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

International Journal of Biological Macromolecules 126 (2019) 1158–1166

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules

journal homepage: http://www.elsevier.com/locate/ijbiomac

Mechanism of the anti-inflammatory activity by a polysaccharide from


Dictyophora indusiata in lipopolysaccharide-stimulated macrophages
Yalin Wang a, Li Lai b, Liping Teng a, Yuhong Li a, Jianqing Cheng a, Jinghua Chen b,⁎, Chao Deng a,⁎
a
Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
b
School of Pharmaceutics Science, Jiangnan University, Wuxi 214122, China

a r t i c l e i n f o a b s t r a c t

Article history: Dictyophora indusiata polysaccharides (DIP) shows antioxidant, anti-tumor and immunostimulatory activities.
Received 1 October 2018 However, the anti-inflammatory roles of DIP in NLRP3 inflammasome in LPS-stimulated macrophages are still
Received in revised form 2 January 2019 not well defined. In this study, we investigated the mechanism of the anti-inflammatory activity of DIP in LPS-
Accepted 4 January 2019
primed RAW264.7 macrophages. Our data showed that DIP inhibited NF-κB signal pathway via modulating
Available online 6 January 2019
TLR4 expression, phosphorylation of IκBα and nuclear translocation of NF-κB-p65 subunit. Meanwhile, DIP re-
Keywords:
duced inflammasome activation via decreasing NLRP3 expression in cytoplasmic pools, limiting self-assembly
Dictyophora indusiata polysaccharide of NLRP3 inflammasome, as well as the subsequent activation of caspase-1 and the secretion of IL-1β and IL-
Anti-inflammatory activity 18. For the first time, we show that the anti-inflammatory activity of DIP is mediated by inhibiting TLR4/NF-κB
NLRP3 inflammasome signal pathway and NLRP3 inflammasome activation during LPS-induced acute inflammation in RAW264.7
macrophages.
© 2019 Elsevier B.V. All rights reserved.

1. Introduction [11]. Upon activation, NF-κB could bind to the promoter of target
genes encoding inflammatory mediators (e.g. IL-6) [12,13].
Inflammation is the normal protective reaction that occurs in re- NOD-like receptor protein 3 (NLRP3) inflammasomes are cytosol-
sponse to infection, tissue injury, trauma or noxious stimuli [1–4]. Lim- localized large multiprotein oligomers consisted mainly of adaptor pro-
ited and excessive inflammatory response to pathogens can increase tein apoptosis-associated speck-like protein (ASC), NLRP3 and caspase-
susceptibility to infection and chronic inflammatory condition, respec- 1. Caspase-1 is a proteolytic enzyme mediating the cleavage and activa-
tively [5–7]. Innate immune cells (e.g. macrophages, mast cells, fibro- tion of secreted IL-18 and IL-1β that are prominent mediators of the in-
blasts, dendritic cells as well as circulating leukocytes) contribute to flammatory response. Increasing evidence indicates that there might be
the identification of a variety of pathogen- and host-derived danger sig- a close relationship between NLRP3 inflammasome and inflammatory
nals with surface or intracellular-expressed pattern recognition recep- diseases including cancer, arthritis, liver diseases and type 2 diabetes
tors (PRRs) [8]. These signals include the whole pathogens and [14–17].
pathogen-associated molecular patterns (PAMPs) (e.g. microbial The activation of NLRP3 inflammasomes requires two distinct sig-
nucleic acids, lipoproteins and carbohydrates), and host-derived signals nals: the priming signal typically induced by TLR4 that triggers the ex-
of cellular damage such as danger-associated molecular patterns pression of pro-IL-1β, pro-IL-18 and the elements of the
(DAMP) and environmental irritants. The early sensing of dangerous inflammasome (NLRP3) through signal transduction pathways (e.g.
signals involves PRRs on the cell surface [e.g., membrane-bound Toll- NF-κB) [18,19]; the activation of the signals in the second stage induced
like receptors (TLRs)] and in the cytoplasm [e.g., NOD-like receptors assembly of the inflammasome complex consisting of ASC, NLRP3 and
(NLRs)] [9]. inactive caspase-1, activation of caspase-1 and the cleavage of precursor
Innate immune system recognizes Gram-negative bacteria through cytokines IL-1β and IL-18 [20,21]. Several molecules with similar func-
TLR4 by identifying lipopolysaccharide (LPS) that is a part of the micro- tion of PAMPs or DAMPs, such as reactive oxygen species (ROS) and ex-
bial outer membrane triggering inflammatory response [10]. LPS in- tracellular ATP, may trigger the second signal [22].
duced TLR4 activation and signal transduction, which then led to Dictyophora indusiata, known as queen of mushrooms, is one of the
nuclear factor κappa B (NF-κB) activation and nuclear translocation most popular edible mushrooms worldwide due to fantastic taste and
nutrition [23]. Also, it is used as the active components for agents with
therapeutic properties. Nowadays, extensive studies have been focused
⁎ Corresponding authors. on the roles of D. indusiata polysaccharide (DIP) with low toxicity and
E-mail addresses: jhchenwhut@126.com (J. Chen), dcao@jiangnan.edu.cn (C. Deng). multiple beneficial potencies, such as antioxidant [24], antitumor

https://doi.org/10.1016/j.ijbiomac.2019.01.022
0141-8130/© 2019 Elsevier B.V. All rights reserved.
Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166 1159

[25,26] and immunostimulatory activities [27]. However, little is known defatted resultant residue was extracted with distilled water at 90 °C.
about the anti-inflammatory roles of DIP in NLRP3 inflammasome in Afterwards, the supernatant was precipitated with ethanol and refriger-
LPS-stimulated macrophages. In this study, we aim to elucidate whether ated at 4 °C for 24 h. Upon removal of free proteins via Sevage method,
DIP can modulate the NF-κB signal transduction pathway and NLRP3 the yield polysaccharide fraction was further purified through gel
inflammasome-mediated IL-18 and IL-1β secretion in LPS-primed and filtration chromatography (Sephadex G-200 chromatogram), followed
ATP-stimulated macrophages. by dialysis and lyophilization. The product we obtained was a homoge-
neous component, with a molecular weight and total sugar content
2. Materials and methods of 650 kDa and 95.67%, respectively. DIP was a β-(1 → 3)-D-glucan
with side branches of β-(1 → 6)-glucosyl units according to gas
2.1. Chemicals chromatography–mass spectrometry and infrared spectrum analysis
[23]. Endotoxin in DIP was very low (b0.01 ng/mL) with a concentration
LPS extracted from Escherichia coli 0111:B4 was purchased from of 25 μg/mL.
Sigma-Aldrich (St. Louis, MO, USA). Fetal bovine serum (FBS), penicillin
G, streptomycin and Dulbecco's modified eagle medium (DMEM) were 2.3. Cell culture and stimulation
purchased from Gibco (Carlsbad, CA, USA). Bay11-7082 was purchased
from Beyotime (Shanghai, China). Macrophage-like cell line RAW264.7, purchased from the Cell Bank
of Chinese Academy of Sciences (Shanghai, China), were cultured in
2.2. DIP preparation DMEM containing 10% FBS, streptomycin (100 μg/mL) and penicillin
(100 U/mL) at 37 °C in a humidified atmosphere containing 5% CO2.
Fruiting bodies of D. indusiata were purchased from Changning, Cells in the exponential growth phase were used for experiments.
China. DIP isolation and purification was carried out according to To determine the roles of DIP in modulating LPS-induced NLRP3
our previous experiments with slight modification [28]. Briefly, inflammasome activation, RAW264.7 cells were divided into the follow-
D. indusiata powder was grinded with ethyl acetate and ethanol to re- ing groups: (i) blank control (cells cultured on DMEM medium); (ii) LPS
moved lipids. Then the water extract was centrifugated to collect the (1 μg/mL) group; (iii–v) LPS + DIP groups, subject to LPS (1 μg/mL) and
polysaccharide-enriched fractions in the supernatant, while the DIP with a concentration of 12.5 μg/mL, 25 μg/mL, or 50 μg/mL for 3 h,

Fig. 1. Inhibitory effects of DIP on LPS-induced expression of TLR4 in RAW264.7 macrophages. (A–C) The expression of TLR4 in the cell surface was detected by flow cytometry. (D) Relative
fluorescence intensity. Different lowercase letters indicated statistical differences (P b 0.05).
1160 Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166

respectively. Afterwards, 5 mM ATP was added in LPS group and LPS at room temperature. Then they were incubated with the rabbit anti-
+ DIP groups and incubated for 1 h to assess NLRP3 inflammasome pro-caspase-1 (Abcam, UK), anti-NLRP3 (Abcam, UK), anti-phospho-
activation. IκBα (Abcam, UK), anti-IκBα (Abcam, UK), anti-GAPDH (Beyotime Bio-
technology, China) or goat anti-IL-1β (R&D system, China) overnight at
2.4. Western blot analysis 4 °C. Subsequently, the membranes were washed with TBST and incu-
bated with horseradish peroxidase-conjugated goat anti-rabbit or donkey
For inflammasome activation analysis, cellular supernatant was col- anti-goat IgG for 1 h. After washing the membrane with TBST thrice for
lected, and remaining cells were rinsed twice with ice-cold PBS and 5 min, proteins were detected using an ECL detection kit (Beyotime,
lysed with RIPA lysis buffer containing proteinase inhibitors. Protein ex- Shanghai, China) according to the manufacturer's instructions. Bands
pression was determined by Western blot analysis according to the stan- were visualized and photographed using Bio-Rad image analysis system
dard procedures. Protein content was measured with BCA method. (Bio-Rad, CA, USA). The same membrane was probed with GAPDH for
Denatured proteins were separated in a 10% SDS-PAGE, and then were loading control. Semiquantitative protein quantification was performed
transferred to polyvinylidene difluoride membrane and blocked for 1 h using the ImageJ software (Bio-Rad, CA, USA).

Fig. 2. DIP inhibited LPS-induced NF-κB transcriptional activation. DIP reduced the LPS-induced activation of NF-κB by decreasing phosphorylation of IκB-α, the nuclear translocation of NF-
κB-p65 subunit and the protein secretion regulated by this signaling pathway. (A) Expression of P-IκBα and IκBα protein determined using Western blot analysis. (B, C) Protein semi-
quantification for P-IκBα and IκBα. (D) Nuclear translocation of NF-κB-p65 was observed under a CLSM. (E) Secretion of IL-6 detected by ELISA. Different lowercase letters indicated
statistical differences (P b 0.05).
Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166 1161

2.5. Determination of Caspase-1 activation 2.8. Effects of DIP on the combination of NLRP3 with ASC

Caspase-1 activity was detected using a commercial kit (Beyotime, The colocalization of NLRP3 with ASC was detected by immunofluo-
Shanghai, China) according to the manufacturer's instructions. In brief, rescence staining with a CLSM. Cells were rinsed thrice with cold PBS
cellular extract was incubated with Ac-YVAD-pNA in a 96-well microti- and fixed with cold 4% paraformaldehyde solution at room temperature
ter plate for 3 h at 37 °C. The absorbance values of pNA were measured for 12 min. Next, the cells were blocked with immunostaining blocking
using a microplate reader at 405 nm (Spectra Max M5, Molecular De- buffer at room temperature for 1 h and then incubated with rat mono-
vices, USA). clonal antibody NLRP3 (1:200, Abcam, UK) and rabbit monoclonal anti-
body ASC (1:200, Abcam, UK) overnight at 4 °C. Cy3-labeled goat anti-
rabbit IgG (Beyotime, Shanghai, China) was added to the cells and incu-
2.6. Measurement of secretion of IL-6, IL-1β and IL-18
bated for 1 h. DAPI was added to each plate and incubated for 5 min at
room temperature in the dark, followed by observation under a CLSM.
Concentration of mature IL-18, IL-1β and IL-6 in RAW264.7 cells was
determined using a commercial ELISA kit (Invitrogen, USA) according to
the manufacturer's protocol. The tests were performed at least in 2.9. Detection of ROS generation
triplicate.
ROS generation was determined using commercial kit (Beyotime,
Shanghai, China) according to the manufacturer's protocol. The alive
2.7. Effect of DIP on NF-κB nuclear translocation cells were collected and detected by flow cytometry.

Immunofluorescence method was applied to determine the effects


of DIP on NF-κB nuclear translocation. RAW264.7 cells (1 × 105) were 2.10. Effect of TLR4 expression
seeded in a confocal plate and incubated at 37 °C in a humidified atmo-
sphere with 5% CO2 for 24 h. Cells were washed thrice with cold PBS and Flow cytometry was applied to detect the effects of DIP on TLR4 ex-
fixed with cold paraformaldehyde (4%) solution at room temperature pression. Briefly, the cells in each group were treated with PE anti-
for 12 min. After washing with cold PBS thrice for 5 min, the cells CD284 (TLR4) antibody (Bio Legend, USA) for 2 h. Finally, cells were
were blocked with immunostaining blocking buffer (Beyotime, Shang- washed with cold PBS thrice and detected by flow cytometry.
hai, China) at room temperature for 1 h, and then were incubated
with rat monoclonal antibody NF-kB p65 (1:200, Beyotime, Shanghai, 2.11. Statistical analysis
China) overnight at 4 °C. The cells were washed with PBS thrice for
5 min and incubated with Cy3-labeled goat anti-rabbit IgG (Beyotime, All data are presented as the means ± standard deviation (SD). Data
Shanghai, China) for 1 h in the dark. DAPI (Beyotime, Shanghai, China) analyses were performed using the SPSS 23.0 software. Significant dif-
was added to each plate and incubated for 5 min. Finally, the cells ferences were evaluated by one-way ANOVA. P b 0.05 was considered
were washed with cold PBS thrice and observed under a CLSM. to be statistically significant.

Fig. 3. Effects of DIP on expression of inflammasome components in priming of inflammasome activation. (A) Western blot analysis of pro-caspase-1 and NLRP3 protein expression. (B–D)
Protein semi-quantifications were shown for pro-caspase-1 and NLRP3. Different lowercase letters indicated statistical differences. (P b 0.05).
1162 Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166

3. Results 3.2. DIP inhibited the transcriptional activation of NF-κB

3.1. DIP inhibited TLR4 expression To examine the roles of DIP in LPS-induced TLR4 signal transduction,
we evaluated the activation of the NF-κB pathway. DIP down-regulated
Compared with the LPS-treatment group, DIP (12.5 μg/mL, 25 μg/mL the expression of P-IκBα (Fig. 2A and B) and inhibited NF-κB nuclear
and 50 μg/mL) significantly reduced fluorescence intensity (P b 0.05, translocation of the p65 subunit (Fig. 2D). Also, it triggered decrease
Fig. 1A–C). This indicated that DIP could inhibit TLR4 expression in of IL-6 (Fig. 2E), of which the gene expression was induced by LPS and
LPS-induced RAW264.7 macrophages. Particularly, DIP (25 μg/mL) regulated through NF-κB in LPS-stimulated RAW264.7 macrophages.
showed the most obvious inhibiting effects on TLR4 expression The inhibition effect of NF-κB transcriptional activation by DIP was the
(Fig. 1D). most obvious at a concentration was 25 μg/mL.

Fig. 4. Effects of DIP on the secretion of IL-1β and IL-18 via activating NLRP3 inflammasome in macrophages. (A) Confocal immunofluorescence images of NLRP3 and ASC in macrophages.
(B) Analysis of caspase-1 activity using Ac-YVAD-pNA; the activity of caspase-1 was reflected by the production of pNA. (C) IL-1β protein expression. (D, E) Protein semi-quantification was
shown for supernatant IL-1β and IL-1β of lysate. (F) Secretion of IL-18 was detected by ELISA. Different lowercase letters indicated statistical differences (P b 0.05).
Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166 1163

Fig. 5. IL-1β secretion was inhibited by DIP in LPS-primed RAW264.7 macrophages. (A, B) IL-1β concentrations in culture supernatant were detected by ELISA. Cells were stimulated with
or without 5 mM ATP for 1 h. Different lowercase letters indicated statistical differences (P b 0.05).

3.3. DIP decreased the inflammasome-priming 3.4. DIP inhibited inflammasome activation, IL-18 and IL-1β secretion

Protein expression in supernatant pro-caspase-1 and cytoplasmic In blank control, the expression of NLRP3 with ASC was very low,
pool of NLRP3 was prominently decreased by the DIP in the LPS- whereas, colocalization NLRP3 with ASC was obviously increased in
primed macrophages (Fig. 3A–D). Among these groups, DIP in different LPS group. Combination of NLRP3 with ASC in experimental groups
concentrations can suppress the inflammasome-priming, and DIP (25 was gradually reduced in the 12.5 μg/mL and 25 μg/mL dose groups,
μg/mL) showed the most evident inhibitory effects. but the inhibitory effects showed attenuation when treating with 50

Fig. 6. Effects of DIP on ROS generation. (A–C) ROS generation in different DIP dose groups (12.5 μg/mL DIP, 25 μg/mL DIP and 50 μg/mL). (D) Relative fluorescence intensity. Different
lowercase letters indicated statistical differences (P b 0.05).
1164 Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166

μg/mL DIP (Fig. 4A). DIP significantly inhibited the production of acti- to block NF-κB, 30 μM) for 3 h before stimulating with ATP (5 mM) for
vated caspase-1, especially at a concentration of 25 μg/mL (Fig. 4B). 1 h. Bay11-7082 inhibited LPS and ATP induced protein expressions in
DIP significantly inhibited caspase-1-mediated IL-1β (Figs. 4D and NLRP3 inflammasome-priming (P-IkBα, supernatant pro-caspase1 and
5A) and IL-18 (Fig. 4F) generation, however, the inhibitory effects NLRP3) and activation phase (supernatant IL-1β, Fig. 7). These results
showed gradual attenuation with the increase of DIP concentration. indicated that NF-κB was critical for the activation of the NLRP3
Moreover, DIP also significantly inhibited the secretion of IL-1β when inflammasome in LPS and ATP-treated cells. DIP (12.5 μg/mL) showed
RAW264.7 macrophages were only primed with LPS in the absence of similar anti-inflammatory effects compared to that of Bay11-7082.
any other NLRP3 inflammasome-activating stimuli (Fig. 5B).
4. Discussion
3.5. DIP inhibited the ROS production
Inflammasome regulation deficiency involves in numerous chronic
ROS production involved in NLRP3 inflammasome activation. On inflammation and autoimmune diseases, such as inflammatory bowel
this basis, we investigated the effects of DIP on ROS production by ana- disease (IBD), systemic sclerosis, systemic lupus erythematosus and
lyzing the fluorescence intensity of DCF-DA. DIP (12.5 μg/mL, 25 μg/mL rheumatoid arthritis [29]. NLRP3 inflammasome plays a major role in
and 50 μg/mL) could significantly inhibit the ROS generation in LPS- the chronic inflammation as it recognizes microbial and cell stress com-
primed and ATP-activated macrophages, and DIP (25 μg/mL) showed ponents. Additionally, it serves as a platform for caspase-1 activation
the strongest inhibiting effect on ROS generation (Fig. 6). and pro-inflammatory cytokine IL-1β and IL-18 during the process of in-
flammatory and autoimmune diseases. Moreover, TLRs are the key me-
3.6. NF-κB was required for the activation of the NLRP3 inflammasome diators of innate host defenses in the intestine involving in the mucosal
immune homoeostasis. TLR4 signaling induces the inflammatory re-
To elucidate the regulatory relationship of NF-κB in LPS and ATP- sponse through the activation of NF-κB [30]. Thus, TLR4/NF-κB signal
stimulated macrophages, cells were treated with LPS in the presence pathway and NLRP3 inflammasome are potential therapeutic targets
or absence of DIP (12.5 μg/mL) or Bay11-7082 (inhibitors with ability for the treatment of inflammatory and autoimmune diseases [31].

Fig. 7. NF-κB was required for the activation of the NLRP3 inflammasome. (A) Western blot analysis findings. (B–H) Semi-quantification analysis of protein in supernatant IL-1β,
supernatant pro-caspase-1, lysis IL-1β, lysis pro-caspase-1, NLRP3, P-IkBα and IkBα. Different lowercase letters indicated statistical differences (P b 0.05).
Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166 1165

Current research on inflammation has focused on the use of natural with ASC), proteolytic cleavage of inactive pro-caspase-1 and the secre-
products as alternative and effective anti-inflammatory therapies with tion of IL-1β and IL-18 in LPS-primed and ATP-activated macrophages.
low toxicities and minimal adverse effects. These polysaccharides We also analyzed IL-1β secretion modulation through DIP in LPS-
from bacteria, fungi and mushrooms show high efficacy and lower tox- primed macrophages in the presence or absence of ATP stimulation.
icity than other therapeutic agents [32,33]. In a previous study, polysac- DIP can inhibit the LPS-induced release of IL-1β independently of
charide of Hericium erinaceus attenuated dextran sodium sulfate (DSS)- inflammasome-triggered signals. Thus, it inhibited IL-1β secretion in
induced colitis in C57BL/6 mice via regulating NF-κB signal pathway and LPS-primed and ATP-mediated NLRP3 inflammasome activation and
the secretion of IL-6, IL-1β and TNF-α [34]. Meanwhile, Lycium also inhibited IL-1β secretion in LPS-primed and non-ATP-stimulated
barbarum polysaccharides improved hepatic injury through NF-κB and inflammasome-activated macrophages. These indicated that LPS was
NLRP3 pathways in a steatohepatitis mouse model subject to a methio- sufficient to promote IL-1β synthesis in the absence of inflammasome-
nine choline deficient diet [35]. In the present study, we found DIP re- activating stimulus independent of the NLRP3 inflammasome sensor in-
pressed the NF-κB signal transduction pathway and NLRP3 duction. However, in LPS-primed macrophages, ATP served as an ago-
inflammasome-mediated IL-1β and IL-18 secretion in LPS-stimulated nist of the second signal of NLRP3 inflammasome activation triggered
macrophages. the maturation and secretion of high amounts of IL-1β that was similar
Cell surface receptors, NF-κB signal transduction pathway and the to the previous studies [11,36].
production of inflammasome components are the priming events es- When a LPS-primed macrophage is subjected to an activating stim-
sential for inflammasome activation and subsequent IL-1β and IL-18 se- ulus in a permissive cellular context, the NLRP3 inflammasome is acti-
cretion in the sequential regulation of inflammasome activation. Our vated by recruiting ASC and caspase-1. However, the exact molecular
results indicated that DIP could reduce TLR4 expression, NF-κB pathway events resulting in NLRP3 activation are still not well defined. Several
activation and signal transduction by inhibiting IκB-α phosphorylation NLRP3 activation models are mutually interacted, such as pore forma-
and nuclear translocation of NF-κB-p65 subunit in LPS-primed macro- tion, ion redistribution, lysosomal disruption, metabolic dysfunction
phages. Also, the secretion of IL-6 that was a proinflammatory bio- and mitochondrial dysfunction models. In these models, the common
marker controlled by NF-κB signaling pathway was inhibited. For mechanisms leading to NLRP3 activation were closely related to the
inflammasome components, DIP inhibited the expression of NLRP3 movement of K ions. High levels of extracellular ATP, that may occur
and pro-caspase-1 of the inflammasome components. However, DIP in cases of dying cells adjacent to the inflammasome-containing cells,
did not trigger a dose-dependent inhibition for these priming events es- also resulted in ion flux by opening the P2X purinoceptor 7 channel.
sential for inflammasome activation, which was possibly related to the Once opening of the channel, the NLRP3 inflammasome was activated
fact that DIP mainly exhibited an effect of inhibiting inflammation at by allowing external flow of K ions. In addition, mitochondrial ROS are
lower concentrations and an effect of promoting inflammation at a also essential for NLRP3 activation (Fig. 8). Many NLRP3 triggers can
higher concentration [28]. cause the increase in mitochondrial ROS production, and NLRP3 is
DIP is reported to modulate inflammasome activation by inhibiting inhibited by pre-incubating with some antioxidants [8,37]. Therefore,
self-assembly of inflammasome (e.g. the oligomerization of NLRP3 the inhibitory effects of DIP on IL-1β and IL-18 secretion may be related

Fig. 8. The anti-inflammatory mechanism of DIP in LPS-primed and ATP-stimulated macrophages.


1166 Y. Wang et al. / International Journal of Biological Macromolecules 126 (2019) 1158–1166

to the intrinsic antioxidant activity, and the potential inhibition of ROS Acknowledgment
production would be associated with the strong anti-inflammatory ef-
fects of DIP. This work was supported by National Natural Science Foundation of
As DIP could inhibit NF-κB signal pathway via modulating the TLR4 China (No. 31700708) and the Natural Science Foundation of Jiangsu
expression and reduce inflammasome activation via decreasing NLRP3 Province (No. BK20170181).
expression, it could be applied in treating the chronic inflammation
and autoimmune diseases associated with TLR4/NF-κB signal pathway References
and NLRP3 inflammasome (e.g. IBD). It has been reported that the pri-
[1] S. Mariathasan, D.M. Monack, Nat. Rev. Immunol. 7 (2007) 31–40.
mary function of intestinal bacteria is to enhance host digestive effi- [2] K. Newton, V.M. Dixit, Cold Spring Harb. Perspect. Biol. 4 (2012), a006049. .
ciency by degrading dietary polysaccharides [38]. In other words, it is [3] C. Schinkel, A. Gaertner, J. Zaspel, S. Zedler, E. Faist, M. Schuermann, Clin. J. Pain 22
suggested that polysaccharides, including DIP, might be degraded intes- (2006) 235–239.
[4] R.P. Abdul, J.K. Song, J. Funct. Foods 38 (2017) 415–426.
tinal environment. However, it has also been reported that lentinan, a [5] D. Bi, Q. Lai, N. Cai, et al., J. Agric. Food Chem. 66 (2018) 2083–2091.
polysaccharide extracted from Lentinus edodes with similar structure [6] P. Libby, Nutr. Rev. 65 (2007) S140–S146.
as DIP, shows therapeutic potential in IBD model mice possibly by [7] K. Deuteraiou, G. Kitas, A. Garyfallos, T. Dimitroulas, Rheumatol. Int. 38 (2018)
1345–1354.
inhibiting TLR4/NF-κB signaling-mediated inflammatory responses [8] M.S. Mangan, E.J. Olhava, W.R. Roush, H.M. Seidel, G.D. Glick, E. Latz, Nat. Rev. Drug
[39]. We speculated that DIP might have similar effects in these inflam- Discov. 17 (2018) 588–606.
mation mice model. In future, more studies are needed to investigate [9] K. Schroder, R. Zhou, J. Tschopp, Science 327 (2010) 296–300.
[10] Q. Yu, S. Nie, J. Wang, et al., Food Chem. Toxicol. 66 (2014) 14–22.
whether DIP could survive the gut and get absorbed in intestinal
[11] N. Martinez-Micaelo, N. González-Abuín, M. Pinent, A. Ardévol, M. Blay, Mol. Nutr.
environment. Food Res. 59 (2015) 262–269.
In conclusion, DIP attenuated TLR4/NF-κB signal pathway activation [12] J. Guo, J. Chen, X. Lu, et al., J. Agric. Food Chem. 66 (2018) 4758–4767.
via modulating TLR4 expression, phosphorylation of IκB-α and the nu- [13] P.P. Tak, G.S. Firestein, J. Clin. Invest. 107 (2001) 7–11.
[14] A. Abderrazak, K.E. Hadri, E. Bosc, et al., J. Pharmacol. Exp. Ther. 357 (2016) 487–494.
clear translocation of NF-κB-p65 subunit. Additionally, DIP was a de- [15] C. Choulaki, G. Papadaki, A. Repa, et al., Arthritis Res. Ther. 17 (2015) 257.
pressor for the activation of NLRP3 inflammasome. It attenuated the [16] C. Lebeaupin, E. Proics, C.H. Bieville, et al., Cell Death Dis. 6 (2015), e1879. .
cytoplasmic pool of NLRP3 expression, ROS production, NLRP3-ASC olig- [17] G. Müzes, F. Sipos, Curr. Drug Targets 16 (2015) 249–257.
[18] L. Liang, X. Tan, Q. Zhou, et al., Invest. Ophthalmol. Vis. Sci. 54 (2013) 402–414.
omerization, caspase-1 activation and IL-1β and IL-18 extracellular se- [19] Y. Qiao, P. Wang, J. Qi, L. Zhang, C. Gao, FEBS Lett. 586 (2012) 1022–1026.
cretion in LPS-primed and ATP-stimulated macrophages. Therefore, [20] J.K. Dowling, L.A. O'Neill, Crit. Rev. Biochem. Mol. Biol. 47 (2012) 424–443.
DIP can be used as an anti-inflammatory agent, especially for NLRP3 [21] W. Xin, Q. Wang, D. Zhang, C. Wang, Eur. J. Pharmacol. 814 (2017) 240–247.
[22] C.A. Janeway, R. Medzhitov, Annu. Rev. Immunol. 20 (2002) 197–216.
inflammasome. [23] C. Deng, Z. Hu, H. Fu, M. Hu, X. Xu, J. Chen, Int. J. Biol. Macromol. 51 (2012) 70–75.
[24] C. Deng, H. Fu, L. Teng, Z. Hu, X. Xu, J. Chen, Int. J. Biol. Macromol. 61 (2013)
Abbreviations 453–458.
[25] S. Han, C. Ma, M. Hu, et al., Cell Biochem. Funct. 35 (2017) 414–419.
[26] H. Fu, C. Deng, L. Teng, et al., Int. J. Med. Mushrooms 17 (2015) 151–160.
DIP Dictyophora indusiata polysaccharide [27] C. Jin, R.A. Flavell, J. Clin. Immunol. 30 (2010) 628–631.
TLR4 Toll-like receptor-4 [28] C. Deng, J. Shang, H. Fu, J. Chen, H. Liu, J. Chen, Int. J. Biol. Macromol. 91 (2016)
752–759.
NF-κB nuclear factor κappa B
[29] X. Yichen, X. Wenna, S. Wenru, Clin. Exp. Allergy 48 (2018) 1080–1091.
IL interleukin [30] S.W. Brubaker, K.S. Bonham, I. Zanoni, J.C. Kagan, Annu. Rev. Immunol. 33 (2015)
PRRs pattern recognition receptors 257–290.
PAMPs pathogen-associated molecular patterns [31] A.P. Perera, D. Kunde, R. Eri, Curr. Pharm. Des. 23 (2017) 2321–2327.
[32] K.H. Lee, M. Park, K.Y. Ji, et al., Immunobiology 219 (2014) 802–812.
DAMPs danger-associated molecular patterns [33] I. Lavi, L. Nimri, D. Levinson, I. Peri, Y. Hadar, B. Schwartz, J. Gastroenterol. 47 (2012)
FBS fetal bovine serum 504–518.
LPS lipopolysaccharide [34] Y. Rena, Y. Geng, Y. Du, et al., J. Nutr. Biochem. 57 (2018) 67–76.
[35] J. Xiao, F. Wang, E.C. Liong, K.F. So, G.L. Tipoe, Int. J. Biol. Macromol. 120 (2018)
NLRP3 NOD-like receptor protein 3 1480–1489.
ASC apoptosis-associated speck-like protein [36] K. Schroder, V. Sagulenko, A. Zamoshnikova, et al., Immunobiology 217 (2012)
ROS Reactive oxygen species 1325–1329.
[37] X. Yin, J. Cell. Mol. Med. 22 (2018) 3264-3.
[38] L.V. Hooper, A.J. Macpherson, Nat. Rev. Immunol. 10 (2010) 159–169.
Conflict of interest [39] Y. Liu, J. Zhao, Y. Zhao, et al., J. Cell. Mol. Med. 00 (2018) 1–11.

The authors declare no conflict of interest.

You might also like