Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Carbohydrate Polymers 237 (2020) 116163

Contents lists available at ScienceDirect

Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol

The effect of doxycycline-containing chitosan/carboxymethyl chitosan T


nanoparticles on NLRP3 inflammasome in periodontal disease
Shuo Xua,b, Qihui Zhouc, Zhongxin Jiangd, Yanwen Wange, Kai Yanga,b, Xiaohui Qiua,b,
Qiuxia Jia,*
a
Department of Periodontology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
b
School of Stomatology of Qingdao University, Qingdao, 266003, China
c
Institute for Translational Medicine, State Key Laboratory of Bio-fibers and Eco-textiles, Qingdao University, Qingdao, 266021, China
d
Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
e
Stuart Country Day School of the Sacred Heart, 1200 Stuart Road, Princeton, New Jersey, 08628, United States

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

Keywords: A polyelectrolyte complex nanoparticle comprising chitosan (CS) and carboxymethyl chitosan (CMCS) was
Chitosan prepared (CS/CMCS-NPs) by ionic gelation, which was then used as a doxycycline carrier (Dox:CS/CMCS-NPs).
Carboxymethyl chitosan The obtained CS/CMCS-NPs and Dox:CS/CMCS-NPs were characterized for various parameters and bacterio-
Doxycycline static ability against Porphyromonas gingivalis. The regulation of related genes and proteins of NLRP3 in-
NLRP3 inflammasome
flammasome and IL-1β in human gingival fibroblasts (HGFs) was characterized by qRT-PCR, western blotting
Periodontitis
and ELISA. The results showed that Dox:CS/CMCS-NPs had an orderly morphology and an excellent cyto-
compatibility. P. gingivalis was strongly inhibited by Dox:CS/CMCS-NPs contrasted with control group. Dox:CS/
CMCS-NPs effectively down-regulated both gene and protein levels of NLRP3 inflammasome and IL-1β in HGFs.
This study provides a new method for rational application of Dox in the clinical treatment of periodontal disease
and a new direction for explaining the mechanism of action of Dox:CS/CMCS-NPs and more drug-carrying
nanoparticles.

1. Introduction through a series of signal transduction pathways and activate acquired


immunity (Schenten & Medzhitov, 2011). NOD-like receptors (NLRs)
Periodontal disease is an infectious bacterial disease occurring in are a class of intracytoplasmic PRRs, with an activation of multiple
periodontal supporting tissues (Sanz et al., 2017). It is a common and protein complexes, or inflammasomes typical structure, after ligand
frequently occurring disease that can lead to continuous irreversible recognition. NLRP3 inflammasome is the most intensively-studied in-
destruction of periodontal supporting tissues, which will eventually flammasome complex, comprising NLRP3, ASC and Caspase-1. After
lead to the loss of teeth and seriously affect quality of life(Kassebaum activation, NLRP3 can interact with the adaptor protein ASC to recruit
et al., 2014). Dental plaque is the initiating factor of periodontitis. Host Caspase-1 and lead to the autocatalysis of Caspase-1, which splits into
responses caused by periodontal pathogens in the subgingival area play active p20 and p10 fragments and shears pro-IL-1β and pro-IL-18. This
a crucial role in the destruction of connective tissue and bone (Graves, generates mature cytokines IL-1β and IL-18, which are released to the
2008). Immune response is closely related to the occurrence and de- outside of the cell and play a role in immunity and inflammation
velopment of periodontal disease (Chatzopoulos, Mansky, Lunos, (Hansen, Vojtech, & Laing, 2011). The main pathogenic bacteria of
Costalonga, & Wolff, 2019; Jing et al., 2019; Zhang & Deng, 2019). periodontal disease include Porphyromonas gingivalis, Tannerella for-
NLRP3 inflammasome plays an important role in regulating innate sythia and Prevotella intermedia (Hajishengallis, 2015). Porphyromonas
immune responses in chronic inflammatory diseases such as period- gingivalis (P. gingivalis), a gram-negative obligate anaerobic bacterium,
ontitis (Olsen & Yilmaz, 2016).Pattern recognition receptors (PRRs) are is the dominant bacteria in periodontal disease, especially in a lesion
an important component of the innate immune system that can re- area or active site of chronic periodontitis. It is closely associated with
cognize pathogen-associated molecular patterns (PAMPs) and danger- NLRP3 inflammasome activation. Inflammasomes recognize the fla-
associated molecular patterns (DAMPs), release inflammatory factors gellin and secreting system components of pathogens by the Naip


Corresponding author.
E-mail address: jqx_1@163.com (Q. Ji).

https://doi.org/10.1016/j.carbpol.2020.116163
Received 10 July 2019; Received in revised form 10 March 2020; Accepted 11 March 2020
Available online 12 March 2020
0144-8617/ © 2020 Elsevier Ltd. All rights reserved.
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

subfamily proteins in the nod-like receptor family (Hajishengallis et al., obtained by MedChemExpress (Monmouth Junction, USA). Human IL-
2011; Huck, Elkaim, Davideau, & Tenenbaum, 2015; Lian et al., 2018). 1β ELISA Kit was purchased by Elabscience (Wuhan, China). RNAiso
Therefore, NLRP3 inflammasome inhibition is a new strategy for Plus, PrimeScript™ RT reagent Kit with gDNA Eraser and TB Green™
treating periodontitis. Premix Ex Taq™ II were provided by Takara (Dalian, China). PCR
Periodontal scaling and periodontal curettage are the main clinical Primers were supplied by Sangon Biotech (Wuhan, China). All other
treatment methods for periodontitis. Recently, local drug delivery sys- reagents were analytical grade and were used without further pur-
tems have gained increasing attention owing to their better perfor- ification. The experiment was approved by the Ethics Committee of The
mance than systemic administration (Jain et al., 2008). Chitosan (CS), Affiliated Hospital of Qingdao University approval no. QYFY WZLL
one of the natural cationic polysaccharide, has good histocompatibility, 25589.
biodegradation and non-toxicity (Goncalves, Antunes, & Barbosa, 2012;
Ragelle et al., 2014). Increasing attention has also been paid to the 2.2. Preparation of CS/CMCS-NPs and Dox:CS/CMCS-NPs
regulatory effect of chitosan nanoparticles (NPs) on immunity (Gjoseva
et al., 2018). Carboxymethyl chitosan (CMCS) is a water-soluble chit- NPs were prepared by a simple ion crosslinking method. A Dox
osan derivative with good biocompatibility and tissue compatibility aqueous solution (1 mg/ml, 1 ml) and CS solution (3 mg/ml, 1 ml,
(Gujarathi, Rane, & Patel, 2012). Negatively-charged CMCS can form dissolved by 1 % acetic acid, pH 6.0) were pre-mixed at room tem-
polyelectrolyte complexes with positively-charged CS through electro- perature and constant velocity for 30 min under magnetic agitation
static interaction, which can then be used to encapsulate drugs and (IKA C-MAG HS 4) and different concentrations of CMCS (4 mg/ml, 1
better penetrate the internal mucosal barrier (Feng et al., 2014). These ml) and TPP (5 mg/ml, 100 μl) were stirred with the mixture for 1 h to
CS/CMCS nanoparticles are more stable than pure CS nanoparticles, form nanoparticles at room temperature and constant velocity under
and have better water solubility and better tolerance to acidic en- magnetic agitation. It is worth mentioning that in the experiment, we
vironment, so we speculated that they are more suitable for periodontal selected 0.5, 1, 2, 5 mg/ml Dox solutions for the synthesis of nano-
pocket (Feng et al., 2014). particles, but only 1 mg/ml Dox solution produced the most stable
Doxycycline (Dox) is a semi-synthetic second-generation tetra- nanoparticles with the most regular appearance, so we finally de-
cycline, an effective tetracycline antibiotic for periodontitis (Buduneli termined the concentration of 1 mg/ml Dox in the experiment. The
et al., 2004; Nelson & Levy, 2011; Preshaw, 2018). Dox can inhibit obtained NPs were washed three times with deionized water, collected
periodontal pathogen activity and down-regulate NLRP3 inflammasome by ultrafast centrifugation BECKMAN COULTER at 12000 RPM for 30
(Hu et al., 2018; Zhang et al., 2017). However, the direct application of min, and then freeze-dried LABCONCO at −50 ℃ and vacuum for 48 h.
Dox in local, especially an oral environment, still has some problems,
such as systemic exposure and off-target toxicity, not sufficient local 2.3. Characterization of the prepared CS/CMCS-NPs and Dox:CS/CMCS-
delivery, poor response rate of this disease to systemic Dox (Golub NPs
et al., 2016). Therefore, we need a new drug carrier, preferably with a
certain degree of targeting and slow release, can better play the role of The morphology of CS/CMCS-NPs and Dox:CS/CMCS-NPs was ob-
Dox. The gums are thin and the gingival sulcus is shallow, making it served via scanning electron microscopy (SEM) (VEGA3, TESCAN,
difficult for traditional drug paste or film to fit into the gingival sulcus. China) and transmission electron microscopy (TEM) (Spectra 300,
Previous studies have shown that nanoparticles are much more absor- Thermo Fisher Scientific, USA).
bent than microparticles, so it would be better to build a mobile drug The size analysis and zeta potential of NPs were determined using a
nanocarrier that can go into the gingival sulcus and function particle size analyzer (Microtrac Nanotrac Wave Ⅱ). The obtained
(Gregoriadis, 1978). samples were not filtered and diluted prior to testing.
At present, there is little research on the mechanism of action of To determine Dox loading and encapsulation efficiency, the free
nanoparticles in reducing inflammation. For periodontal disease, the Dox in the supernatants was filtered (0.45 μm), and the absorbance of
inflammasome pathway may be a good entry point (Xu et al., 2019). So, Dox at 273 nm was recorded by a spectrophotometer. The encapsula-
this study aimed to construct the effect of Dox:CS/CMCS-NPs on NLRP3 tion efficiency (EE) (Eq. 1) and loading capacity (LC) (Eq. 2) were ex-
inflammasome and IL-1β in a human gingival fibroblasts (HGFs) in- pressed by the following equation (Feng et al., 2014):
flammatory model stimulated by Porphyromonas gingivalis-lipopoly-
total amount of Dox added − free Dox
saccharide (P. gingivalis-LPS) and find a new pathway for the treatment EE (%) = × 100
total amount of Dox added (1)
of periodontal disease.
total amount of Dox added − free Dox
2. Materials and methods LC (%) = × 100
weight of nanoparticles (2)

2.1. Materials
2.4. Bacteriostatic testing
CS (molecular weight, MW:10 kDa, degree of deacetylation, DD: 89
%) and CMCS (MW: 12 kDa, DD: 81 %, degree of substitution, DS: 92 P. gingivalis ATCC 33277 was used to identify antibacterial activity
%) was supplied by Department of Marine biology, Ocean University of of CS/CMCS-NPs and Dox:CS/CMCS-NPs. It was inoculated in culture
China. Triphenyl phosphate (TPP) was purchased from Sigma (St. Louis, bottle at 37 °C, under anaerobic conditions, and cultured by water bath
USA). Alpha minimum essential medium (α-MEM), fetal bovine serum, thermostat JBXL-70X, Jingbo at 200 rpm for 5 days. In the experimental
penicillin/ streptomycin and trypsin in this experiment were purchased group, 500 μg/ml CS/CMCS-NPs and Dox:CS/CMCS-NPs were added
from Biological Industries (Kibbutz Beit Haemek, Israel). Doxycycline, and cultured for 5 days under the same conditions. The cultured bac-
phosphate buffer saline (PBS) solution, Tryptic Soy Agar (TSA) medium terial liquid was coated on the blood plate with TSA medium, and the
and Cell Counting Kit-8 (CCK-8) assay were provided by Solarbio number of bacterial colonies was observed after culture at 37 °C for 5
(Shanghai, China). Porphyromonas gingivalis ATCC 33277 was supplied days.
by microbial culture preservation center of Guangdong Province
Guangzhou, China. Bacterial culture bottle was supplied by BD BACTEC 2.5. Cell culture, identification and effect of Dox:CS/CMCS-NPs on cell
Benex Limited Dun Laoghaire, Ireland. Porphyromonas gingivalis viability
Lipopolysaccharide (P. gingivalis-LPS) was supplied provided by
InvivoGen (San Diego, USA). Adenosine Triphosphate (ATP) was Primary HGFs were derived from adolescent gingival tissues

2
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

removed during alveolar surgery at the Department of Stomatology, the 2.8. Statistical analysis
Affiliated Hospital of Qingdao University. The present study with the
patients’ informed consent was approved by the Research Ethics Data are presented as mean ± SD. Statistical analyses were per-
Committee of the Affiliated Hospital of Qingdao University, P.R. China. formed by one-way ANOVA followed by the Student's t test. Differences
The cells were cultured in complete medium, α-MEM containing 10 % were considered significant at p < 0.05.
fetal bovine serum and 1 % penicillin/streptomycin, in a humidified
incubator of 5 % CO2 at 37 °C. Cells passaged three to five times were 3. Results and discussion
used in the following analysis. HGFs were identified by immuno-
fluorescence staining with anti-vimentin (VIM) and anti-cytokeratin 3.1. Preparation and characterization Dox:CS/CMCS-NPs
(CK).
Studies on cytocompatibility of P. gingivalis-LPS were carried out in As shown in Fig. 1, Dox:CS/CMCS-NPs were produced by ionic ge-
the supplementary material (Figs. S1 and S2) to determine the required lation in Dox, positively-charged CS and negatively-charged CMCS at
concentration of LPS in the experiment. HGFs at a density of 103 were pH 6.0. TPP was used as a crosslinking agent to stabilize the structure of
incubated with CS/CMCS-NPs and Dox:CS/CMCS-NPs (0–1000 μg/ml) the NPs. According to Feng et al., the most stable content were obtained
for 24, 48 and 72 h at 37 °C in a 96-well cell culture plate and 10 μl/ at a ratio of 3:4:0.5 of CS: CMCS: TPP (Feng et al., 2013). At this ratio,
well CCK-8 was added. After incubating for 2 h at 37 °C, the optical CS/CMCS-NPs had a stable mean particle size (203.1 ± 10.51 nm), PDI
density of the samples at 450 nm was measured. (0.23 ± 0.10) and zeta potential (+32.3 ± 0.4 mV). In our study, we
used a ratio of 1:3:4:0.5 for Dox: CS: CMCS: TPP. At this ratio, Dox:CS/
CMCS-NPs had a stable mean particle size, PDI and zeta potential
2.6. Total RNA isolation and quantitative real-time polymerase chain shown in Table 2. The absolute value of the zeta potential was greater
reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) than 30 mV indicated that the prepared NP system was stable.
Scanning electron micrographs, transmission electron micrographs
The total RNA from induced, treated and normal HGFs was isolated and particle size distribution of CS/CMCS-NPs and Dox:CS/CMCS-NPs
using the RNAiso Plus (TRIzol method). RNA was reverse transcribed were shown in Fig. 2. The results showed that both NPs were typically
using the RT reagent Kit with gDNA Eraser. The cDNA was synthesized spherical and capsular. They had good dispersion, clear edges, no ag-
for SYBR Assay in the Light Cycler LC480 (Roche). mRNA expression glomeration and uniform particle distribution. The average particle size
was normalized to the levels of the housekeeping gene GAPDH. The of CS/CMCS-NPs and Dox:CS/CMCS-NPs were showed in volume frac-
relative level of expression of a gene was calculated using the 2−ΔΔCt tion (Table 2). The results were consistent with those obtained by the
method. Primers are listed in the Supplementary Table 1. particle size analyzer.
The cell culture supernatant from HGFs were collected, centrifuged Most of the current studies on local drug delivery systems in-
by centrifuge tube (1 ml) for 10 min at 250×g and stored at −20 °C for vestigate the effect, and few studies investigate the mechanism of action
cytokine measurement. The level of IL-1β was measured with a com- (Ji, Zhao, Deng, & Lu, 2010; Sah, Dewangan, & Suresh, 2019). In this
mercial ELISA kit following the manufacturers’ protocols. study, a novel drug-loaded NP system, Dox:CS/CMCS-NPs, was suc-
cessfully constructed as a periodontal pocket administration. Both the
anti-inflammatory effect on cell inflammation model and deeper un-
2.7. Western blot analysis derstanding of the mechanism of action were explored. The particle size
measurement showed that the Dox:CS/CMCS-NPs loaded with the drug
The total protein from induced, treated and normal HGFs was col- was approximately 50 nm larger than that of CS/CMCS-NPs, and the
lected by lysing in (5×) Laemmli SDS buffer containing 0.01 % bro- PDI was smaller. Encapsulation efficiency (75 ± 7.21) % and loading
mophenol blue, 1 % β-mercaptoethanol, and a protease inhibitor capacity (28 ± 4.01) % had good performance which indicated that
cocktail (Roche). Subsequently, equal amounts of protein were sepa- Dox was successfully packed. Previous studies have shown that CS/
rated by SDS-PAGE gels and transferred onto PVDF membranes. After CMCS-NPs have slow-release properties, resulting in the Dox:CS/CMCS-
blocking with 5 % fat-free milk, the membranes were washed with NPs to release Dox continuously over longer periods of time, thus
TBST and incubated with primary and secondary antibodies according achieving better efficacy (Feng et al., 2013).
to the manufacturers’ instructions. Primary antibodies against NLRP3
(1:1000), ASC (1:1000), caspase-1 (1:1000) and IL-1β (1:1000) were 3.2. Culture and identification of HGFs
from Cell Signaling Technology. Secondary HRP-linked antibodies
against rabbit IgG (1:8000) and anti-GAPDH (1:5000) were from To identify gingival fibroblasts, we observed the cell morphology
Elabscience. The protein was analyzed using an enhanced chemilumi- and characterized the specific protein markers by immunofluorescence.
nescence kit and the band intensity was quantified with Image J 1.46 r Adhesion cells of human gingival tissue appeared about 7 days after the
and GraphPad Prism 7.04 software. GAPDH served as the loading primary culture, showing a spindle shape, which is one of the most
control. characteristic shapes of gingival fibroblasts (Park, Salem, Semlali,
Leung, & Rouabhia, 2017). Additionally, the cells retained their spindle
Table 1 shape after passage (Fig. 3A). Immunofluorescence images showed that
Sequence of primers used for quantitative polymerase chain reaction assays. intracellular anti-vimentin staining was positive (Fig. 3B) and in-
Gene Primer Sequence (5′-3′) tracellular anti-keratin staining was negative (Fig. 3C), which further
indicated that the obtained cells were gingival fibroblasts (Mohd Nor,
NLRP3 FORWARD: TGGCTGTAACATTCGGAGATTGTGG Berahim, Azlina, Mokhtar, & Kannan, 2017). In summary, these results
REVERSE: GCTTCTGGTTGCTGCTGAGGAC
suggest that the cells originating from human tissue masses were gin-
Caspase-1 FORWARD: GGTGCTGAACAAGGAAGAGATGGAG
REVERSE: TGCCTGAGGAGCTGCTGAGAG gival fibroblasts.
PYCARD (ASC) FORWARD: GCTGCTGGATGCTCTGTA
REVERSE: GGCTGGTGTGAAACTGAAG 3.3. Bacteriostatic properties of CS/CMCS-NPs and Dox:CS/CMCS-NPs
IL-1β FORWARD: CAGTGGCAATGAGGATGA
REVERSE: TAGTGGTGGTCGGAGATT
The results showed that the liquid mediums of P. gingivalis (negative
GAPDH FORWARD: TGAAGGTCGGAGTCAACGGATTTGGT
REVERSE: CATGTGGGCCATGAGGTCCACCAC control) and P. gingivalis with CS/CMCS-NPs were cloudy, whereas the
liquid medium of P. gingivalis with Dox and Dox:CS/CMCS-NPs was

3
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

Fig. 1. Schematic illustrations showing the preparation and action pattern of Dox:CS/CMCS-NPs.

Table 2 effects. Bacterial adhesion growth on plaque biofilm is difficult to re-


Mean particle size, PDI and zeta potential of CS/CMCS-NPs and Dox:CS/CMCS- move and can resist the killing effect of antibiotics and host defense
NPs. function (Sanz, van Winkelhoff, & Working Group 1 of Seventh
Mean particle size (nm) PDI Zeta potential (mV) European Workshop on, 2011). Among them, P. gingivalis, especially in
the lesion area or active site of chronic periodontitis, can activate a host
CS/CMCS-NPs 203.1 ± 10.51 0.23 ± +32.3 ± 0.4 inflammatory reaction and destroy periodontal tissues (Trindade et al.,
0.10
2014). In this study, the antibacterial effect of Dox:CS/CMCS-NPs on P.
Dox:CS/CMCS-NPs 252.3 ± 4.78 0.12 ± +30.6 ± 0.4
0.08 gingivalis was very effective. We speculated that because chitosan has
good bacteriostatic properties and has been proven to inhibit P. gingi-
valis (Liu et al., 2019; Peng, Hsieh, Chen, Tsai, & Chen, 2016). As a
clear after the oscillating culture for 5 days (Fig. 4A). The formation of powerful antibiotic, Dox is superior in bacteriostasis (Golub,
bacterial colony was not observed in the Dox and Dox:CS/CMCS-NPs, Ramamurthy, McNamara, Greenwald, & Rifkin, 1991). It should be
while P. gingivalis in CS/CMCS-NPs, which were cultured in the same noted that P. gingivalis grows in a specific environment that requires a
condition, were denser (Fig. 4B). The colonies were counted, and the high demand for an oxygen-free environment and nutrition (Song et al.,
number of colonies were showed in Fig. 4C. 2019). Dox:CS/CMCS-NPs changed the composition of the liquid cul-
The surface of the Dox:CS/CMCS-NPs has a positive charge, which ture medium and the growth environment of bacteria, which has a
enables better targeting of plaque biofilm with a negative charge (Ng, synergistically effect on the improvement of bacteriostatic properties of
Ke, Lee, Hedrick, & Yang, 2013). In other words, the packed Dox can be Dox:CS/CMCS-NPs.
delivered to the plaque biofilm more accurately and kill bacteria more
effectively (Fig. 1). This was the advantage of the composite nano- 3.4. Cytocompatibility of Dox:CS/CMCS-NPs
system constructed in the experiment. It could be greatly improved the
efficiency of Dox, reduce the dosage, and thus reduce the risk of side We investigated the effects of different concentrations of Dox:CS/

Fig. 2. SEM, TEM and particle size distribution (volume fraction) of CS/CMCS-NPs (A) and Dox:CS/CMCS-NPs (B).

4
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

Fig. 3. Images of passaged cells (A), the staining of specific proteins for cell identification (B, C). HGFs were observed under the microscope (A). Immunofluorescence
images showed that intracellular anti-vimentin staining was positive (B) and intracellular anti-cytokeratin staining was negative (C).

Fig. 4. Bacteriostatic testing (P. g referred to Porphyromonas gingivalis) of CS/CMCS-NPs (500 μg/ml), Dox (10 μg/ml) and Dox:CS/CMCS-NPs (500 μg/ml) cultured
with Porphyromonas gingivalis for 5 days at 37 ℃ and anaerobic environment (A). Agar culture of bacterial suspension (B) and the number of individual colonies (C).

5
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

Fig. 5. Treated HGFs with CS/CMCS-NPs and Dox:CS/CMCS-NPs in different concentration (62.5, 125, 250, 500, 1000 μg/ml) for 24 h, 48 h, and 72 h. (A) Images of
cells after 24 h and 72 h treated with Dox:CS/CMCS-NPs. (B) Cell viability detected by CCK-8 assay.

CMCS-NPs on HGFs to determine the appropriate concentration of NPs in the blank control, indicating a model of cellular inflammation.
for cell stimulation. After 24 h culture, there was no significant dif- However, adding Dox:CS/CMCS-NPs to the LPS + ATP group sig-
ference in cell morphology or the cell number of the experimental and nificantly inhibited the protein expression of IL-1β. The above results
control groups. After 72 h culture, the number of cells in the 1000 μg/ all proved that Dox:CS/CMCS-NPs had anti-inflammatory effect.
ml group increased compared with that in the blank control group, and To detect the effect of Dox:CS/CMCS-NPs on P. gingivalis-LPS +
24 h group. The difference was statistically significant; however, the ATP-induced NLRP3 inflammasome (Fig. 7A and B) and IL-1β (Fig. 7C),
morphology did not change (Fig. 5A). the expression of related proteins on the total intracellular protein of
The CCK-8 experiment showed that cell activity slightly increased HGFs was studied. LPS + ATP-induced HGFs stimulated with 1 μg/ml
with the increase of NP concentration at 24 h, 48 h or 72 h, while the P. gingivalis-LPS for 11 h followed by stimulation with 5 mM ATP for 1
number of cells in the 72 h group increased compared with that in the h. LPS + ATP + Dox-induced HGFs stimulated with 1 μg/ml P. gingi-
24 h group, and the difference was statistically significant (Fig. 5B). At valis-LPS for 11 h followed by stimulation with 5 mM ATP and 10 μg/ml
62.5, 125, 250, 500, 1000 μg/ml, the Dox:CS/CMCS-NPs had no ob- Dox for 1 h. LPS + ATP + NPs-induced HGFs stimulated with 1 μg/ml
vious inhibitory effect on cell viability, and the concentrations can be P. gingivalis-LPS for 11 h followed by stimulation with 5 mM ATP and
used. 500 μg/ml Dox:CS/CMCS-NGs for 1 h. Based on the gray value of the
The effect of CS/CMCS-NPs on cells is mild, even slightly promoting Western Blot strip, the LPS + ATP addition increased the expression of
cell growth (Wang et al., 2016). We found that the CS/CMCS-NPs had NLRP3 inflammasome and IL-1β in HGFs, while Dox:CS/CMCS-NPs
no obvious inhibitory effect on cell viability at 62.5, 125, 250, 500 or inhibited LPS + ATP-induced NLRP3 (Fig. 7D), ASC (Fig. 7E), Pro-
1000 μg/ml (Fig. 5B). This proved that the CS/CMCS-NPs were not caspase-1 (Fig. 7F), Cleaved caspase-1 (Fig. 7G), Pro- IL-1β (Fig. 7H)
toxic to HGFs. In this experiment, the action of Dox:CS/CMCS-NPs with and Cleaved IL-1β (Fig. 7I) in HGFs. Doxycycline alone could also
a concentration less 1000 μg/ml for 72 h showed no decrease in cell down-regulate protein level of NLRP3 inflammasome and IL-1β (Fig. 7),
activity, which proves that Dox:CS/CMCS-NPs were cell-friendly. which is consistent with our previous results (Xu et al., 2019). However,
there is no significance in the inhibitory effect of Dox:CS/CMCS-NPs
3.5. Effects of Dox:CS/CMCS-NPs on the expression of NLRP3 and doxycycline on NLRP3 inflammasome and IL-1β, which proved that
inflammasome and IL-1β induced by P. gingivalis-LPS and ATP our nano-drug delivery system has good physical properties but does
not reduce the effectiveness of doxycycline.
The mRNA level of NLRP3, ASC and Caspase-1 in the LPS + ATP The regulatory effect of Dox and chitosan on immunity should not
group were significantly higher than those in the blank control group be ignored (Chang, Wu, Wu, Huang, & Tsai, 2019; Li et al., 2015;
(Fig. 6A–C). This implied that LPS + ATP had a synergistic effect on the Preshaw, 2018). Dox alone has been shown to down-regulate the ex-
activation of NLRP3 inflammasome and we had successfully established pression of NLRP3 inflammasome, and our previous study also found
the model of inflammasome. However, Dox:CS/CMCS-NPs significantly that Dox has a similar effect in HGFs (Zhang et al., 2017). This was
inhibited the expression of LPS + ATP-induced NLRP3, ASC and Cas- illustrated in the supplementary materials (Figs. S3 and S4). CS/CMCS-
pase-1 at the gene level. NPs alone had no down-regulation effect on NLRP3 inflammasomes
To study the effect of LPS + ATP and Dox:CS/CMCS-NPs on in- whether in mRNA or protein level. This was illustrated in the supple-
flammasome activation and IL-1β production, the mRNA in HGFs and mentary materials (Fig. S5). Furthermore, the regulatory effect of
their cellular supernatant induced by LPS + ATP was investigated. Dox:CS/CMCS-NPs on HGFs and the NLRP3 inflammasome model on
Fig. 6D illustrated Dox:CS/CMCS-NPs did not increase the expression of related genes and proteins was observed. Our results demonstrated that
IL-1β. The Dox:CS/CMCS-NPs addition in the LPS + ATP group in- Dox:CS/CMCS-NPs had a strong inhibitory effect on LPS + ATP-in-
hibited the expression of IL-1β at the gene level. In Fig. 6E, the protein duced NLRP3 inflammasome by down-regulating the expression of
expression of IL-1β in the group with LPS + ATP was greater than that NLRP3, ASC and Caspase-1 in HGFs. Importantly, the expression of IL-

6
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

Fig. 6. (A, B, C, D, E) HGFs infected by LPS + ATP and treated by Dox:CS/CMCS-NPs. (A, B, C, D) The level of mRNA of NLRP3, ASC, Caspase-1 and IL-1β analyzed
by quantitative real-time polymerase chain reaction. (E) The level of protein of IL-1β analyzed by ELISA. The mRNA levels of cytokines in untreated controls were set
as 1.0. Bars show the levels of cytokines with mean SD (n = 3) and analyzed by the Student's t test. *p < 0.05. **p < 0.01. ***p < 0.001 versus control.

1β in protein or at the gene level was down-regulated in HGFs treated experiments in this study and drafted the article. Professor Qihui Zhou
by Dox:CS/CMCS-NPs. That means that Dox:CS/CMCS-NPs had an anti- directed the synthesis of the materials, analysis and interpretation of
inflammatory effect in HGFs. There may be two possible mechanisms by data, and revise this article critically for important intellectual content.
which Dox:CS/CMCS-NPs down-regulates LPS + ATP-induced NLRP3 Zhongxin Jiang contributed to acquisition of data of cell identification
inflammasome in HGFs or Dox:CS/CMCS-NPs killed bacteria and in- and wrote this part of the article. Yanwen Wang and Xiaohui Qiu
teracted with their endotoxins (Henehan, Montuno, & De Benedetto, contributed to acquisition, analysis and interpretation of data of genes
2017). On the other hand, as an immune-related pattern recognition as well as wrote this part of the article. Kai Yang contributed to ac-
receptor, inflammasome could be directly regulated by Dox:CS/CMCS- quisition, analysis and interpretation of data of protein as well as wrote
NPs through the regulation of immune pathways (Fig. 1) (Broz & Dixit, this part of the article. Professor Qiuxia Ji contributed to the concep-
2016). tion, designed of the study and revise this article critically for important
intellectual content. All authors have read and approved the final
4. Conclusion submission.

The new drug-loaded nanoparticles Dox:CS/CMCS-NPs prepared by


ion crosslinking method have a complete morphology and stable Acknowledgements
structure, and have good bacteriostasis against Porphyromonas gingi-
valis. Dox:CS/CMCS-NPs can effectively down-regulate the mRNA and This work was primarily supported by the National Natural Science
protein levels of NLRP3 inflammasome and IL-1β induced by LPS + Foundation of China (Grant No. 81401526), the Scientific Research
ATP in HGFs. It provides a potential new option for the rational ad- Foundation of Qingdao University (Grant No. DC1900009689), the
ministration of doxycycline in the clinical treatment of periodontal Natural Science Foundation of Shandong Province, China (Grant No.
disease. ZR2019QC007 and Grant No. ZR2019MH003), China Postdoctoral
Science Foundation (Grant no. RZ1900011066), the Major Research
Plan of the National Natural Science Foundation of China (Grant no.
Author contribution 91849209).

Shuo Xu is the first author of this paper, completed most of the

7
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

Fig. 7. LPS + ATP increasing the expression and Dox and Dox:CS/CMCS-NPs inhibiting the expression of protein levels of NLRP3 and ASC (A), Pro-caspase-1 and
Cleaved caspase-1 (B), Pro-IL-1β and Cleaved IL-1β (C) in HGFs. Western blot were performed to assess the protein level of NLRP3 (D), ASC (E), Pro-caspase-1 (F),
Cleaved caspase-1 (G), IL-1β (H) and Cleaved IL-1β (I). The blots were stripped and re-probed with GAPDH as a loading control. Data are presented as mean SEM of
three independent experiments. *p < 0.05. **p < 0.01. ***p < 0.001 versus control.

Appendix A. Supplementary data 249–260 discussion 260-241.


Graves, D. (2008). Cytokines that promote periodontal tissue destruction. The Journal of
Periodontology, 79(8 Suppl), 1585–1591.
Supplementary material related to this article can be found, in the Gregoriadis, G. (1978). Liposomes in therapeutic and preventive medicine: The devel-
online version, at doi:https://doi.org/10.1016/j.carbpol.2020.116163. opment of the drug-carrier concept. Annals of the New York Academy of Sciences, 308,
343–370.
Gujarathi, N. A., Rane, B. R., & Patel, J. K. (2012). pH sensitive polyelectrolyte complex of
References O-carboxymethyl chitosan and poly (acrylic acid) cross-linked with calcium for sus-
tained delivery of acid susceptible drugs. International Journal of Pharmaceutics,
Broz, P., & Dixit, V. M. (2016). Inflammasomes: Mechanism of assembly, regulation and 436(1–2), 418–425.
signalling. Nature Reviews Immunology, 16(7), 407–420. Hajishengallis, G. (2015). Periodontitis: From microbial immune subversion to systemic
Buduneli, E., Vardar, S., Buduneli, N., Berdeli, A. H., Turkoglu, O., Baskesen, A., & Atilla, inflammation. Nature Reviews Immunology, 15(1), 30–44.
G. (2004). Effects of combined systemic administration of low-dose doxycycline and Hajishengallis, G., Liang, S., Payne, M. A., Hashim, A., Jotwani, R., Eskan, M. A., & Curtis,
alendronate on endotoxin-induced periodontitis in rats. The Journal of Periodontology, M. A. (2011). Low-abundance biofilm species orchestrates inflammatory periodontal
75(11), 1516–1523. disease through the commensal microbiota and complement. Cell Host & Microbe,
Chang, S. H., Wu, G. J., Wu, C. H., Huang, C. H., & Tsai, G. J. (2019). Oral administration 10(5), 497–506.
with chitosan hydrolytic products modulates mitogen-induced and antigen-specific Hansen, J. D., Vojtech, L. N., & Laing, K. J. (2011). Sensing disease and danger: A survey
immune responses in BALB/c mice. International Journal of Biological Macromolecules, of vertebrate PRRs and their origins. Developmental and Comparative Immunology,
131, 158–166. 35(9), 886–897.
Chatzopoulos, G. S., Mansky, K. C., Lunos, S., Costalonga, M., & Wolff, L. F. (2019). Henehan, M., Montuno, M., & De Benedetto, A. (2017). Doxycycline as an anti-in-
Sclerostin and WNT-5a gingival protein levels in chronic periodontitis and health. flammatory agent: Updates in dermatology. Journal of the European Academy of
Journal of Periodontal Research Supplement. Dermatology and Venereology: JEADV, 31(11), 1800–1808.
Feng, C., Sun, G., Wang, Z., Cheng, X., Park, H., Cha, D., & Chen, X. (2014). Transport Hu, F., Zhou, Z., Xu, Q., Fan, C., Wang, L., Ren, H., & Chen, X. (2018). A novel pH-
mechanism of doxorubicin loaded chitosan based nanogels across intestinal epithe- responsive quaternary ammonium chitosan-liposome nanoparticles for periodontal
lium. European Journal of Pharmaceutics and Biopharmaceutics: Official Journal of treatment. International Journal of Biological Macromolecules, 129, 1113–1119.
Arbeitsgemeinschaft Fur Pharmazeutische Verfahrenstechnik EV, 87(1), 197–207. Huck, O., Elkaim, R., Davideau, J. L., & Tenenbaum, H. (2015). Porphyromonas gingi-
Feng, C., Wang, Z., Jiang, C., Kong, M., Zhou, X., Li, Y., & Chen, X. (2013). Chitosan/o- valis-impaired innate immune response via NLRP3 proteolysis in endothelial cells.
carboxymethyl chitosan nanoparticles for efficient and safe oral anticancer drug Innate Immunity, 21(1), 65–72.
delivery: in vitro and in vivo evaluation. International Journal of Pharmaceutics, Jain, N., Jain, G. K., Javed, S., Iqbal, Z., Talegaonkar, S., Ahmad, F. J., & Khar, R. K.
457(1), 158–167. (2008). Recent approaches for the treatment of periodontitis. Drug Discovery Today,
Gjoseva, S., Geskovski, N., Sazdovska, S. D., Popeski-Dimovski, R., Petrusevski, G., 13(21–22), 932–943.
Mladenovska, K., & Goracinova, K. (2018). Design and biological response of dox- Ji, Q. X., Zhao, Q. S., Deng, J., & Lu, R. (2010). A novel injectable chlorhexidine ther-
ycycline loaded chitosan microparticles for periodontal disease treatment. mosensitive hydrogel for periodontal application: Preparation, antibacterial activity
Carbohydrate Polymers, 186, 260–272. and toxicity evaluation. Journal of Materials Science Materials in Medicine, 21(8),
Golub, L. M., Elburki, M. S., Walker, C., Ryan, M., Sorsa, T., Tenenbaum, H., & Gu, Y. 2435–2442.
(2016). Non-antibacterial tetracycline formulations: Host-modulators in the treat- Jing, L., Kim, S., Sun, L., Wang, L., Mildner, E., Divaris, K., & Offenbacher, S. (2019). IL-
ment of periodontitis and relevant systemic diseases. International Dental Journal, 37- and IL-35/IL-37-Producing plasma cells in chronic periodontitis. Journal of Dental
66(3), 127–135. Research22034519847443.
Golub, L. M., Ramamurthy, N. S., McNamara, T. F., Greenwald, R. A., & Rifkin, B. R. Kassebaum, N. J., Bernabe, E., Dahiya, M., Bhandari, B., Murray, C. J., & Marcenes, W.
(1991). Tetracyclines inhibit connective tissue breakdown: New therapeutic im- (2014). Global burden of severe periodontitis in 1990-2010: A systematic review and
plications for an old family of drugs. Critical Reviews in Oral Biology and Medicine: an meta-regression. Journal of Dental Research, 93(11), 1045–1053.
Official Publication of the American Association of Oral Biologists, 2(3), 297–321. Li, R., Luo, X., Pan, Q., Zineh, I., Archer, D. F., StanWilliams, R., & Chegini, N. (2015).
Goncalves, R. M., Antunes, J. C., & Barbosa, M. A. (2012). Mesenchymal stem cell re- Doxycycline alters the expression of inflammatory and immune-related cytokines and
cruitment by stromal derived factor-1-delivery systems based on chitosan/poly chemokines in human endometrial cells: Implications in irregular uterine bleeding.
(gamma-glutamic acid) polyelectrolyte complexes. European Cells & Materials, 23, Human Reproduction, 30(1), 249.

8
S. Xu, et al. Carbohydrate Polymers 237 (2020) 116163

Lian, D., Dai, L., Xie, Z., Zhou, X., Liu, X., Zhang, Y., & Chen, Y. (2018). Periodontal periodontal drug delivery. Colloids and Surfaces B, Biointerfaces, 178, 185–198.
ligament fibroblasts migration injury via ROS/TXNIP/Nlrp3 inflammasome pathway Sanz, M., Ceriello, A., Buysschaert, M., Chapple, I., Demmer, R. T., Graziani, F., & Vegh,
with Porphyromonas gingivalis lipopolysaccharide. Molecular Immunology, 103, D. (2017). Scientific evidence on the links between periodontal diseases and diabetes:
209–219. Consensus report and guidelines of the joint workshop on periodontal diseases and
Liu, J., Xiao, Y., Wang, X., Huang, L., Chen, Y., & Bao, C. (2019). Glucose-sensitive de- diabetes by the international diabetes Federation and the European Federation of
livery of metronidazole by using a photo-crosslinked chitosan hydrogel film to inhibit periodontology. Diabetes Research and Clinical Practice.
Porphyromonas gingivalis proliferation. International Journal of Biological Sanz, M., van Winkelhoff, A. J., & Working Group 1 of Seventh European Workshop on, P.
Macromolecules, 122, 19–28. (2011). Periodontal infections: Understanding the complexity–Consensus of the se-
Mohd Nor, N. H., Berahim, Z., Azlina, A., Mokhtar, K. I., & Kannan, T. P. (2017). venth European workshop on periodontology. Journal of Clinical Periodontology,
Identification and characterization of intraoral and dermal fibroblasts revisited. 38(Suppl 11), 3–6.
Current Stem Cell Research & Therapy, 12(8), 675–681. Schenten, D., & Medzhitov, R. (2011). The control of adaptive immune responses by the
Nelson, M. L., & Levy, S. B. (2011). The history of the tetracyclines. Annals of the New York innate immune system. Advances in Immunology, 109, 87–124.
Academy of Sciences, 1241, 17–32. Song, J. M., Woo, B. H., Lee, J. H., Yoon, S., Cho, Y., Kim, Y. D., & Park, H. R. (2019). Oral
Ng, V. W., Ke, X., Lee, A. L., Hedrick, J. L., & Yang, Y. Y. (2013). Synergistic co-delivery of administration of Porphyromonas gingivalis, a major pathogen of chronic period-
membrane-disrupting polymers with commercial antibiotics against highly oppor- ontitis, promotes resistance to paclitaxel in mouse xenografts of oral squamous cell
tunistic bacteria. Advanced Material, 25(46), 6730–6736. carcinoma. International Journal of Molecular Sciences, 20(10).
Olsen, I., & Yilmaz, O. (2016). Modulation of inflammasome activity by Porphyromonas Trindade, F., Oppenheim, F. G., Helmerhorst, E. J., Amado, F., Gomes, P. S., & Vitorino, R.
gingivalis in periodontitis and associated systemic diseases. Journal of Oral (2014). Uncovering the molecular networks in periodontitis. Proteomics Clinical
Microbiology, 8, 30385. Applications, 8(9–10), 748–761.
Park, H. J., Salem, M., Semlali, A., Leung, K. P., & Rouabhia, M. (2017). Antimicrobial Wang, J., Xu, M., Cheng, X., Kong, M., Liu, Y., Feng, C., & Chen, X. (2016). Positive/
peptide KSL-W promotes gingival fibroblast healing properties in vitro. Peptides, 93, negative surface charge of chitosan based nanogels and its potential influence on oral
33–43. insulin delivery. Carbohydrate Polymers, 136, 867–874.
Peng, P. C., Hsieh, C. M., Chen, C. P., Tsai, T., & Chen, C. T. (2016). Assessment of Xu, S., Zhou, Q., Fan, C., Zhao, H., Wang, Y., Qiu, X., & Ji, Q. (2019). Doxycycline inhibits
photodynamic inactivation against periodontal Bacteria Mediated by a chitosan hy- NAcht Leucine-rich repeat protein 3 inflammasome activation and interleukin-1beta
drogel in a 3D gingival model. International Journal of Molecular Sciences, 17(11). production induced by Porphyromonas gingivalis-lipopolysaccharide and adenosine
Preshaw, P. M. (2018). Host modulation therapy with anti-inflammatory agents. triphosphate in human gingival fibroblasts. Archives of Oral Biology, 107, 104514.
Periodontology 2000, 76(1), 131–149. Zhang, L., & Deng, S. (2019). Effects of astragaloside IV on inflammation and immunity in
Ragelle, H., Riva, R., Vandermeulen, G., Naeye, B., Pourcelle, V., Le Duff, C. S., ... Preat, rats with experimental periodontitis. Brazilian Oral Research, 33, e032.
V. (2014). Chitosan nanoparticles for siRNA delivery: Optimizing formulation to in- Zhang, W., Xie, X., Wu, D., Jin, X., Liu, R., Hu, X., & Cao, Y. (2017). Doxycycline at-
crease stability and efficiency. Journal of Controlled Release : Official Journal of the tenuates leptospira-induced IL-1beta by suppressing NLRP3 inflammasome priming.
Controlled Release Society, 176, 54–63. Frontiers in Immunology, 8, 857.
Sah, A. K., Dewangan, M., & Suresh, P. K. (2019). Potential of chitosan-based carrier for

You might also like