ArticlePDF Available

Acacetin inhibits inflammation by blocking MAPK/NF-κB pathways and NLRP3 inflammasome activation

Frontiers
Frontiers in Pharmacology
Authors:

Abstract and Figures

Objective: Our preliminary research indicates that acacetin modulates the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome, providing protection against Alzheimer’s Disease (AD) and cerebral ischemic reperfusion injury. The mechanisms of acacetin to inhibit the activation of the NLRP3 inflammasome remain fully elucidated. This study aims to investigate the effects and potential mechanisms of acacetin on various agonists induced NLRP3 inflammasome activation. Methods: A model for the NLRP3 inflammasome activation was established in mouse bone marrow-derived macrophages (BMDMs) using Monosodium Urate (MSU), Nigericin, Adenosine Triphosphate (ATP), and Pam3CSK4, separately. Western blot analysis (WB) was employed to detect Pro-caspase-1, Pro-Interleukin-1β (Pro-IL-1β) in cell lysates, and caspase-1, IL-1β in supernatants. Enzyme-Linked Immunosorbent Assay (ELISA) was used to measured the release of IL-1β, IL-18, and Tumor Necrosis Factor-alpha (TNF-α) in cell supernatants to assess the impact of acacetin on NLRP3 inflammasome activation. The lactate dehydrogenase (LDH) release was also assessed. The Nuclear Factor Kappa B (NF-κB) and Mitogen-Activated Protein Kinase (MAPK) signaling pathways related proteins were evaluated by WB, and NF-κB nuclear translocation was observed via laser scanning confocal microscopy (LSCM). Disuccinimidyl Suberate (DSS) cross-linking was employed to detect oligomerization of Apoptosis-associated Speck-like protein containing a Caspase Recruitment Domain (ASC), and LSCM was also used to observe Reactive Oxygen Species (ROS) production. Inductively Coupled Plasma (ICP) and N-(6-methoxyquinolyl) acetoethyl ester (MQAE) assays were utilized to determined the effects of acacetin on the efflux of potassium (K+) and chloride (Cl-) ions. Results: Acacetin inhibited NLRP3 inflammasome activation induced by various agonists, reducing the release of TNF-α, IL-1β, IL-18, and LDH. It suppressed the expression of Lipopolysaccharides (LPS)-activated Phosphorylated ERK (p-ERK), p-JNK, and p-p38, inhibited NF-κB p65 phosphorylation and nuclear translocation. Acacetin also reduced ROS production and inhibited ASC aggregation, thus suppressing NLRP3 inflammasome activation. Notably, acacetin did not affect K+ and Cl-ions efflux during the activation process. Conclusion: Acacetin shows inhibitory effects on both the priming and assembly processes of the NLRP3 inflammasome, positioning it as a promising new candidate for the treatment of NLRP3 inflammasome-related diseases.
This content is subject to copyright.
Acacetin inhibits inammation by
blocking MAPK/NF-κB pathways
and NLRP3 inammasome
activation
Juan Bu
1
, Yeledan Mahan
1
, Shengnan Zhang
1
, Xuanxia Wu
1
,
Xiaoling Zhang
1
, Ling Zhou
1
* and Yanmin Zhang
2
*
1
Medical and Translational Research Center, Peoples Hospital of Xinjiang Uygur Autonomous Region,
Urumqi, China,
2
Scientic Research and Education Center, Peoples Hospital of Xinjiang Uygur
Autonomous Region, Urumqi, China
Objective: Our preliminary research indicates that acacetin modulates the
nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain
containing 3 (NLRP3) inammasome, providing protection against Alzheimers
Disease (AD) and cerebral ischemic reperfusion injury. The mechanisms of
acacetin to inhibit the activation of the NLRP3 inammasome remain fully
elucidated. This study aims to investigate the effects and potential
mechanisms of acacetin on various agonists induced NLRP3 inammasome
activation.
Methods: A model for the NLRP3 inammasome activation was established in
mouse bone marrow-derived macrophages (BMDMs) using Monosodium Urate
(MSU), Nigericin, Adenosine Triphosphate (ATP), and Pam3CSK4, separately.
Western blot analysis (WB) was employed to detect Pro-caspase-1, Pro-
Interleukin-1β(Pro-IL-1β) in cell lysates, and caspase-1, IL-1βin supernatants.
Enzyme-Linked Immunosorbent Assay (ELISA) was used to measured the release
of IL-1β, IL-18, and Tumor Necrosis Factor-alpha (TNF-α) in cell supernatants to
assess the impact of acacetin on NLRP3 inammasome activation. The lactate
dehydrogenase (LDH) release was also assessed. The Nuclear Factor Kappa B
(NF-κB) and Mitogen-Activated Protein Kinase (MAPK) signaling pathways related
proteins were evaluated by WB, and NF-κB nuclear translocation was observed
via laser scanning confocal microscopy (LSCM). Disuccinimidyl Suberate (DSS)
cross-linking was employed to detect oligomerization of Apoptosis-associated
Speck-like protein containing a Caspase Recruitment Domain (ASC), and LSCM
was also used to observe Reactive Oxygen Species (ROS) production. Inductively
Coupled Plasma (ICP) and N-(6-methoxyquinolyl) acetoethyl ester (MQAE) assays
were utilized to determined the effects of acacetin on the efux of potassium (K+)
and chloride (Cl-) ions.
Results: Acacetin inhibited NLRP3 inammasome activation induced by various
agonists, reducing the release of TNF-α, IL-1β, IL-18, and LDH. It suppressed the
expression of Lipopolysaccharides (LPS)-activated Phosphorylated ERK (p-ERK),
p-JNK, and p-p38, inhibited NF-κB p65 phosphorylation and nuclear
translocation. Acacetin also reduced ROS production and inhibited ASC
aggregation, thus suppressing NLRP3 inammasome activation. Notably,
acacetin did not affect K+ and Cl-ions efux during the activation process.
OPEN ACCESS
EDITED BY
David M. Pereira,
University of Porto, Portugal
REVIEWED BY
Junping Zhang,
First Teaching Hospital of Tianjin University of
Traditional Chinese Medicine, China
Muhammad Furqan Akhtar,
Riphah International University, Pakistan
*CORRESPONDENCE
Ling Zhou,
zlrmyy@sina.com
Yanmin Zhang,
1518486455@qq.com
RECEIVED 31 August 2023
ACCEPTED 26 January 2024
PUBLISHED 08 February 2024
CITATION
Bu J, Mahan Y, Zhang S, Wu X, Zhang X, Zhou L
and Zhang Y (2024), Acacetin inhibits
inammation by blocking MAPK/NF-κB
pathways and NLRP3 inammasome activation.
Front. Pharmacol. 15:1286546.
doi: 10.3389/fphar.2024.1286546
COPYRIGHT
© 2024 Bu, Mahan, Zhang, Wu, Zhang, Zhou
and Zhang. This is an open-access article
distributed under the terms of the Creative
Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is
permitted, provided the original author(s) and
the copyright owner(s) are credited and that the
original publication in this journal is cited, in
accordance with accepted academic practice.
No use, distribution or reproduction is
permitted which does not comply with these
terms.
Frontiers in Pharmacology frontiersin.org01
TYPE Original Research
PUBLISHED 08 February 2024
DOI 10.3389/fphar.2024.1286546
Conclusion: Acacetin shows inhibitory effects on both the priming and assembly
processes of the NLRP3 inammasome, positioning it as a promising new candidate
for the treatment of NLRP3 inammasome-related diseases.
KEYWORDS
inammasome, NLRP3, acacetin, MAPK pathway, NF-κB pathway
1 Introduction
Inammation represents a biological paradox. While moderate
inammatory responses can confer benets to the host, aiding in the
combat against foreign pathogen invasions, excessive or chronic
inammation may lead to tissue damage and consequential
pathological changes. The nucleotide-binding oligomerization
domain (NOD)-like receptor pyrin domain containing 3
(NLRP3) inammasome plays a critical role in these
inammatory responses. Comprised of Apoptosis-associated
Speck-like protein containing a Caspase Recruitment Domain
(ASC), NLRP3, and pro-caspase-1, the NLRP3 inammasome,
upon activation, catalyzes the activation of caspase-1, which
mediates the discharge of inammatory cytokines Interleukin-1β
(IL-1β) and Interleukin-18 (IL-18), thereby leading to inammation
and pyroptosis (Xu Y. et al., 2023;Ma, 2023). Studies support the
implication of mitogen-activated protein kinase (MAPK) and
Nuclear Factor Kappa-B (NF-κB) pathways in modulating
NLRP3 inammasome activation, thereby participating in the
inammatory response (He et al., 2013;Robblee et al., 2016). The
activation of NLRP3 inammasome correlates with a variety of
diseases, including peritonitis (Qin and Zhao, 2023), arthritis (Wang
et al., 2023), Alzheimers disease (AD) (Lv et al., 2023), and ischemic
stroke (Han and Le, 2023), etc. Hence, the therapeutic efcacy of
NLRP3 inammasome inhibitors, which have manifested signicant
prognostic improvements in animal models of these diseases,
underscores the imperative for their discovery.
At present, identied inhibitors of NLRP3 inammasome
encompass agents such as sulforaphane (Kiser et al., 2021),
oridonin (He et al., 2018), BAY11-7082 (Irrera et al., 2017),
INF39 (Shi et al., 2021), MCC950 (Luo et al., 2019), CY-09
(Wang et al., 2022), INF-200 (Gastaldi et al., 2023), SB-222200
(Zhou et al., 2023), Alantolactone (Li W. et al., 2023), and
Tabersonine (Xu H.-W. et al., 2023), among others. Nevertheless,
these therapeutic agents encounter drawbacks including limited
specicity, short half-lives, suboptimal bioavailability, and an
uncertain clinical application outlook. Acacetin, also referred to
as robinin, is a avonoid compound extracted from various plants,
including chrysanthemums, Robinia pseudoacacia, and Saussurea
involucrata. This compound, characterized by its small molecular
weight, ability to penetrate the blood-brain barrier, minimal toxicity,
and extensive pharmacological activities (Singh et al., 2020). Our
previous study (Bu et al., 2019) revealed that acacetin mitigated
ischemic stroke in mouse models by inhibiting microglial
overactivation, modulating the NF-κB/NLRP3 pathway, and
downregulating inammatory cytokines such as TNF-α, IL-1β,
and IL-6. In addition, acacetin showed signicant neuroprotective
properties by diminishing infarct volume and ameliorating
neurological scores in these models. In vitro study, acacetin
increased the survival rate of microglial cells following the
oxygen-glucose deprivation and reoxygenation (OGD/R) injury,
decreased lactate dehydrogenase (LDH) release, stimulated
autophagy, and inhibited NLRP3 inammasome activation (Bu
et al., 2023). Furthermore, acacetin has demonstrated an ability
to impede NLRP3 inammasome activation, reduce inammatory
cytokine release, attenuate senile plaques development in AD mice,
improve cognitive and exploratory abilities in AD mice, and thereby
impart a protective role against AD (Bu et al., 2022). Given the
critical role that NLRP3 inammasome plays in the onset and
progression of inammatory diseases and the signicant
protective effects of acacetin on these diseases, questions arise
regarding the potential relationship between acacetins protective
role and its capacity to inhibit NLRP3 inammasome activation.
Herein, we initially isolated macrophages from mouse bone
marrow to explore the impact of acacetin on the activation of
NLRP3 inammasome induced by both canonical (i.e., by
activators monosodium urate (MSU), Nigericin, and adenosine
triphosphate (ATP)) and non-canonical pathways. The aim was
to clarify whether acacetin could suppress NLRP3 inammasome
activation, thus exerting anti-inammatory effects. Subsequently, we
investigated whether acacetin impacts the NF-κB and MAPK
pathways, sequentially inhibiting the priming of
NLRP3 inammasome. Following this, the study employed
Disuccinimidyl Suberate (DSS) cross-linking to detect ASC
oligomerization and laser scanning confocal microscopy (LSCM)
to monitor the production of Reactive Oxygen Species (ROS). The
impact of acacetin on potassium (K+) and chloride (Cl-) ions efux
was quantied using Inductively Coupled Plasma (ICP) and N-(6-
methoxyquinolyl) acetoethyl ester (MQAE) assays, respectively.
These methods facilitated a precise understanding of acacetins
role in the assembly and upstream processes of the
NLRP3 inammasome.
2 Materials and methods
2.1 Cell preparation, stimulation,
and grouping
Bone marrow-derived macrophages (BMDMs) were isolated
from 68 weeks old C57BL/6J mice supplied by Xinjiang Medical
University (License Number: SCXK (Xin) 20180002). Following
euthanization and sterilization, both femurs and tibias were excised
and the bone marrow was ushed into DMEM (C11965500BT,
Gibco, United States) medium using sterile phosphate buffer saline
(PBS, ZLI-9062, ZSGB-Bio, China). The obtained cell suspension
was centrifuged at 110 gfor 10 min at room temperature, after which
the supernatant was discarded. The resultant cell pellet was
resuspended in 2 mL of red blood cell lysis buffer and
maintained on ice for 5 min. Subsequently, 8 mL of DMEM
Frontiers in Pharmacology frontiersin.org02
Bu et al. 10.3389/fphar.2024.1286546
supplemented with 10% fetal bovine serum (FBS, FND500, Excell
Bio, China) and 1% penicillin-streptomycin (15070063, Gibco,
United States) was added. After mixing, the solution was
centrifuged at 220 g(with a centrifuge radius of 10 cm) for 5 min
at room temperature. The supernatant was discarded, and the cells
were then resuspended in DMEM containing 10 ng/mL of
Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF,
P00184, Solarbio, Beijing, China), 10% FBS, and 1% penicillin-
streptomycin. Ensuring a homogenous suspension through
repeated pipetting, the cells were cultured in a 37°C incubator.
After 3 days in culture, a complete medium replacement was
conducted, followed by a half medium change on day 5.
Microscopic examination revealed that the adherent cells had
satisfactory attachment, extending pseudopodia, and adopted a
spindle shape.
When cell conuency reached 80%, BMDMs cells (5 × 10
5
) were
seeded to a 12-well plate and then treated with LPS (50 ng/mL,
L2630, Sigma, United States) for 3 h. Afterwards, the cells were
treated with Acacetin (00017, Sigma, United States) for 0.5 h and
respectively stimulated with MSU (150 μg/mL, tlrl-msu/MSU-
4107, InvivoGen, France) for 4 h, Nigericin (10 μM, tlrl-nig,
InvivoGen, France) for 0.5 h, and ATP (2.5 mM, A2383-5G/
SLCD1216, Sigma, United States) for 0.5 h. For the activation of
non-canonical pathways, cells were pretreated with Pam3CSK4
(400 ng/mL, Tlrl-pms/PMS-41-03, InvivoGen, France) for 3 h,
then treated with Acacetin for 0.5 h, and then transfected with
LPS (500 ng/mL) using Lipofectamine 3000 (L3000-008/2218558,
ThermoFisher, United States of America) for 16 h. Both the
supernatant and cell pellets were collected for subsequent analyses.
Groups for assessing acacetins impact on NF-κB and MAPK
signaling pathways included: a control group, groups treated with
LPS for varying durations (0 min, 10 min, 30 min, 60 min), and
groups rst treated with LPS for these durations followed by the
addition of acacetin (10 μM). For evaluating acacetins effect on
NLRP3 inammasome activation, the study encompassed a control
group, an LPS treated group, an LPS + Nigericin group, and groups
treated with LPS + Nigericin followed by different concentrations of
acacetin (2.5 μM, 5 μM, and 10 μM). Further details are available in
the gure legends.
2.2 Cell samples collection and proteins
extraction
After the above treatments, supernatants from each group were
collected and combined with an equal volume of methanol and a
quarter volume of chloroform. This mixture was then vortexed
thoroughly and centrifuged at 14,000 × g at room temperature for
5 min. Following centrifugation, the resulting tri-layered solution
had proteins localized in the intermediate layer. The top layer was
carefully discarded, and 500 μL of methanol was added to the
remaining solution. This was followed by another centrifugation
at 16,000 × g for 5 min, after which the protein precipitate settled at
the bottom. The supernatant was removed, and the resultant pellet
was air-dried for about 5 min to get rid of residual methanol. The
dried protein pellet was then reconstituted in sample buffer and
subjected to heating at 101°C in a metal bath for 10 min. The
prepared protein sample was deemed t either for immediate
Western Blotting assays or storage at 20°C.
For the cellular proteins, cells were enzymatically digested
using 0.25% Trypsin-EDTA (25200056, Gibco, United States)
and gathered. These cells were then resuspended in 100 μLof
radioimmunoprecipitation assay (RIPA) buffer (AR0105, Boster
Bio, China), ensuring a homogenous mixture. The cell-lysis
mixture was allowed to incubate at 4°Cfor60min.A
centrifugation step at 14,000 × g for 15 min at 4°C allowed us
to collect the supernatant, which was subsequently mixed with
an appropriate volume of 5 × SDS-PAGE loading buffer infused
with β-mercaptoethanol. The denaturation of proteins was
achieved by placing this mixture in a 100°C-water bath for
5 min. After this, a nal centrifugation at 14,000 × g for
5 min was done to obtain a clear supernatant. Lastly, the
protein content was quantied using the standard BCA
method (Easy II Protein Quantitative Kit, DQ111-01,
TransGen Biotech, China), from which the volume equivalent
to 30 μg of protein was calculated, setting the stage for
downstream experimental endeavors.
2.3 Western blot
Electrophoresis conditions: the stacking gel was set at 80 V and
the separating gel at 100 V. Following electrophoresis, proteins from
the gel were transferred onto PVDF membranes (ISEQ00010/
IPVH00010, Millipore, United States). Then the membrane was
blocked with 5% bovine serum albumin (BSA) in TBST at room
temperature for 2 h. It was then incubated with primary antibodies
overnight at 4°C(Table 1). After washing thrice with TBST for
10 min each, the membrane was incubated with corresponding
secondary antibodies (diluted 1:5,000) at room temperature for
2 h. Another three 10-min TBST washes were done, followed by
an application of chemiluminescent reagent (mixed 1:1 of Solution
A and B, 2 mL each) on the membrane. Protein bands were detected
and captured using a chemiluminescence imaging system
(Chemiscope 3000, Clinx, China). The relative expression level
was calculated based on the absorbance ratio of the target gene
band to the β-actin band.
2.4 Enzyme-linked immunosorbent assay
Cytokine levels of IL-1β, IL-18, and TNF-αin collected cell
supernatants were measured using ELISA kits (EK201B/3-48,
EK218-48 and EK282/3-48, LiankeBio, China) following the
manufacturers instructions. After adding a stop solution to
terminate the reaction, the color change was measured
spectrophotometrically at a dual wavelength of 450 nm
(maximum absorption wavelength) and 570 nm (reference
wavelength) using a microplate reader (xMarkTM, Bio-Rad,
United States). The optical density (OD) at 570 nm was
subtracted from the OD at 450 nm to correct for optical
imperfections. The corrected OD values were used to determine
the concentrations of the cytokines in the supernatants by
comparing them to the standard curve.
Frontiers in Pharmacology frontiersin.org03
Bu et al. 10.3389/fphar.2024.1286546
2.5 LDH release assay
To quantify the level of cellular cytotoxicity and necrosis, a LDH
release assay was performed following the manufacturers
instructions. LDH is a stable enzyme, released upon cell lysis,
and it serves as a marker of cellular damage or death.
Supernatants from each group were collected and assayed for
LDH activity. The LDH assay was conducted using the LDH
Cytotoxicity Detection Kit (A020-2, NJJCBio, China). This kit
utilizes the conversion of lactate to pyruvate by LDH, with
simultaneous conversion of a tetrazolium salt (INT) into a red
formazan product. The amount of formazan product, which is
directly proportional to the LDH activity in the sample, can be
quantied by measuring the absorbance at 450 nm using a
microplate reader. The calculation formula of LDH activity is:
LDH Activity U/L
()
Absorbance of sample Absorbance of blank
Absorbance of standard Absorbance of standard blank
× Standard concentration × 1000
2.6 NF-κB nuclear translocation
The nuclear translocation of NF-κB was visualized using laser
scanning confocal microscopy (LSCM). After the above treatments,
the coverslips were thoroughly washed with three 2-min wash cycles
using phosphate-buffered saline (PBS). Subsequent steps were
xation with 4% paraformaldehyde for 20 min, permeabilization
with 0.5% Triton X-100 for 5 min, and blocking with 1% BSA
for 30 min, each followed by identical wash cycles. Following
blocking, cells were incubated at 37°C with primary NF-κB
p65 antibody (2 μg/mL, ab76302, Abcam, United Kingdom) for
2 h. A post-incubation wash cycle was performed before the cells
were exposed to a goat anti-rabbit IgG H&L secondary antibody
(2 μg/mL, ab205718, United Kingdom) for 1 h at 37°C in the dark.
Another wash cycle preceded DAPI counterstaining (1 μg/mL,
C0065, Solarbio, Beijing, China) for 5 min at room temperature.
After a nal wash cycle, cells were mounted with 50% glycerol and
imaged using a laser scanning confocal microscope (LSM700, Carl
Zeiss AG, Germany).
2.7 K + ions concentration
Prepared cell suspensions were centrifuged at 110 gfor 10 min,
discarding the supernatant and retaining the cellular pellet. Cells were
then washed twice with PBS, followed by centrifugation at 110 gfor
10 min, retaining the cellular pellet. The cellular pellet was resuspended
in 0.2 mL of deionized water and homogenized in an ice bath.
Homogenateswerethencentrifugedat450gfor 5 min, and 20 μL
of the supernatant was mixed with 180 μL of protein precipitant. This
was further centrifuged at 1370 gfor 5 min, and 50 μLofthe
supernatant was collected for potassium measurement. Protein
concentration in the samples was also determined to normalize the
K+ ions concentration. The concentration of K+ ions was measured
using a potassium ion assay kit (C001-2-1, NJJCBio, China), following
the manufacturers instructions. The calculation formula of the K+ ions
concentration in mmol/g of protein (mmol/gprot) is:
Potassium ions concentration mmolgprot

Absorbance of sample Absorbance of blank
Absorbance of standard Absorbance of blank
× standard concentration× sample dilution factor
÷ protein concentration in sample
TABLE 1 Primary antibodies used in Western blot analysis.
Protein Cat no. Dilution Transfer time (min) Company Info
NLRP3 ab263899 1:1000 120 abcam, United Kingdom
Caspase-1 ab179515 1:1000 60 abcam, United Kingdom
IL-1βab9722 1:500 60 abcam, United Kingdom
NF-κB p65 ab32536 1:800 60 abcam, United Kingdom
Phospho-NF-κB p65 ab76302 1:800 60 abcam, United Kingdom
IκBαab32518 1:1000 60 abcam, United Kingdom
Phospho-IκBα5209s 1:800 60 CST, United States
ERK ab184699 1:800 60 abcam, United Kingdom
Phospho-ERK ab201015 1:800 60 abcam, United Kingdom
JNK ab179461 1:1000 60 abcam, United Kingdom
Phospho-JNK ab76572 1:800 60 abcam, United Kingdom
p38 ab170099 1:1000 60 abcam, United Kingdom
Phospho-p38 ab195049 1:800 60 abcam, United Kingdom
β-Actin 100166-MM10 1:1000 60 Sino Biological, China
Frontiers in Pharmacology frontiersin.org04
Bu et al. 10.3389/fphar.2024.1286546
2.8 Cl- ions concentration
The concentration of Cl-ions was determined using MQAE
uorescent indicator. After the previous preparation, the
supernatant was discarded. The cells were then lysed with
ultrapure water and incubated at 37°C for 15 min. The lysate was
transferred to a new 1.5 mL EP tube and placed at 80°C for 30 min.
After centrifuging at 6200 × g for 5 min, the supernatant was
transferred to a fresh 1.5 mL EP tube. An equal volume of 50 μL
of MQAE (HY-D0090, MedChemExpress, United States) at a
concentration of 10 μM was added to the supernatant, and the
mixture was vortexed to ensure uniform mixing. Then, 80 μL of the
mixed solution was dispensed into a 96-well plate. Fluorescence
intensity was measured using a uorometer (VLB000D2,
ThermoFisher, United States), with the excitation wavelength set
at 355 nm and the emission wavelength at 460 nm.
2.9 Mitochondrial reactive oxygen
species levels
The levels of mitochondrial ROS were evaluated using LSCM.
Cells were adjusted to a concentration of 1 × 10
5
cells/mL and plated
onto adhesive glass coverslips (200 μL per coverslip). Following the
intervention, cells were incubated with 100 μL MitoSOX Red
(M36008, ThermoFisher, United States) mitochondrial
superoxide indicator at a nal concentration of 5 μM for 5 min at
37°C. Then, cells were washed with a three 2-min wash cycle using
PBS. Subsequently, cells were xed at room temperature for 20 min
using 4% paraformaldehyde, followed by another wash cycle. The
nuclei were counterstained with DAPI (1 μg/mL) in a dark room for
5 min at room temperature, followed by a nal wash cycle.
Coverslips were nally mounted using 50% glycerol and cells
were visualized using a laser scanning confocal microscope.
Quantication of ROS levels was performed using ImageJ
software (National Institutes of Health, United States).
2.10 ASC oligomerization assay
Cells were collected and lysed in 100 µL of RIPA buffer
(supplemented with protease and phosphatase inhibitors). The
lysate was incubated in an ice bath for 60 min before
centrifugation at 14000 g,4
°C for 15 min, with the supernatant
collected for further processing. The cell lysate was then treated for
oligomerized protein processing. The cells were digested with
trypsin (25200056, Gibco, United States) and resuspended in
0.5 mL of ice-cold buffer A. The lysate was centrifuged at 1800 g
at 4°C for 8 min to remove the precipitate. A 30 μL aliquot of lysate
was reserved for Western Blot of ASC as input controls. The
remaining supernatant was diluted in a 1:1 ratio with buffer A
and centrifuged at 2000 gat 4°C for 5 min. The supernatant was
collected and diluted with an equal volume of CHAPS buffer before
a centrifugation at 5000 gfor 8 min to precipitate ASC oligomers.
The supernatant was discarded and the pellet was resuspended in
50 µL of CHAPS buffer containing 4 mM of DSS (S1885, Sigma,
United States) and incubated at room temperature for 30 min for
protein cross-linking. The mixture was then centrifuged again at
5000 × g for 8 min at 4°C. The supernatant was discarded, and the
pellet was resuspended in 30 µL of protein loading buffer and
denatured at 90°C for 2 min. Subsequent steps were conducted
according to standard Western Blot procedures with anti-ASC
antibody (sc-514414, Santa Cruz, United States).
2.11 Statistical analysis
Data were analyzed using GraphPad Prism 9 software
(GraphPad Software Inc., United States). The Shapiro-Wilk test
was used for normality, while the Brown-Forsythe test was applied
to test for homogeneity of variances. For multiple comparisons, one-
way analysis of variance (ANOVA) was used, and post hoc pairwise
comparisons were conducted using the Least Signicant Difference
(LSD) method. Data are presented as means ± standard error of the
mean (SEM). A p-value of less than 0.05 was considered statistically
signicant.
3 Results
3.1 Effect of acacetin on
NLRP3 inammasome activation induced
by Nigericin
To investigate whether Acacetin could inhibit the activation of the
NLRP3 inammasome, several methods were conducted. Western blot
analysis was used to quantify the expression levels of pro-caspase-1, pro-
IL-1β, and NLRP3 in the cell lysates, as well as caspase-1 and IL-1βin the
cell supernatant (Figure 1A). ELISA was utilized to measure the
expression of TNF-α, IL-1β, and IL-18 in the supernatant (Figures
1BD), and the LDH assay was conducted to evaluate cell death
(Figure 1E). The results revealed that, compared to the control group,
Nigericin induced the activation of NLRP3 inammasome, leading to a
signicant increase in the expression of caspase-1, IL-1β,andNLRP3in
the supernatant and cell lysates. Intervention with Acacetin effectively
reduced the expression of caspase-1, IL-1β, and NLRP3. In comparison to
the control group, the expression of inammatory cytokines TNF-α,IL-
1β, IL-18, and LDH release was signicantly elevated, while acacetin
inhibited the expression of them. It indicates acacetins inhibition of
Nigericin-induced NLRP3 inammation activation. Among the tested
concentrations, the inhibitory effect was most prominent at a dose of
10 μM. Consequently, this dose was selected for subsequent studies.
3.2 Effect of acacetin on
NLRP3 inammasome activation induced by
multiple agonists
We further investigated the effects of Acacetin on
NLRP3 inammasome activation induced by the canonical
activators MSU, ATP, and Nigericin. The results revealed that
compared to the control group, all three activatorsMSU, ATP,
and Nigericinwere able to induce the activation of
NLRP3 inammasome, leading to an increase in the expression
of caspase-1 and IL-1β(Figure 2A). There was also an elevation in the
release of TNF-α,IL-1β,IL-18,andLDHactivity(Figures 2BE).
Frontiers in Pharmacology frontiersin.org05
Bu et al. 10.3389/fphar.2024.1286546
FIGURE 1
Effect of Acacetin on NLRP3 inammasome activation induced by Nigericin. After pretreatment with LPS (50 ng/mL) for 3 h, BMDMs were treated
with different concentrations of Acacetin (2.5 μM, 5 μM, and 10 μM) for 0.5 h, and then Nigericin (10 μM) was added for stimulation for 0.5 h. The culture
supernatant and cell lysate were collected. (A) Western blot detected IL-1βand caspase-1 in the supernatant, and pro-IL-1β, pro-caspase-1, and NLR P3 in
the cell lysate. (BD) ELISA detected levels of IL-1β(B), IL-18 (C), and TNF-α(D) in the supernatant. (E) LDH activity was assessed. The data is
presented as mean ± SEM (n= 3) and was analyzed with ANOVA followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org06
Bu et al. 10.3389/fphar.2024.1286546
FIGURE 2
Effect of Acacetin on NLRP3 inammasome activation induced by multiple agonists. BMDMs cells were pretreated with LPS (50 ng/mL) for 3 h and
then treated with Acacetin (10 μM) for 0.5 h. After that, the cells were stimulated with MSU (150 μg/mL for 4 h), ATP (2.5 mM for 0.5 h), and Nigericin
(10 μM for 0.5 h). The culture supernatant and cell lysate were collected. (A) Western blot detected IL-1βand caspase-1 in the supernatant, and pro-IL-1β
and pro-caspase-1 in the cell lysate. (BD) ELISA detected the levels of IL-1β(B), IL-18 (C) and TNF-α(D) in the supernatant. (E) LDH activity was
assessed. The data is presented as mean ± SEM (n= 3) and was analyzed with ANOVA followed by LSD *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org07
Bu et al. 10.3389/fphar.2024.1286546
FIGURE 3
Effect of Acacetin on non-canonical NLRP3 inammasome activation. After pretreatment with Pam3CSK4 (400 ng/mL) for 3 h, BMDMs were
treated with acacetin of different concentrations (2.5 μM, 5 μM, and 10 μM) for 0.5 h. Subsequently, the cells were transfected with LPS (500 ng/mL) using
Lipofectamine 3000 for 16 h. The culture supernatant and cell lysate were collected. (A) Western blot detected IL-1βand caspase-1 in the supernatant,
and pro-IL-1βand pro-caspase-1 in the cell lysate. (BD) ELISA detected the levels of IL-1β(B), IL-18 (C) and TNF-α(D) in the supernatant. (E) LDH
activity was also assessed. The data is presented as mean ± SEM (n= 3) and was analyzed with ANOVA followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org08
Bu et al. 10.3389/fphar.2024.1286546
FIGURE 4
Effect of Acacetin on the NF-kB pathway. BMDMs were primed with LPS for different durations (0, 10 min, 30 min, and 60 min) and then treated with
acacetin (10 μM). (A) Western blot was used to detect the relative expression levels of NF-κB p65 (B),p-NF-κB p65 (C),IκBα(D), and p-IκBα(E) in the cells.
The data is presented as mean ± SEM (n = 3) and was analyzed with ANOVA followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org09
Bu et al. 10.3389/fphar.2024.1286546
Remarkably, acacetin demonstrated an inhibitory effect on
NLRP3 inammasome activation by these canonical activators and
decreased the secretion of TNF-α,IL-1β, IL-18, and LDH release.
3.3 Effect of acacetin on non-canonical
NLRP3 inammasome activation
To explore the potential effects of acacetin on non-canonical
NLRP3 inammasome activation, BMDMs were pre-treated with
Pam3CSK4 and then stimulated with LPS. Our ndings
demonstrated that acacetin effectively interrupted the cleavage of
caspase-1 during non-canonical NLRP3 inammasome activation.
This interruption led to a subsequent inhibition of the secretion of
IL-1βand IL-18, along with the release of LDH. Interestingly, the
expression of TNF-αremained unaffected by acacetin under these
conditions. Figure 3 summarizes these results.
3.4 Effect of acacetin on the NF-κB pathway
Western Blot was employed to detect the protein expression
levels of phosphorylated p65 (p-p65), p65, phosphorylated IκBα
(p-IκBα), and IκBα. As shown in Figure 4, the protein expression
levels of NF-κB p65 and IκBαdid not differ in all groups. Compared
to the control group, the expression levels of p-65 and p-IκBαwere
upregulated with the progression of LPS stimulation time. Upon
treatment with acacetin, the expression of both p-65 and p-IκBαwas
markedly reduced.
3.5 Effect of acacetin on nuclear
translocation of NF-κB p65
The results from LSCM revealed that in the blank control group,
NF-κB p65 (green) was broadly distributed within the cytoplasm,
FIGURE 5
Effect of Acacetin on nuclear localization of NF-κB p65. BMDMs were primed with LPS for different durations (0, 10 min, 30 min, and 60 min) and
then treated with acacetin (10 μM). (A) Immunouorescence staining of NF-κB p65 was performed. NF-κB p65 is depicted in gre en, and the cell nuclei are
shown in blue. Scale bar: 50 μm. (B) Quantication of relative uorescence intensity of NF-κB p65. The data is presented as mean ± SEM (n= 3) and was
analyzed with ANOVA followed by LSD. ***p<0.001.
Frontiers in Pharmacology frontiersin.org10
Bu et al. 10.3389/fphar.2024.1286546
with no observable localization within the cell nucleus (blue). Upon
LPS stimulation, a substantial amount of NF-κB p65 was observed to
localize to the cell nucleus, indicating that LPS promotes the
activation and nuclear translocation of NF-κB p65. Following
treatment with acacetin, the levels of LPS-mediated NF-κB
p65 localized to the nucleus were signicantly reduced, indicating
FIGURE 6
Effect of Acacetin on the MAPK pathway. BMDMs were primed with LPS for different durations (0, 10 min, 30 min, and 60 min), then treated with
acacetin (10 μM). (A) Western blot was conducted to detect the relative expressions of ERK (B), p-ERK (C),JNK(D),p-JNK(E), p38 (F), and p-p38 (G) in the
cells. The data is presented as mean ± SEM (n = 3) and was analyzed with ANOVA followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org11
Bu et al. 10.3389/fphar.2024.1286546
that acacetin signicantly inhibits LPS-mediated NF-κB p65 nuclear
translocation. A visual overview of the results is given in Figure 5.
3.6 Effect of acacetin on the mitogen-
activated protein kinase pathway
The MAPK family signaling pathways include p38, ERK, and
JNK. To assess their activity, Western blot analysis was conducted to
measure the expression levels of phosphorylated ERK (p-ERK),
ERK, phosphorylated JNK (p-JNK), JNK, phosphorylated p38
(p-p38), and p38. In the blank control group, the expression
levels of p-ERK, p-JNK, and p-p38 were found to be at a low
level. Upon LPS stimulation, there was an increase in the expression
of these phosphorylated proteins. Acacetin effectively inhibited the
LPS-induced expression of p-ERK, p-JNK, and p-p38, underscoring
its modulatory effect on the MAPK pathway. The results are shown
in Figure 6.
3.7 Effect of acacetin on K+ and Cl- ions
efux during NLRP3 inammasome
activation
Many studies have highlighted the important role of K+ and
Cl-ions in the activation process of the NLRP3 inammasome. In
our investigation, compared to the blank control group, the
expression of intracellular K+ and Cl-ions decreased in the
LPS + Nigericin group. The intervention of acacetin did not
reverse the reduction of K+ and Cl-ions within the cells,
suggesting that acacetin does not affect the efux of K+ and
Cl-ions induced by Nigericin. Therefore, acacetin does not affect
NLRP3 inammasome activation through the efux of K+ and
Cl-ions. See Figure 7 for the results.
3.8 Effect of acacetin on mitochondrial
damage and reactive oxygen species during
NLRP3 inammasome activation
ROS are key upstream signals in the NLRP3 inammasome
activation. We utilized MitoSOX to label ROS and observed their
expression levels in cells using a laser confocal microscope. The
study found compared to the control group, ROS production
increased following Nigericin induction. Intervention with acacetin
subsequently led to reduced ROS production in a dose-dependent
manner. Thus, acacetin may inuence NLRP3 inammasome
activation by modulating ROS production (Figure 8).
3.9 Effect of acacetin on ASC aggregation
during NLRP3 inammasome activation
ASC aggregation is an important hallmark of
NLRP3 inammasome activation. In comparison with the control
group, Nigericin was observed to induce ASC oligomerization.
FIGURE 7
Effect of Acacetin on K+ and Cl- Ions Efux During NLRP3 Inammasome Activation. BMDMs were primed with LPS (50 ng/mL) for 3 h, followed by
treatment with different concentrations of acacetin (2.5 μM, 5 μM, and 10 μM) for 0.5 h, and then stimulated with Nigericin (10 μM) for 0.5 h. The cell
suspension was collected and processed. (A) K+ ions concentration was measured using a potassium ion assay kit (mmol/gprot). (B) Cl-ions
concentration was determined using MQAE uorescent indicator. The data is presented as mean ± SEM (n= 3) and was analyzed with ANOVA
followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org12
Bu et al. 10.3389/fphar.2024.1286546
Treatment with Acacetin led to a dose-dependent reduction in ASC
oligomerization. The outcome is illustrated in Figure 9.
4 Discussion
Acacetin has been recognized for its pharmacological properties,
including antioxidant and anti-inammatory effects, and has
demonstrated therapeutic potential against various diseases in
cellular or animal studies. Multiple studies have found that
acacetin can exert protective effects on cardiovascular diseases by
regulating pathways such as TGF-β/Smad3, MAPK, and PI3K/Akt,
inhibiting the expression of inammatory cytokines, and promoting
the secretion of anti-inammatory factors (Liu et al., 2016;Wu W.-
Y. et al., 2018;Li Z.-Y. et al., 2023). Wu D. et al., 2018 found in
cellular experiments that acacetin can inhibit the production of ROS,
increase the activity of HO-1 and Nrf2, and suppress the expression
of inammatory factors TNF-αand IL-1β, thereby alleviating LPS-
induced lung injury. In vitro studies by Ha et al. (Ha et al., 2012)
showed that acacetin could suppress the expression of NO, iNOS,
FIGURE 8
Effect of Acacetin on Mitochondrial Damage and ROS During NLRP3 Inammasome Activation. BMDMs were primed with LPS (50 ng/mL) for 3 h,
followed by treatment with different concentrations of acacetin (2.5 μM, 5 μM, and 10 μM) for 0.5 h, and then stimulated with Nigericin (10 μM) for 0.5 h.
The cells were then incubated with MitoSOX Red Indicator (5 μM) to detect the levels of mitochondrial ROS. The images (A) were acquired using laser
scanning confocal microscopy and the bar plot (B) was quantitated by uorescen ce intensity. Scale bar: 20 μm. The data is presented as mean ± SEM
(n= 3) and was analyzed with ANOVA followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org13
Bu et al. 10.3389/fphar.2024.1286546
PGE2, COX2, TNF-α, and IL-1βin LPS-stimulated BV2 cells.
Moreover, acacetin has been found to inhibit 6-OHDA-induced
neuronal death and ROS-mediated apoptotic cascades, thereby
exerting protective effects against Parkinsons Disease (PD) (Kim
et al., 2017). Wang et al. (Wang et al., 2015) discovered that acacetin
could mediate the transcriptional regulation of APP and BACE-1,
downregulate their protein expression, reduce Aβformation, and
also inhibit APP synthesis, thus lessening the formation of senile
plaques and providing protection against Alzheimers Disease (AD).
Although these studies conrm the anti-inammatory effect of
acacetin, the specic mechanisms remain to be further elucidated,
and it is still unclear whether acacetin could become a candidate
drug for inhibiting the NLRP3 inammasome. In this study, we
found that acacetin inhibits the activation of the
NLRP3 inammasome induced by canonical activators such as
MSU, ATP, and Nigericin. Acacetin is able to suppress caspase-1
cleavage, inhibit the secretion of IL-1βand IL-18, reduce LDH
activity, and furthermore, it can also inhibit non-classical
NLRP3 inammasome activation.
The activation of the NLRP3 inammasome involves two
stepspriming step and activation step. The priming step mainly
activates the NF-κB pathway, thereby upregulating the expression of
NLRP3 and Pro-IL-1β. The activation step primarily triggers the
assembly of the NLRP3 inammasome, leading to the secretion of
IL-1βand IL-18 mediated by caspase-1, as well as pyroptosis
(Bauernfeind et al., 2009;Swanson et al., 2019). LPS, a major
outer membrane component of Gram-negative bacteria, binds to
toll-like receptor 4 (TLR4), activates the MAPK pathway, and
FIGURE 9
Effect of Acacetin on ASC Aggregation During NLRP3 Inammasome Activation. BMDMs were primed with LPS (50 ng/mL) for 3 h, followed by
treatment with different concentrations of acacetin (2.5 μM, 5 μM, and 10 μM) for 0.5 h, and then stimulated with Nigericin (10 μM) for 0.5 h. The cells
were lysed with pellets crosslinked with DSS to detect ASC aggregation. (A) Western blot was performed to detect the relative expression of Polymers (B),
Dimers (C), and ASC in pellets (D) in the cells, with ASC in INPUT serving as the loading control. The data is presented as mean ± SEM (n = 3) and was
analyzed with ANOVA followed by LSD. *p<0.05, **p<0.01, ***p<0.001.
Frontiers in Pharmacology frontiersin.org14
Bu et al. 10.3389/fphar.2024.1286546
induces an NF-κB-dependent inammatory cascade, resulting in the
overproduction of pro-inammatory mediators such as nitric oxide
(NO) and prostaglandin E2 (PGE2) as well as cytokines like TNF-α
and IL-6 (Zeng et al., 2017).
Here, we explored the role of acacetin during the
NLRP3 inammasome priming step by isolating and inducing
mouse BMDMs with LPS. We found that LPS increased TNF-α
secretion and NLRP3 expression in BMDMs. While acacetin could
inhibit TNF-αsecretion and reduce NLRP3 expression, indicating its
inuence on the priming of NLRP3 inammasome activation. Several
NLRP3 inammasome inhibitors can regulate the priming process.
Compounds such as curcumin (Hasanzadeh et al., 2020), resveratrol
(Liu et al., 2019), green tea polyphenols (Wang et al., 2019), BAY11-
7082 (Irrera et al., 2017), and INF-39 (Shi et al., 2021) have all shown
inhibitory effects on the NF-κB pathway. Chloroquine (Chen et al.,
2017), Sinigrin (Lee et al., 2017), and ECGG (Luo et al., 2021)can
inhibit both NF-κB and MAPK pathways, thereby regulating the
transcriptional levels of NLRP3 protein components and modulating
NLRP3 inammasome activation. Whether acacetin plays an
inhibitory role during the priming is not yet clear, and whether it
inhibits NLRP3 inammasome activation by suppressing the NF-κB
and MAPK pathways remains unreported. In our study, we
discovered that acacetin can inhibit the expression of p-65 and
p-IκBα, suppress nuclear translocation of NF-κB, and inhibit the
expression of p-ERK, p-JNK, and p-P38, thus modulating the
priming process.
Following the priming step, the activation step of the
NLRP3 inammasome is marked by pivotal roles of ASC
aggregation, ROS production, K+ and Cl-ions efux. Various
inhibitors of NLRP3, such as MNS (He et al., 2014), NBC6
(Baldwin et al., 2017), and BOT-4-one (Shim et al., 2017), have
been shown to prevent the formation of ASC specks during the
activation of the NLRP3 inammasome. Youm et al. (He et al.,
2018) demonstrated that β-hydroxybutyrate (BHB) could
specically inhibit NLRP3 inammasome activation by
suppressing K+ ions efux. Similarly, MCC950 was found to
attenuate Cl-ions efux during NLRP3 inammasome activation
(Jiang et al., 2017). Yang et al., 2016 pinpointed that sulforaphane
can impede NLRP3 inammasome activation via inhibiting
unsaturated fatty acid-induced ROS production through the
AMP-activated protein kinase (AMPK) autophagy pathway. In
ourpreviousstudies(Bu et al., 2019;2022;2023), we identied that
acacetin could diminish ROS level in microglial cells post-oxygen
glucose deprivation (OGD) injury. However, whether acacetin
affects ASC aggregation, as well as the efux of K+ and Cl-ions,
thereby modulating NLRP3 inammasome activation, remains
unreported. In this study, we observed that acacetin inhibited
ASC aggregation and ROS production following
NLRP3 inammasome activation but did not alter the efux of
K+ and Cl-ions during the activation step.
In summary, this study has unveiled that acacetin can inhibit the
activation of the NLRP3 inammasome. Specically, acacetin
modulates both the priming and assembly of NLRP3 inammasome
activation by suppressing the NF-κBandMAPKpathways,
subsequently hindering ASC aggregation and ROS production.
While acacetin displays inhibitory effects on NLRP3 inammasome
activation, further studies are warranted to determine its protective role
against diseases mediated by the NLRP3 inammasome.
Data availability statement
The original contributions presented in the study are included in
the article/Supplementary material, further inquiries can be directed
to the corresponding authors.
Ethics statement
The animal study was approved by the Ethics Committee of
Peoples Hospital of Xinjiang Uygur Autonomous Region. The study
was conducted in accordance with the local legislation and
institutional requirements.
Author contributions
JB: Conceptualization, Data curation, Funding acquisition,
Investigation, Project administration, Writingoriginal draft,
Writingreview and editing. YM: Data curation, Formal Analysis,
Funding acquisition, Writingreview and editing, Writingoriginal
draft. SZ: Formal Analysis, Investigation, Writingreview and
editing. XW: Formal Analysis, Investigation, Writingreview and
editing. XZ: Formal Analysis, Investigation, Writingreview and
editing. LZ: Conceptualization, Data curation, Project
administration, Supervision, Writingreview and editing. YZ:
Supervision, Writingreview and editing.
Funding
The author(s) declare nancial support was received for the
research, authorship, and/or publication of this article. This research
was funded by grants from Tianshan Youth Projectof Program for
Fostering Excellent Young Talents in Science and Technology of
Xinjiang Uygur Autonomous Region (Grant No. 2020Q046),
Natural Science Foundation of Xinjiang Uygur Autonomous
Region (Grant No. 2020D01C095), and Program for Fostering
Excellent Young Talents in Science and Technology of Urumqi,
China to JB and Foundation of Peoples Hospital of Xinjiang Uygur
Autonomous Region (Grant No. 20210110) to YM
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Frontiers in Pharmacology frontiersin.org15
Bu et al. 10.3389/fphar.2024.1286546
References
Baldwin, A. G., Rivers-Auty, J., Daniels, M. J. D., White, C. S., Schwalbe, C. H.,
Schilling, T., et al. (2017). Boron-based inhibitors of the NLRP3 inammasome. Cell
Chem. Biol. 24, 13211335. doi:10.1016/j.chembiol.2017.08.011
Bauernfeind, F. G., Horvath, G., Stutz, A., Alnemri, E. S., MacDonald, K., Speert, D.,
et al. (2009). Cutting edge: NF-kappaB activating pattern recognition and cytokine
receptors license NLRP3 inammasome activation by regulating NLRP3 expression.
J. Immunol. 183, 787791. doi:10.4049/jimmunol.0901363
Bu, J., Ji, G., Mahan, Y., Wang, Z., Wu, X., and Niu, X. (2023). Acacetin protects
against cerebral ischemia-reperfusion injury by regulating ROS/NLRP3 signaling
pathway via autophagy. J. Apoplexy Nerv. Dis. 40, 99102. doi:10.19845/j.cnki.
zfysjjbzz.2023.0025
Bu, J., Shi, S., Wang, H.-Q., Niu, X.-S., Zhao, Z.-F., Wu, W.-D., et al. (2019). Acacetin
protects against cerebral ischemia-reperfusion injury via the NLRP3 signaling pathway.
Neural Regen. Res. 14, 605612. doi:10.4103/1673-5374.247465
Bu,J.,Zhang,Y.,Mahan,Y.,Shi,S.,Wu,X.,Zhang,X.,etal.(2022).Acacetinimproves
cognitive function of APP/PS1 Alzheimers disease model mice via the NLRP3 inammasome
signaling pathway. Transl. Neurosci. 13, 390397. doi:10.1515/tnsci-2022-0254
Chen,X.,Wang,N.,Zhu,Y.,Lu,Y.,Liu,X.,andZheng,J.(2017).Theantimalarial
chloroquine suppresses LPS-induced NLRP3 inammasome activation and confers protection
against murine endotoxic shock. Mediat. Inamm. 2017, 6543237. doi:10.1155/2017/6543237
Gastaldi, S., Rocca, C., Gianquinto, E., Granieri, M. C., Boscaro, V., Blua, F., et al.
(2023). Discovery of a novel 1,3,4-oxadiazol-2-one-based NLRP3 inhibitor as a
pharmacological agent to mitigate cardiac and metabolic complications in an
experimental model of diet-induced metaammation. Eur. J. Med. Chem. 257,
115542. doi:10.1016/j.ejmech.2023.115542
Ha, S. K., Moon, E., Lee, P., Ryu, J. H., Oh, M. S., and Kim, S. Y. (2012). Acacetin
attenuates neuroinammation via regulation the response to LPS stimuli in vitro and in
vivo.Neurochem. Res. 37, 15601567. doi:10.1007/s11064-012-0751-z
Han, Q.-Q., and Le, W. (2023). NLRP3 inammasome-mediated neuroinammation
and related mitochondrial impairment in Parkinsons disease. Neurosci. Bull. 39,
832844. doi:10.1007/s12264-023-01023-y
Hasanzadeh, S., Read, M. I., Bland, A. R., Majeed, M., Jamialahmadi, T., and Sahebkar,
A. (2020). Curcumin: an inammasome silencer. Pharmacol. Res. 159, 104921. doi:10.
1016/j.phrs.2020.104921
He, H., Jiang, H., Chen, Y., Ye, J., Wang, A., Wang, C., et al. (2018). Oridonin is a
covalent NLRP3 inhibitor with strong anti-inammasome activity. Nat. Commun. 9,
2550. doi:10.1038/s41467-018-04947-6
He, W., Qu, T., Yu, Q., Wang, Z., Lv, H., Zhang, J., et al. (2013). LPS induces IL-8
expression through TLR4, MyD88, NF-kappaB and MAPK pathways in human dental
pulp stem cells. Int. Endod. J. 46, 128136. doi:10.1111/j.1365-2591.2012.02096.x
He, Y., Varadarajan, S., Muñoz-Planillo, R., Burberry, A., Nakamura, Y., and Núñez,
G. (2014). 3,4-methylenedioxy-β-nitrostyrene inhibits NLRP3 inammasome
activation by blocking assembly of the inammasome. J. Biol. Chem. 289,
11421150. doi:10.1074/jbc.M113.515080
Irrera, N., Vaccaro, M., Bitto, A., Pallio, G., Pizzino, G., Lentini, M., et al. (2017). BAY
11-7082 inhibits the NF-κB and NLRP3 inammasome pathways and protects against
IMQ-induced psoriasis. Clin. Sci. (Lond) 131, 487498. doi:10.1042/CS20160645
Jiang, H., He, H., Chen, Y., Huang, W., Cheng, J., Ye, J., et al. (2017). Identication of a
selective and direct NLRP3 inhibitor to treat inammatory disorders. J. Exp. Med. 214,
32193238. doi:10.1084/jem.20171419
Kim,S.M.,Park,Y.J.,Shin,M.-S.,Kim,H.-R.,Kim,M.J.,Lee,S.H.,etal.(2017).Acacetin
inhibits neuronal cell death induced by 6-hydroxydopamine in cellular Parkinsonsdisease
model. Bioorg Med. Chem. Lett. 27, 52075212. doi:10.1016/j.bmcl.2017.10.048
Kiser, C., Gonul, C. P., Olcum, M., and Genc, S. (2021). Inhibitory effects of
sulforaphane on NLRP3 inammasome activation. Mol. Immunol. 140, 175185.
doi:10.1016/j.molimm.2021.10.014
Lee, H.-W., Lee, C. G., Rhee, D.-K., Um, S. H., and Pyo, S. (2017). Sinigrin inhibits
production of inammatory mediators by suppressing NF-κB/MAPK pathways or
NLRP3 inammasome activation in macrophages. Int. Immunopharmacol. 45,
163173. doi:10.1016/j.intimp.2017.01.032
Li, W., Xu, H., Shao, J., Chen, J., Lin, Y., Zheng, Z., et al. (2023a). Discovery of
alantolactone as a naturally occurring NLRP3 inhibitor to alleviate NLRP3-driven
inammatory diseases in mice. Br. J. Pharmacol. 180, 16341647. doi:10.1111/bph.16036
Li, Z.-Y., Lv, S., Qiao, J., Wang, S.-Q., Ji, F., Li, D., et al. (2023b). Acacetin alleviates
cardiac brosis via TGF-β1/Smad and AKT/mTOR signal pathways in spontaneous
hypertensive rats. Gerontology 69, 10761094. doi:10.1159/000531596
Liu, H., Yang, L., Wu, H.-J., Chen, K.-H., Lin, F., Li, G., et al. (2016). Water-soluble
acacetin prodrug confers signicant cardioprotection against ischemia/reperfusion
injury. Sci. Rep. 6, 36435. doi:10.1038/srep36435
Liu, T., Ma, Y., Zhang, R., Zhong, H., Wang, L., Zhao, J., et al. (2019). Resveratrol
ameliorates estrogen deciency-induced depression- and anxiety-like behaviors and
hippocampal inammation in mice. Psychopharmacology 236, 13851399. doi:10.1007/
s00213-018-5148-5
Luo, Y., Lu, J., Ruan, W., Guo, X., and Chen, S. (2019). MCC950 attenuated early brain
injury by suppressing NLRP3 inammasome after experimental SAH in rats. Brain Res.
Bull. 146, 320326. doi:10.1016/j.brainresbull.2019.01.027
Luo, Z.-L., Sun, H.-Y., Wu, X.-B., Cheng, L., and Ren, J.-D. (2021). Epigallocatechin-
3-gallate attenuates acute pancreatitis induced lung injury by targeting mitochondrial
reactive oxygen species triggered NLRP3 inammasome activation. Food Funct. 12,
56585667. doi:10.1039/d1fo01154e
Lv, J., Shen, X., Shen, X., Zhao, S., Xu, R., Yan, Q., et al. (2023). NPLC0393 from
Gynostemma pentaphyllum ameliorates Alzheimers disease-like pathology in mice by
targeting protein phosphatase magnesium-dependent 1A phosphatase. Phytother. Res.
37, 47714790. doi:10.1002/ptr.7945
Ma, Q. (2023). Pharmacological inhibition of the NLRP3 inammasome: structure,
molecular activation, and inhibitor-NLRP3 interaction. Pharmacol. Rev. 75, 487520.
doi:10.1124/pharmrev.122.000629
Qin, Y., and Zhao, W. (2023). Posttranslational modications of NLRP3 and their
regulatory roles in inammasome activation. Eur. J. Immunol. 53, e2350382. doi:10.
1002/eji.202350382
Robblee, M. M., Kim, C. C., Porter Abate, J., Valdearcos, M., Sandlund, K. L. M.,
Shenoy, M. K., et al. (2016). Saturated fatty acids engage an IRE1α-dependent pathway
to activate the NLRP3 inammasome in myeloid cells. Cell Rep. 14, 26112623. doi:10.
1016/j.celrep.2016.02.053
Shi, Y., Lv, Q., Zheng, M., Sun, H., and Shi, F. (2021). NLRP3 inammasome inhibitor
INF39 attenuated NLRP3 assembly in macrophages. Int. Immunopharmacol. 92,
107358. doi:10.1016/j.intimp.2020.107358
Shim, D.-W., Shin, W.-Y., Yu, S.-H., Kim, B.-H., Ye, S.-K., Koppula, S., et al. (2017).
BOT-4-one attenuates NLRP3 inammasome activation: NLRP3 alkylation leading to
the regulation of its ATPase activity and ubiquitination. Sci. Rep. 7, 15020. doi:10.1038/
s41598-017-15314-8
Singh, S., Gupta, P., Meena, A., and Luqman, S. (2020). Acacetin, a avone with
diverse therapeutic potential in cancer, inammation, infections and other metabolic
disorders. Food Chem. Toxicol. 145, 111708. doi:10.1016/j.fct.2020.111708
Swanson, K. V., Deng, M., and Ting, J. P.-Y. (2019). The NLRP3 inammasome:
molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 19, 477489.
doi:10.1038/s41577-019-0165-0
Wang,D.,Zhang,M.,Wang,T.,Cai,M.,Qian,F.,Sun,Y.,etal.(2019).Greenteapolyphenols
prevent lipopolysaccharide-induced inammatory liver injury in mice by inhibiting
NLRP3 inammasome activation. Food Funct. 10, 38983908. doi:10.1039/C9FO00572B
Wang, H.-Y., Lin, X., Huang, G.-G., Zhou, R., Lei, S.-Y., Ren, J., et al. (2023).
Atranorin inhibits NLRP3 inammasome activation by targeting ASC and protects
NLRP3 inammasome-driven diseases. Acta Pharmacol. Sin. 44, 16871700. doi:10.
1038/s41401-023-01054-1
Wang,X.,Perumalsamy,H.,Kwon,H.W.,Na,Y.-E.,andAhn,Y.-J.(2015).
Effects and possible mechanisms of action of acacetin on the behavior and eye
morphology of Drosophila models of Alzheimersdisease.Sci. Rep. 5, 16127.
doi:10.1038/srep16127
Wang, Y., Liu, Y.-J., Zhang, M.-M., Zhou, H., Gao, Y.-H., Cheng, W.-J., et al. (2022).
CY-09 alleviates the depression-like behaviors via inhibiting NLRP3 inammasome-
mediated neuroinammation in lipopolysaccharide-induced mice. ACS Chem.
Neurosci. 13, 32913302. doi:10.1021/acschemneuro.2c00348
Wu, D., Wang, Y., Zhang, H., Du, M., and Li, T. (2018a). Acacetin attenuates mice
endotoxin-induced acute lung injury via augmentation of heme oxygenase-1 activity.
Inammopharmacol 26, 635643. doi:10.1007/s10787-017-0398-0
Wu, W.-Y., Li, Y.-D., Cui, Y.-K., Wu, C., Hong, Y.-X., Li, G., et al. (2018b). The
natural avone acacetin confers cardiomyocyte protection against hypoxia/
reoxygenation injury via AMPK-mediated activation of Nrf2 signaling pathway.
Front. Pharmacol. 9, 497. doi:10.3389/fphar.2018.00497
Xu, H.-W., Li, W.-F., Hong, S.-S., Shao, J.-J., Chen, J.-H., Chattipakorn, N., et al.
(2023a). Tabersonine, a natural NLRP3 inhibitor, suppresses inammasome activation
in macrophages and attenuate NLRP3-driven diseases in mice. Acta Pharmacol. Sin. 44,
12521261. doi:10.1038/s41401-022-01040-z
Xu, Y., Biby, S., Kaur, B., and Zhang, S. (2023b). A patent review of NLRP3 inhibitors
to treat autoimmune diseases. Expert Opin. Ther. Pat. 33, 455470. doi:10.1080/
13543776.2023.2239502
Yang, G., Lee, H. E., and Lee, J. Y. (2016). A pharmacological inhibitor of
NLRP3 inammasome prevents non-alcoholic fatty liver disease in a mouse model
induced by high fat diet. Sci. Rep. 6, 24399. doi:10.1038/srep24399
Zeng,J.,Chen,Y.,Ding,R.,Feng,L.,Fu,Z.,Yang,S.,etal.(2017).Isoliquiritigeninalleviates
early brain injury after experimental intracerebral hemorrhage via suppressing ROS- and/or
NF-κB-mediated NLRP3 inammasome activation by promoting Nrf2 antioxidant pathway.
J. Neuroinammation 14, 119. doi:10.1186/s12974-017-0895-5
Zhou, Y., Yang, Z., Ou, Y., Cai, H., Liu, Z., Lin, G., et al. (2023). Discovery of a selective
NLRP3-targeting compound with therapeutic activity in MSU-induced peritonitis and
DSS-induced acute intestinal inammation. Cell Mol. Life Sci. 80, 230. doi:10.1007/
s00018-023-04881-x
Frontiers in Pharmacology frontiersin.org16
Bu et al. 10.3389/fphar.2024.1286546
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The aberrant activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is known to contribute to the pathogenesis of various human inflammation-related diseases. However, to date, no small-molecule NLRP3 inhibitor has been used in clinical settings. In this study, we have identified SB-222200 as a novel direct NLRP3 inhibitor through the use of drug affinity responsive target stability assay, cellular thermal shift assay, and surface plasmon resonance analysis. SB-222200 effectively inhibits the activation of the NLRP3 inflammasome in macrophages, while having no impact on the activation of NLRC4 or AIM2 inflammasome. Furthermore, SB-222200 directly binds to the NLRP3 protein, inhibiting NLRP3 inflammasome assembly by blocking the NEK7 − NLRP3 interaction and NLRP3 oligomerization. Importantly, treatment with SB-222200 demonstrates alleviation of NLRP3-dependent inflammatory diseases in mouse models, such as monosodium urate crystal-induced peritonitis and dextran sulfate sodium-induced acute intestinal inflammation. Therefore, SB-222200 holds promise as a lead compound for the development of NLRP3 inhibitors to combat NLRP3-driven disease and serves as a versatile tool for pharmacologically investigating NLRP3 biology.
Article
Full-text available
The NACHT, LRR, and PYD domains‐containing protein 3 (NLRP3) inflammasome is a multimolecular complex that plays a fundamental role in inflammation. Optimal activation of NLRP3 inflammasome is crucial for host defense against pathogens and the maintenance of immune homeostasis. Aberrant NLRP3 inflammasome activity has been implicated in various inflammatory diseases. Posttranslational modifications (PTMs) of NLRP3, a key inflammasome sensor, play critical roles in directing inflammasome activation and controlling the severity of inflammation and inflammatory diseases, such as arthritis, peritonitis, inflammatory bowel disease, atherosclerosis, and Parkinson's disease. Various NLRP3 PTMs, including phosphorylation, ubiquitination, and SUMOylation, could direct inflammasome activation and control inflammation severity by affecting the protein stability, ATPase activity, subcellular localization, and oligomerization of NLRP3 as well as the association between NLRP3 and other inflammasome components. Here, we provide an overview of the PTMs of NLRP3 and their roles in controlling inflammation and summarize potential anti‐inflammatory drugs targeting NLRP3 PTMs.
Article
Full-text available
Parkinson's disease (PD) is a common neurodegenerative disorder caused by the loss of dopamine neurons in the substantia nigra and the formation of Lewy bodies, which are mainly composed of alpha-synuclein fibrils. Alpha-synuclein plays a vital role in the neuroinflammation mediated by the nucleotide-binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome in PD. A better understanding of the NLRP3 inflammasome-mediated neuroinflammation and the related mitochondrial impairment during PD progression may facilitate the development of promising therapies for PD. This review focuses on the molecular mechanisms underlying NLRP3 inflammasome activation, comprising priming and protein complex assembly, as well as the role of mitochondrial impairment and its subsequent inflammatory effects on the progression of neurodegeneration in PD. In addition, the therapeutic strategies targeting the NLRP3 inflammasome for PD treatment are discussed, including the inhibitors of NLRP3 inflammatory pathways, mitochondria-focused treatments, microRNAs, and other therapeutic compounds.
Article
Full-text available
The nucleotide-binding, oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is a multiprotein complex that combines sensing, regulation, and effector functions to regulate inflammation in health and disease. NLRP3 is activated by a diverse range of inflammation-instigating signals known as pathogen associated molecular patterns and danger associated molecular patterns. Upon activation, NLRP3 oligomerizes and recruits partner proteins to form a supramolecular platform to process the maturation and release of interleukin (IL)-1β, IL-18, and gasdermin D, major mediators of inflammation and inflammatory cell death termed pyroptosis. The NLRP3 inflammasome has been implicated in the pathogenesis of a wide range of disease conditions, including chronic inflammatory disease that are associated with lifestyle and dietary changes, aging, and environmental exposures, and have become the leading cause of death worldwide. Pharmacological targeting of NLRP3 and signaling demonstrated promising efficacy in ameliorating a list of disease conditions in animal models. These findings underscore the potential and importance of NLRP3 as a druggable target for treating a range of diseases. In this review, recent progress in understanding the structure and mechanism of action of the NLRP3 inflammasome is discussed with focus on pharmacological inhibition of NLRP3 by small molecule inhibitors. New structural and mechanistic insights into NLRP3 activation and inhibitor-NLRP3 interactions would aid in the rational design and pharmacological evaluation of NLRP3 inhibitors for treatment of human disease. SIGNIFICANCE STATEMENT: The NLRP3 inflammasome plays central role in innate immune sensing and control of inflammation. Pharmacological inhibition of NLRP3 demonstrated promising efficacy in a range of diseases in animal models. Recent elucidation of the structure and inhibitor binding of NLRP3 generated new insights into its mode of action and inhibitor-NLRP3 interaction at molecular levels, providing new framework for developing small chemical inhibitors of NLRP3 with improved efficacy and specificity against chronic disease that has become major health concerns worldwide.
Article
Full-text available
Background and Purpose The NLR family pyrin domain‐containing 3 (NLRP3) inflammasome is activated in many inflammatory conditions. So far, no low MW compounds inhibiting NLRP3 have entered clinical use. Identification of naturally occurring NLRP3 inhibitors may be beneficial to the design and development of compounds targeting NLRP3. Alantolactone is a phytochemical from a traditional Chinese medicinal plant with anti‐inflammatory activity, but its precise target remains unclear. Experimental Approach A bank of phytochemicals was screened for inhibitors of NLRP3‐driven production of IL‐1β in cultures of bone‐marrow‐derived macrophages from female C57BL/6 mice. Models of gouty arthritis and acute lung injury in male C57BL/6J mice were used to determine the in vivo effects of the most potent compound. Key Results Among the 150 compounds screened in vitro, alantolactone exhibited the highest inhibitory activity against LPS + ATP‐induced production of IL‐1β in macrophages, suppressing IL‐1β secretion, caspase‐1 activation and pyroptosis. Alantolactone directly bound to the NACHT domain of NLRP3 to inhibit activation and assembly of NLRP3 inflammasomes. Molecular simulation analysis suggested that Arg335 in NLRP3 was a critical residue for alantolactone binding, leading to suppression of NLRP3–NEK7 interaction. In vivo studies confirmed significant alleviation by alantolactone of two NLRP3‐driven inflammatory conditions, acute lung injury and gouty arthritis. Conclusion and Implications The phytochemical alantolactone inhibited activity of NLRP3 inflammasomes by directly targeting the NACHT domain of NLRP3. Alantolactone shows great potential in the treatment of NLRP3‐driven diseases and could lead to the development of novel NLRP3 inhibitors.
Article
Introduction: NOD-like receptor family pyrin domain containing 3 (NLRP3) can sense a plethora of exogenous and endogenous dangers. Upon activation, a multimeric protein complex, the NLRP3 inflammasome, is formed to initiate the innate immune responses. Emerging studies have implicated the pathophysiological roles of this protein complex in human disorders, highlighting that it represents a druggable target for therapeutics development. Areas covered: The current review summarizes the functional facets of the NLRP3 inflammasome, its association with autoimmune diseases, and recent patents on the development of NLRP3 inhibitors. Literature search was conducted using SciFinder and Google Patents with the key word NLRP3 and NLRP3 inhibitors. Expert opinion: Although significant advances have been made in understanding the NLRP3 inflammasome, more studies are still needed to elucidate the molecular mechanisms underlying its roles in autoimmune diseases. A number of NLRP3 inhibitors have been patented, however, none of them have been approved for clinical use. Due to the complex nature of the NLRP3 inflammasome, novel screening assays along with target engagement methods could benefit the drug discovery and clinical translation. In addition, clinical trials on NLRP3 inhibitors are still in their early stages, and continuous investigations are needed to fully assess their safety and effectiveness.
Article
Alzheimer's disease (AD) is a neurodegenerative disease with clinical hallmarks of progressive cognitive impairment and memory loss. Gynostemma pentaphyllum ameliorates cognitive impairment, but the mechanisms remain obscure. Here, we determine the effect of triterpene saponin NPLC0393 from G. pentaphyllum on AD-like pathology in 3×Tg-AD mice and elucidate the underlying mechanisms. NPLC0393 was administered daily in vivo by intraperitoneal injection for 3 months and its amelioration on the cognitive impairment in 3×Tg-AD mice was assessed by new object recognition (NOR), Y-maze, Morris water maze (MWM), and elevated plus-maze (EPM) tests. The mechanisms were investigated by RT-PCR, western blot, and immunohistochemistry techniques, while verified by the 3×Tg-AD mice with protein phosphatase magnesium-dependent 1A (PPM1A) knockdown (KD) through brain-specific injection of adeno-associated virus (AAV)-ePHP-KD-PPM1A. NPLC0393 ameliorated AD-like pathology targeting PPM1A. It repressed microglial NLRP3 inflammasome activation by reducing NLRP3 transcription during priming and promoting PPM1A binding to NLRP3 to disrupt NLRP3 assembly with apoptosis-associated speck-like protein containing a CARD and pro-caspase-1. Moreover, NPLC0393 suppressed tauopathy by inhibiting tau hyperphosphorylation through PPM1A/NLRP3/tau axis and promoting microglial phagocytosis of tau oligomers through PPM1A/nuclear factor-κB/CX3CR1 pathway. PPM1A mediates microglia/neurons crosstalk in AD pathology, whose activation by NPLC0393 represents a promising therapeutic strategy for AD.
Article
Introduction: Attenuation of the activation of cardiac fibroblasts contributes to reduce the excessive extracellular matrix deposition and cardiac structural remodeling in hypertensive hearts. Acacetin has shown to play a protective role in doxorubicin-induced cardiomyopathy. The aim of this study was to investigate the potential molecular mechanisms underlying the protective role of acacetin on hypertension-induced fibrosis in the heart. Methods: Echocardiography, pathological morphological and western-blotting techniques were used to evaluate the anti-fibrosis effects in spontaneous hypertensive rat (SHR) which were daily intragastric administrated with acacetin (10mg/kg and 20mg/kg) for 6 weeks. Angiotensin II (Ang II) was used to induce cellular fibrosis in human cardiac fibroblasts in the absence and presence of acacetin treatment for 48 hours. Results: Acacetin alleviated hypertension-induced left ventricular posterior wall thickness and relieved cardiac remodeling in SHR. The expressions of collagen-1, collagen-III and alpha-SMA were remarkedly decreased after treatment with acacetin (n=6, P<0.05). The wound closure rate and the number of migratory cells per field were decreased by 3 and 10 µmol/L acacetin in Ang II-treated fibroblasts (n=6, P < 0.05). Acacetin (10µmol/L) inhibited Ang II-induced upregulation of collagen-1 and collagen-III (n=6, P < 0.05), and downregulation of the expression of alpha-SMA reversed the differentiation of fibroblasts to myofibroblasts. Acacetin also decreased the expression of TGF-β1, the fraction of p-Smad3/Smad3, and p-AKT and p-mTOR, but increased the expression of Smad7 (n=6, P < 0.05). Furthermore, acacetin inhibited TGF-β1 agonist SRI and AKT agonist SC79 caused fibrotic effect. Conclusion: In summary, acacetin inhibits the hypertension-associated cardiac fibrotic processes through regulating TGF-β/Smad3, AKT/mTOR signal transduction pathways.
Article
Inspired by the recent advancements in understanding the binding mode of sulfonylurea-based NLRP3 inhibitors to the NLRP3 sensor protein, we developed new NLRP3 inhibitors by replacing the central sulfonylurea moiety with different heterocycles. Computational studies evidenced that some of the designed compounds were able to maintain important interaction within the NACHT domain of the target protein similarly to the most active sulfonylurea-based NLRP3 inhibitors. Among the studied compounds, the 1,3,4-oxadiazol-2-one derivative 5 (INF200) showed the most promising results being able to prevent NLRP3-dependent pyroptosis triggered by LPS/ATP and LPS/MSU by 66.3 ± 6.6% and 61.6 ± 11.5% and to reduce IL-1β release (35.5 ± 8.8% μM) at 10 μM in human macrophages. The selected compound INF200 (20 mg/kg/day) was then tested in an in vivo rat model of high-fat diet (HFD)-induced metaflammation to evaluate its beneficial cardiometabolic effects. INF200 significantly counteracted HFD-dependent "anthropometric" changes, improved glucose and lipid profiles, and attenuated systemic inflammation and biomarkers of cardiac dysfunction (particularly BNP). Hemodynamic evaluation on Langendorff model indicate that INF200 limited myocardial damage-dependent ischemia/reperfusion injury (IRI) by improving post-ischemic systolic recovery and attenuating cardiac contracture, infarct size, and LDH release, thus reversing the exacerbation of obesity-associated damage. Mechanistically, in post-ischemic hearts, IFN200 reduced IRI-dependent NLRP3 activation, inflammation, and oxidative stress. These results highlight the potential of the novel NLRP3 inhibitor, INF200, and its ability to reverse the unfavorable cardio-metabolic dysfunction associated with obesity.
Article
Aberrant NLRP3 activation has been implicated in the pathogenesis of numerous inflammation-associated diseases. However, no small molecular inhibitor that directly targets NLRP3 inflammasome has been approved so far. In this study, we show that Atranorin (C19H18O8), the secondary metabolites of lichen family, effectively prevents NLRP3 inflammasome activation in macrophages and dendritic cells. Mechanistically, Atranorin inhibits NLRP3 activation induced cytokine secretion and cell pyroptosis through binding to ASC protein directly and therefore restraining ASC oligomerization. The pharmacological effect of Atranorin is evaluated in NLRP3 inflammasome-driven disease models. Atranorin lowers serum IL-1β and IL-18 levels in LPS induced mice acute inflammation model. Also, Atranorin protects against MSU crystal induced mice gouty arthritis model and lowers ankle IL-1β level. Moreover, Atranorin ameliorates intestinal inflammation and epithelial barrier dysfunction in DSS induced mice ulcerative colitis and inhibits NLRP3 inflammasome activation in colon. Altogether, our study identifies Atranorin as a novel NLRP3 inhibitor that targets ASC protein and highlights the potential therapeutic effects of Atranorin in NLRP3 inflammasome-driven diseases including acute inflammation, gouty arthritis and ulcerative colitis.