) with × 400 magnification. Five
27
were selected as the STZ-induced DM group. slides per sample were analyzed, and the percentage of pNrf2-posi-
CPDM group: 1 month after confirmation of diabetes, periodon- tive cells was calculated.
titis was induced for another 1 month.
CPDM treatment groups: rats were administered with BCI di-
luted in PBS by oral gavage for 1 month at three dose groups 2.14 | Statistical analysis
(50, 100, or 200 mg/kg/d).
All experiments were performed in triplicate and repeated at least
three times. Data were presented as the means  ±  standard devia-
2.10 | Alveolar bone absorption tions (SD). One-way analysis of variance followed by Dunnett's tests
was performed to determine the significant differences between
All rats were sacrificed after treatments via cervical dislocation groups by SPSS 16.0. Statistically significant differences were con-
under ketamine (100  mg/kg) and xylazine (20  mg/kg) anesthesia. sidered at P < .05.
The alveolar bone absorption was examined by Quantum GX micro-
computed tomography (micro-CT) (PerkinElmer). The changes of al-
veolar bone height at the maxillary second molars were assessed via 3 | R E S U LT S
measuring the distance of the cemento-enamel junction (CEJ) to the
alveolar bone crest (ABC). The CEJ-ABC distance along the long axis 3.1 | HGECs were challenged with P gingivalis LPS
of the buccal roots of each maxillary second molar was measured in the context of high glucose to establish a stable
and presented in millimeters (mm). model of CPDM in vitro

A stable model of CPDM in vitro has been widely used by chal-


2.11 | Fasting blood glucose lenging the periodontal cells with P gingivalis LPS in high-glucose
environments. 28,29 MTT results showed that 25 mmol/L glucose sig-
The fasting blood glucose level of Sprague Dawley rats was moni- nificantly reduced hGECs cell viability, when compared to the con-
tored three days after the rats were intraperitoneally injected with trol group of hGECs in 5.5 mmol/L glucose (Figure S1A). In addition,
35 mg/kg STZ. The fasting blood glucose level of the rats was meas- proliferative activity of hGECs was reduced by 10 and 20 µg/mL LPS
ured and recorded by a blood glucose meter (ACCU-CHEK, Roche challenges, respectively (Figure S1B). Then, we verified the valid-
Company) using tail tip blood sampling after fasting 12 hours. ity of the model by detecting the pro-inflammatory mediators (Tnf-
α, Il-1β, Il-6, and Il-8) that were typically expressed in periodontitis
with DM. HGECs treated with 25 mmol/L glucose + 20 µg/mL LPS
2.12 | Insulin sensitivity index significantly up-regulated the gene expression of pro-inflammatory
cytokines, including Tnf-α, Il-1β, Il-6, and Il-8 (Figure S1C). According
The rat insulin (INS) ELISA kit (Mercodia) was used to detect the to the results of MTT and real-time PCR, cells were divided into the
serum insulin content of the rats. The assay process was performed following groups: (a) Con group, cells were incubated in 5.5 mmol/L
according to the kit's instructions. The absorbance (OD value) at glucose; (b) HG group, cells were incubated in 25 mmol/L glucose;
450 nm was measured by a microplate reader to determine the in- (c) LPS group, cells were incubated in 5.5 mmol/L glucose + 20 µg/
sulin concentration. The insulin sensitivity index was calculated ac- mL LPS; (d) GP group, cells were incubated in 25  mmol/L glu-
cording to the formula: ISI = 22.5/(FBG × INS). cose  +  20  µg/mL LPS. These four groups simulated the state of
health, periodontitis, DM, and periodontitis with DM in human at
the cellular level.
2.13 | Immunohistochemical analysis of pNrf2

Animals were sacrificed via cervical dislocation after euthanized with 3.2 | GP stimulated the ROS production,
ketamine (100 mg/kg) and xylazine (20 mg/kg). One-side maxillary suppressed the gene expression level of antioxidant
specimens were harvested and immediately fixed in 4% (w/v) para- enzymes and phosphorylation of Nrf2 in hGECs
formaldehyde for at least two days, and sectioned at 5 μm as previ-
ously described. 26 The sections were then stained with anti-pNrf2 Reactive oxygen species production is augmented in response
primary antibody (1:50; Abcam, cat. no. 76  026) at 4℃ overnight. to inflammatory mediators. 30 Furthermore, oxidative stress, as
|
386       ZHU et al.

F I G U R E 1   Intracellular ROS, Nrf2 gene and its downstream antioxidant genes, pNrf2 translocation and protein levels in physiological
and high concentration of glucose to hGECs with or without LPS. A, Intracellular ROS levels were indicated using DCFH-DA fluorescence
by FACS and the DCF fluorescence intensity was statistical analyzed. B, The mRNA levels of five antioxidant genes as indicated were
determined by real-time PCR and statistically analyzed. C, The translocation of pNrf2 (S40) was determined by LSCM (representative images
and statistical analysis). The nuclei was stained with DAPI. Scale bar = 30 μm. D, The protein levels of pNrf2 (S40) and total Nrf2 (t-Nrf2)
were determined by western blot analysis (representative images and statistical analysis). Data from representative results were expressed
as mean ± SD from a minimum of three replicates per experiment. *P < .05 represents significant differences compared to the Con group;
#
P < .05 represents significant differences compared to the GP group

a result of elevated intracellular ROS, has been proved as an im- pNrf2 (Figure 1D). Taken together, the results from Figure 1 indi-
portant contributor to CPDM. 5,6 FACS analysis revealed a shift of cated that Nrf2-mediated signaling pathway might be compromised
2',7'-dichlorofluorescein (DCF)-specific fluorescence signal after by stimulation of GP in hGECs.
HG, LPS, and GP treatment. Moreover, DCF-specific fluorescence
signal was significantly elevated in the GP group compared with
DCF-specific fluorescence signal in HG or LPS group (Figure 1A). 3.3 | BCI prevented hGECs from oxidative damage
As a response to excess intracellular ROS, cells normally have caused by GP via the Nrf2 signaling pathway
several protective mechanisms to scavenge ROS, such as the activa-
tion of antioxidant enzymes.31 In the present study, we found that Cells were incubated with various concentrations of the compound
the mRNA levels of some representative anti-antioxidant-related (0.01, 0.1, 0.5, 1, and 10 µg/mL) for 24 hours in order to investigate
genes (Cat, Gclc, Sod1, Sod2, and Nrf2) were significantly decreased the effect of BCI on the viability of hGECs. Treatment with BCI at
after hGECs were treated with GP (Figure 1B). It indicated that the concentrations of 0.01, 0.1, and 0.5  µg/mL increased cell viability
antioxidant systems were compromised when the hGECs were ex- of the hGECs in a dose-dependent manner (Figure S2A), while 1 and
posed to GP environment. 10 µg/mL BCI inhibited the cell viability compared with the control.
The antioxidant enzymes mentioned above were regulated by Accordingly, BCI at concentrations of 0.01, 0.1, and 0.5 µg/mL was
the nuclear transcription factor Nrf2. Immunofluorescence analysis selected for subsequent experiments. And the MTT results showed
showed that the fluorescent intensities in the nucleus of pNrf2 were that 0.01 to 0.5 µg/mL BCI could promote the viability of hGECs in
decreased in groups challenged with HG, LPS, or GP. Furthermore, BCI + GP groups (Figure S2B).
the relative fluorescence intensities of pNrf2 in the GP-treated The anti-antioxidant activity of BCI against GP-induced oxidative
groups were weaker compared with the fluorescent intensities of damage was further examined. FACS analysis indicated that pre-treat-
pNrf2 in the HG group and LPS group (Figure 1C), and this result was ment with BCI could significantly suppress the excess ROS production
further confirmed by western blot using a specific antibody against induced by GP (Figure 2A). Moreover, GP caused a loss of MMP as
ZHU et al. |
      387

F I G U R E 2   Effects of BCI on ROS production and MMP in high concentration of glucose to hGECs with LPS. A, Intracellular ROS levels
were indicated using the DCF fluorescence intensity (representative images and statistical analysis). B, MMP was measured by LSCM
analysis of JC-1 staining (representative images and statistical analysis). Scale bar = 50 μm. Data from representative results were shown
as mean ± SD from a minimum of three replicates per experiment. *P < .05 represents significant differences compared to the Con group;
#
P < .05 represents significant differences compared to the GP group

showed by the loss of the red signal and the increase of green signal. than that in the Con group (Figure S3C). DM groups exhibited hyper-
However, pre-treatment with BCI reversed the MMP loss (Figure 2B). glycemia with higher fasting blood glucose levels (Figure S3D) and
We further examined the production of antioxidant enzymes of significantly decreased insulin sensitivity index (Figure S3E) than the
BCI-mediated cytoprotection. Cells that pre-treated with 0.01, 0.1, non-DM groups. These results indicated that a DM model could be
and 0.5  µg/mL BCI in BCI  +  GP groups significantly increased the induced in rats by treating the animals with a high-fat and high-sugar
mRNA levels of Nrf2, Sod1, Sod2, Cat and Gclc as compared with the diet before intraperitoneally injection of 35 mg/kg STZ. Thread liga-
GP group without BCI treatment (Figure 3A). Moreover, pNrf2 was tion combined with P gingivalis ATCC 33  277 local injection at the
augmented in the BCI  +  GP treatment groups than the GP group, maxillary second molars could induce a periodontitis model. Rats un-
as showed by increased fluorescent intensities of pNrf2 detected derwent these two combined methods exhibited a CPDM phenotype.
by LSCM (Figure 3B). The western blot analysis also showed an in-
creased ratio of pNrf2/total Nrf2 expression in BCI + GP treatment
groups (Figure 3C). 3.5 | BCI treatment had no effect on fasting blood
glucose and insulin sensitivity index in CPDM in vivo

3.4 | Characterizations of the stable CPDM model Our previous study demonstrated that 50, 100, and 200 mg/mL BCI
in vivo significantly inhibited the absorption of alveolar bone in rats with no
side effects. 26 Accordingly, rats in the present study were adminis-
The stable CPDM model in Sprague Dawley rats used in the present trated with BCI at the same doses. Individual diabetes effected the
study was established and illustrated in Figure S3A,B. All animals development of periodontitis.32 In order to examine whether BCI af-
enrolled in the study were maintained in good systemic health dur- fects the diabetic level, we tested the fasting blood glucose and insu-
ing the observation period. The lengths of CEJ-ABC at the maxillary lin sensitivity index. The results showed that there was no significant
second molars in the PD and CPDM groups were significantly greater difference after BCI treatment in CPDM in vivo (Figure S4).
|
388       ZHU et al.

F I G U R E 3   Effects of BCI on the expression of antioxidant enzymes, nucleus accumulation and protein expression of pNrf2 in high
concentration of glucose to hGECs with LPS. A, The mRNA levels of five antioxidant genes as indicated were determined by real-time
PCR and statistically analyzed. B, The translocation of pNrf2 (S40) was determined by LSCM (representative images and statistical
analysis). Nuclei were stained with DAPI. Scale bar = 25 μm. C, The protein level of pNrf2 (S40) and t-Nrf2 was determined by western
blot (representative images and statistical analysis). Data from representative results were shown as mean ± SD from a minimum of three
replicates per experiment. *P < .05 represents significant differences compared to the Con group; #P < .05 represents significant differences
compared to the GP group

3.6 | BCI increased pNrf2 expression and damage in the state of oxidative stress which has been also recognized
mitigated the alveolar bone loss in rats as an important contributor to periodontal destruction in CPDM.6
Studies reported that the systemic level of ROS in serum of patients
Immunohistochemical analysis revealed that treatment with 50, 100, with CPDM was excessively increased.5 Previous results further im-
and 200 mg/kg BCI markedly increased the number of pNrf2-posi- plied a disturbance in antioxidant defence system that the activities
tive cells compared with CPDM group (Figure 4). We further inves- of SOD and CAT were reduced in saliva.35 This may be attributable
tigated the effect of BCI on the prevention of alveolar bone loss by to excessive ROS production from glucose and glycated antioxidative
micro-CT (Figure 5). The results showed that administering rats with enzymes which destroy their capacity to scavenge ROS. The results
BCI (50, 100, 200  mg/kg body weight) dramatically mitigated the of the present study showed that ROS was elevated, mitochondria
CPDM-mediated alveolar bone loss. membrane potential was reduced, pNrf2 and Nrf2-related antioxidant
genes were decreased after hGECs were stimulated with 25 mmol/L
glucose plus 20 µg/mL LPS. Accordingly, the model that hGECs stim-
4 |  D I S CU S S I O N ulated with 25  mmol/L glucose plus 20  µg/mL LPS can be used for
evaluating the level of oxidative stress for CPDM in vitro.
It is well known that ROS in redox biology state act as signaling mol- Periodontitis is caused by the host response to various bac-
ecules to the maintenance of the cells’ normal actions. The balance terial infections.1 The gingival epithelium serves as the primary
between redox physiological and biochemical homeostasis is precisely physical and chemical barrier affecting the immune system and
regulated by antioxidant system including a series of enzymes that can responding to the foreign bacteria. 36 Thus, gingival epithelial
scavenge ROS and dietary-derived molecules, for example Nrf2, Cat, cells play a pivotal role in the maintenance of periodontal health.
Gclc, Sod1, and Sod2 33,34 However, the excessive ROS leads to tissue However, it has been documented that inflammatory conditions
ZHU et al. |
      389

F I G U R E 4   Effects of BCI on pNrf2 (S40) expression in periodontal tissue in rats with CPDM. PNrf2-positive cells were detected by
immunohistochemical analysis (representative images and statistical analysis). Data from representative results were shown as mean ± SD
from a minimum of three replicates per experiment. *P < .05 represents significant differences compared to the CPDM group

such as periodontitis could reduce gingival epithelium functions groups. Epidemiological evidence has demonstrated a bidirectional
associated with periodontal homeostasis. 37 Thus, it is important to relationship between periodontitis and DM.41 In our study, BCI ad-
enhance the proliferation of gingival epithelium cells of the peri- ministration for one month inhibited the absorption of alveolar bone
odontium. Li et al suggested that the cell proliferation activity in with no effect on blood glucose. However, the long-term therapeutic
primary hGECs was significantly decreased at the concentration effect of BCI on blood glucose requires further observation.
of BCI > 4.7 μg/mL. 38 In the present work, more than 1 µg/mL BCI Nrf2 has been recognized as a possible drug target for enhancing
decreased the proliferation of hGECs. These results indicated that antioxidant production of the host.31 Several antioxidant compounds
the treatment with high concentrations of BCI was not conducive such as resveratrol, paeonol, and isorhamnetin were reported to be
to cell proliferation. Interestingly, the BCI concentration used in able to promote the expression of Nrf2.42-44 A recent study has
this study was lower than Li's report, this probably because the described an increase in the oxidative damage indicators and a de-
hGECs cell line used in the present study was more sensitive to crease of total Nrf2 in an experiment CPDM model.45 However, the
BCI stimulation than the primary hGECs. phosphorylation form of Nrf2 at serine40 has been confirmed to
The association of periodontitis and DM has been well docu- play a key mechanistic role in regulating the resistance to oxidative
mented recently. Based on the latest classification standard 2018, stress.16 Thus, under the oxidative stress condition, the phosphor-
DM was believed and defined as an important descriptor in the stag- ylation form of Nrf2 should be examined instead of the total level
ing and grading process of periodontitis.39 DM shifts the immune of Nrf2. Posttranslational regulation of Nrf2 is known to be mod-
defence outside their normal boundary responses to bacterial chal- ulated by different kinases that phosphorylate Nrf2 and affect its
lenges, which can negatively affect periodontal tissues, resulting in activation. Previous study showed that selective inhibitors of phos-
the severity and prognosis of periodontitis. In our present study, phoinositol-3-kinase/AKT could block the cytoprotective effect of
diabetic rats with periodontitis exhibited a more severe periodon- BCI.46 In our study, immunofluorescence technique and western
titis status. It has been demonstrated that glycemic control of indi- blot were used to evaluate the activation of pNrf2 and its related
viduals affects the progress of periodontitis.40 Thus, we detected downstream genes; however, further investigations are needed to
the fasting blood glucose level and insulin sensitivity index after BCI clarify the detailed molecular mechanism on Nrf2 activation.
administration. The results showed that there were no significant A relationship between periodontitis and cytoprotective en-
differences for these two indexes between CPDM and CPDM + BCI zymes themselves has also been demonstrated. 31 Genetic mutation

F I G U R E 5   Effects of BCI on alveolar


bone loss in rats with CPDM. The distance
of CEJ to ABC was measured by micro-
CT (representative images and statistical
analysis). Data from representative results
were shown as mean ± SD from a
minimum of three replicates per
experiment. *P < .05 represents significant
differences compared to the CPDM group
|
390       ZHU et al.

of SOD was identified as a risk factor for periodontitis.47 In addi- National Natural Science Foundation of China (Grant No. 81901019
tion, both Nrf2 and sirtuin 1 have been reported to play important and 81870798), and from the Fundamental Research Funds for the
roles in regulating SOD. 31 Therefore, not only Nrf2-dependent an- Central Universities (Grant No. xjj2017025 and xjj2017097).
tioxidant axis but also sirtuin 1 mediated cytoprotective mecha-
nisms may play roles in control periodontal homeostasis. Further C O N FL I C T O F I N T E R E S T
study is needed to investigate the regulatory role of other antiox- The authors declare no conflicts of interest with respect to the au-
idant signaling pathways and their relationship with CPDM. Apart thorship and/or publication of this article.
from the role in the antioxidant system, Nrf2 has been reported
to participate in inflammatory responses via activation of its tar- ORCID
31
get genes. The present study revealed that exposure of hGECs Dandan Pei  https://orcid.org/0000-0003-4470-5400
to GP up-regulated the pro-inflammatory factors and at the same Ang Li  https://orcid.org/0000-0001-6720-0785
time reduced Nrf2 expression. It was reported that Nrf2 activa-
tion could inhibit the oxidative stress driven by the nuclear factor REFERENCES
kappa beta-mediated inflammatory response via heme oxygenase 1. Chapple ILC, Van der Weijden F, Doerfer C, et al. Primary pre-
1 end-products.48 In contrast, the nuclear factor kappa beta p65 vention of periodontitis: managing gingivitis. J Clin Periodontol.
2015;42(Suppl 16):S71-76.
subunit also repressed the Nrf2/ARE pathway at the transcriptional
2. Costa KL, Taboza ZA, Angelino GB, et al. Influence of periodontal
level via deprivation of cAMP response element binding protein disease on changes of glycated hemoglobin levels in patients with
from Nrf2.49 Activation of Nrf2 by BCI promoted the antioxidant type 2 diabetes mellitus: a retrospective cohort study. J Periodontol.
ability of the hGECs and subsequently down-regulated the inflam- 2017;88:17-25.
3. Stanko P, Izakovicova HL. Bidirectional association between di-
matory responses to protect alveolar bone, as shown by mitigation
abetes mellitus and inflammatory periodontal disease. A review.
of CPDM-induced alveolar bone resorption in rats. Biomed Pap. 2014;158:35-38.
Oral administration of BCI is safe and well tolerated. The com- 4. Gurav AN. Management of diabolical diabetes mellitus and
pound exhibited favorable pharmacokinetics (100-2800 mg daily).50 periodontitis nexus: are we doing enough? World J Diabetes.
2016;7:50-66.
In our study, rats were orally gavaged with BCI at three doses (50,
5. Patil VS, Patil VP, Gokhale N, Acharya A, Kangokar P. Chronic peri-
100, and 200  mg/mL), and no side effects of BCI were observed odontitis in type 2 diabetes mellitus: oxidative stress as a common
during the study. In addition, BCI significantly up-regulated the factor in periodontal tissue injury. J Clin Diagn Res. 2016;10:BC12-16.
expression of pNrf2 and inhibited the absorption of alveolar bone 6. Vincent RR, Appukuttan D, Victor DJ, Balasundaram A. Oxidative
in rats with CPDM. However, oral bioavailability of BCI is very low stress in chronic periodontitis patients with type II diabetes melli-
tus. Eur J Dent. 2018;12:225-231.
which may be due to its poor solubility and short half-life.51 The
7. Tamaki N, Hayashida H, Fukui M, et al. Oxidative stress and anti-
clinical applications of BCI must be highly improved if we develop body levels to periodontal bacteria in adults: the Nagasaki Islands
drugs to treat periodontitis via periodontal pocket in the future, and study. Oral Dis. 2014;20:e49-e56.
different delivery ways need to be studied, such as BCI-loaded nano- 8. Luigi N, Nikos D. Periodontitis and redox status: a review. Curr
Pharm Design. 2013;19:2687-2697.
structured lipid carriers.38
9. Tian Y, Li X, Xie H, et al. Protective mechanism of the antioxidant
baicalein toward hydroxyl radical-treated bone marrow-derived
mesenchymal stem cells. Molecules. 2018;23:223.
5 |  CO N C LU S I O N 10. Patwardhan RS, Sharma D, Thoh M, Checker R, Sandur SK. Baicalein
exhibits anti-inflammatory effects via inhibition of NF-κB transacti-
vation. Biochem Pharmacol. 2016;108:75-89.
In the present study, we examined the mechanisms by which BCI 11. Tsou LK, Lara-Tejero M, RoseFigura J, et al. Antibacterial flavonoids
prevented oxidative stress via Nrf2 signaling pathway in two experi- from medicinal plants covalently inactivate type iii protein secre-
mental CPDM models in vitro and in vivo. This study demonstrated tion substrates. J Am Chem Soc. 2016;138:2209-2218.
12. Shen YC, Chiou WF, Chou YC, Chen CF. Mechanisms in mediating
that BCI significantly reduced ROS formation, increased the expres-
the anti-inflammatory effects of baicalin and baicalein in human
sion of Nrf2 and its downstream genes (Cat, Gclc, Sod1, and Sod2). leukocytes. Eur J Pharmacol. 2003;465:171-181.
Furthermore, rats administrated with BCI markedly increased the 13. Jiang M, Li Z, Zhu G. Protective role of flavonoid baicalin from
number of pNrf2-positive cells and mitigated the CPDM-mediated Scutellaria baicalensis in periodontal disease pathogenesis: a litera-
ture review. Complement Ther Med. 2018;38:11-18.
alveolar bone loss. Our findings suggested that BCI promoted peri-
14. Liu J, Wang S, Sun J, et al. Screening of osteoanagenesis-active
odontal health by stimulating Nrf2-mediated antioxidant defence compounds from scutellaria baicalensis georgi by hPDLC/CMC–on-
systems. On the basis of the present studies, we proposed that BCI line-HPLC/MS. Fitoterapia. 2014;93:105-114.
might be developed as a novel therapeutic drug for the clinical treat- 15. Kraft AD, Lee JM, Johnson DA, Kan YW, Johnson JA. Neuronal
ment of periodontitis with DM. sensitivity to kainic acid is dependent on the Nrf2-mediated ac-
tions of the antioxidant response element. J Neurochem. 2006;98:
1852-1865.
AC K N OW L E D G E M E N T S 16. D'Autréaux B, Toledano MB. ROS as signalling molecules: mecha-
We thank Drs Jianmin Yang and Zhongbo Liu for carefully read- nisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell
ing the manuscript. This work was supported by a grant from the Bio. 2007;8:813-824.
ZHU et al. |
      391

17. Ma J, Li S, Zhu L, et al. Baicalein protects human vitiligo melanocytes 37. Huang X, Yang XI, Ni J, et al. Hyperglucose contributes to periodon-
from oxidative stress through activation of NF-E2-related factor2 titis: involvement of the NLRP3 pathway by engaging the innate im-
(Nrf2) signaling pathway. Free Radical Bio Med. 2018;129:492-503. munity of oral gingival epithelium. J Periodontol. 2015;86:327-335.
18. Choi E-O, Jeong J-W, Park C, et al. Baicalein protects C6 glial cells 38. Li X, Luo W, Ng TW, et al. Nanoparticle-encapsulated baicalein
against hydrogen peroxide-induced oxidative stress and apoptosis markedly modulates pro-inflammatory response in gingival epithe-
through regulation of the Nrf2 signaling pathway. Int J Mol Med. lial cells. Nanoscale. 2017;9:12897-12907.
2016;37:798-806. 39. Tonetti MS, Greenwell H, Kornman KS. Staging and grading of peri-
19. Cui G, Chui Wah Luk S, Li RA, et al. Cytoprotection of baicalein odontitis: framework and proposal of a new classification and case
against oxidative stress-induced cardiomyocytes injury through the definition. J Periodontol. 2018;89:S159-S172.
Nrf2/Keap1 pathway. J Cardiovasc Pharm. 2015;65:39-46. 4 0. Teshome A, Yitayeh A. The effect of periodontal therapy on gly-
20. Zhang Y, Cui L, Guan G, et al. Matrine suppresses cardiac fibrosis by cemic control and fasting plasma glucose level in type 2 diabetic
inhibiting the TGF-β/Smad pathway in experimental diabetic car- patients: systematic review and meta-analysis. Bmc Oral Health.
diomyopathy. Mol Med Rep. 2018;17:1775-1781. 2017;17:31.
21. Ren Y, Li Y, Yan J, et al. Adiponectin modulates oxidative stress-in- 41. Khazaii R, Kamareh S. Relationship between diabetes and periodon-
duced mitophagy and protects C2C12 myoblasts against apoptosis. tal disease: a review of literature. Ann Dent Spec. 2018;6:57-60.
Sci Rep. 2017;7:3209. 42. Tamaki N, Orihuela-Campos RC, Inagaki Y, Fukui M, Nagata T, Ito
22. Yerra VG, Kalvala AK, Sherkhane B, Areti A, Kumar A. Adenosine HO. Resveratrol improves oxidative stress and prevents the pro-
monophosphate-activated protein kinase modulation by berberine gression of periodontitis via the activation of the Sirtl/AMPK and
attenuates mitochondrial deficits and redox imbalance in experimen- the Nrf2/antioxidant defense pathways in a rat periodontitis model.
tal diabetic neuropathy. Neuropharmacology. 2018;131:256-270. Free Radical Bio Med. 2014;75:222-229.
23. Zhou H, Yue Y, Wang J, Ma Q, Chen Y. Melatonin therapy for di- 43. Li J, Li YP, Pan S, Zhang L, He L, Niu Y. Paeonol attenuates liga-
abetic cardiomyopathy: a mechanism involving Syk-mitochondrial tion-induced periodontitis in rats by inhibiting osteoclastogen-
complex I-SERCA pathway. Cell Signal. 2018;47:88-100. esis via regulating Nrf2/NF-kappaB/NFATc1 signaling pathway.
24. Wang D, Sun H, Song G, et al. Resveratrol improves muscle atrophy Biochimie. 2019;156:129-137.
by modulating mitochondrial quality control in STZ-induced dia- 4 4. Qi F, Sun JH, Yan JQ, Li CM, Lv XC. Anti-inflammatory effects of isor-
betic mice. Mol Nutr Food Res. 2018;62:e1700941. hamnetin on LPS-stimulated human gingival fibroblasts by activat-
25. Sun L, Zhao M, Yang Y, et al. Acetylcholine attenuates hypoxia/re- ing Nrf2 signaling pathway. Microb Pathogenesis. 2018;120:37-41.
oxygenation injury by inducing mitophagy through PINK1/Parkin 45. Bi C-S, Wang J, Qu H-L, et al. Calcitriol suppresses lipopolysaccha-
signal pathway in H9c2 cells. J Cell Physiol. 2016;231:1171-1181. ride-induced alveolar bone damage in rats by regulating T helper
26. Sun J-Y, Li D-L, Dong Y, et al. Baicalin inhibits toll-like receptor 2/4 cell subset polarization. J Periodontal Res. 2019;1–12.
expression and downstream signaling in rat experimental periodon- 46. Zhang Z, Cui W, Li G, et al. Baicalein protects against 6-OHDA-
titis. Int Immunopharmacol. 2016;36:86-93. induced neurotoxicity through activation of Keap1/Nrf2/HO-1 and
27. Li X, Sun X, Zhang X, et al. Enhanced oxidative damage and Nrf2 involving PKCα and PI3K/AKT signaling pathways. J Agr Food Chem.
downregulation contribute to the aggravation of periodontitis by 2012;60:8171-8182.
diabetes mellitus. Oxid Med Cell Longev. 2018;2018:9421019. 47. Kazemi E, Moradi MT, Yari K, Mousavi SAR, Kahrizi D. Association
28. Yang XI, Zhang J, Ni J, et al. Toll-like receptor 4-mediated hyper-re- between manganese superoxide dismutase (MnSOD Val-9Ala) gen-
sponsiveness of gingival epithelial cells to lipopolysaccharide in otypes with the risk of generalized aggressive periodontitis disease.
high-glucose environments. J Periodontol. 2014;85:1620-1628. Cell Mol Biol. 2015;61:49-52.
29. Chiu HC, Fu M, Yang T, et al. Effect of high glucose, porphyromonas 48. Morgan MJ, Liu Z. Crosstalk of reactive oxygen species and NF-
gingivalis lipopolysaccharide and advanced glycation end-prod- kappa B signaling. Cell Res. 2011;21:103-115.
ucts on production of interleukin-6/-8 by gingival fibroblasts. J 49. Liu GH, Qu J, Shen X. NF-kappa B/p65 antagonizes Nrf2-ARE path-
Periodontal Res. 2017;52:268-276. way by depriving CBP from Nrf2 and facilitating recruitment of
3 0. Lee IT, Yang C. Role of NADPH oxidase/ROS in pro-inflamma- HDAC3 to MafK. Biochim Biophys Acta. 2008;1783:713-727.
tory mediators-induced airway and pulmonary diseases. Biochem 50. Lai M-Y, Hsiu S-L, Tsai S-Y, Hou Y-C, Chao P-D. Comparison of met-
Pharmacol. 2012;84:581-590. abolic pharmacokinetics of baicalin and baicalein in rats. J Pharm
31. Kanzaki H, Wada S, Narimiya T, et al. Pathways that regulate ROS Pharmacol. 2003;55:205-209.
scavenging enzymes, and their role in defense against tissue de- 51. Zhang J, Lv H, Jiang K, Gao Y. Enhanced bioavailability after oral
struction in periodontitis. Front Physiol. 2017;8:351. and pulmonary administration of baicalein nanocrystal. Int J
32. Caton JG, Armitage G, Berglundh T, et al. A new classification Pharmaceut. 2011;420:180-188.
scheme for periodontal and peri-implant diseases and conditions
- introduction and key changes from the 1999 classification. J
Periodontol. 2018;89:S1-S8. S U P P O R T I N G I N FO R M AT I O N
33. Kakkar R, Kalra J, Mantha SV, Prasad K. Lipid peroxidation and Additional supporting information may be found online in the
activity of antioxidant enzymes in diabetic rats. Mol Cell Biochem.
Supporting Information section. 
1995;151:113-119.
3 4. Netto LES, Antunes F. The roles of peroxiredoxin and thioredoxin
in hydrogen peroxide sensing and in signal transduction. Mol Cells.
2016;39:65-71. How to cite this article: Zhu C, Zhao Y, Wu X, et al. The
35. Trivedi S, Lal N, Mahdi AA, Mittal M, Singh B, Pandey S. Evaluation therapeutic role of baicalein in combating experimental
of antioxidant enzymes activity and malondialdehyde levels in pa- periodontitis with diabetes via Nrf2 antioxidant signaling
tients with chronic periodontitis and diabetes mellitus. J Periodontol.
pathway. J Periodont Res. 2020;55:381–391. https​://doi.
2014;85:713-720.
36. Takuji N, Hideki S, Hitoshi K, et al. Syntheses of prostaglandin E2 org/10.1111/jre.12722​
and E-cadherin and gene expression of beta-defensin-2 by human
gingival epithelial cells in response to Actinobacillus actinomycetem-
comitans. Inflammation. 2003;27:341-349.

You might also like