Accepted Manuscript Chrysanthemum indicum extract inhibits NLRP3 and AIM2 inflammasome activation via regulating ASC phosphorylation Sang-Hyeun Yu, Xiao Sun, Myong-Ki Kim, Mahbuba Akther, Jun-Hyuk Han, TaeYeon Kim, Jun Jiang, Tae-Bong Kang, Kwang-Ho Lee PII:
S0378-8741(19)30342-3
DOI:
https://doi.org/10.1016/j.jep.2019.111917
Article Number: 111917 Reference:
JEP 111917
To appear in:
Journal of Ethnopharmacology
Received Date: 24 January 2019 Revised Date:
22 April 2019
Accepted Date: 23 April 2019
Please cite this article as: Yu, S.-H., Sun, X., Kim, M.-K., Akther, M., Han, J.-H., Kim, T.-Y., Jiang, J., Kang, T.-B., Lee, K.-H., Chrysanthemum indicum extract inhibits NLRP3 and AIM2 inflammasome activation via regulating ASC phosphorylation, Journal of Ethnopharmacology (2019), doi: https:// doi.org/10.1016/j.jep.2019.111917. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT 1
Chrysanthemum indicum extract inhibits NLRP3 and AIM2 inflammasome activation
2
via regulating ASC phosphorylation
3
Sang-Hyeun Yua,†, Xiao Suna,†, Myong-Ki Kimb, Mahbuba Akthera, Jun-Hyuk Hana,
5
Tae-Yeon Kima, Jun Jiangc, Tae-Bong Kangd, Kwang-Ho Leea,d,*
RI PT
4
SC
6
7
a
8
Program of Neutraceuticals Development, Konkuk University, Chungju, Korea
9
b
10
c
11
(Ministry of Education), College of Agriculture, Yanbian University, Yanji, Jilin, China
12
d
13
of Inflammatory Diseases, Konkuk University, Chungju, Korea
Department of Food Science and Engineering, Seowon University, Cheongju, Korea
Key Laboratory of Natural Resource of Changbai Mountain and Functional Molecules
†
16
EP
TE D
Department of Biotechnology, College of Biomedical & Health Science, Research Institute
These authors contributed equally to this work
AC C
14
15
M AN U
Department of Applied Life Science, Graduate School, BK21 Plus Glocal Education
17
*
18
Science, Konkuk University, Chungju, Korea
19
Tel: +82 43 840 3613; Fax: +82 43 851 5235
20
E-mail address:
[email protected] (K.-H. Lee)
Corresponding author: Department of Biotechnology, College of Biomedical & Health
ACCEPTED MANUSCRIPT ABSTRACT
22
Ethnopharmacological relevance: Chrysanthemum indicum (C. indicum), a perennial plant,
23
has long been used to treat inflammation-related disorders, such as pneumonia, hypertension,
24
gastritis, and gastroenteritis.
25
Aim of the study: The inhibitory effect of C. indicum extract (C.I) on inflammasome
26
activation was investigated to validate its potential in treating inflammation related disorders.
27
Materials and methods: LPS-primed bone marrow-derived macrophages (BMDMs) were
28
used to confirm the inhibitory effect of C.I on selective inflammasome activation in vitro. A
29
monosodium urate (MSU)-induced murine peritonitis model was employed to study the
30
effect of C.I in vivo.
31
Results: C.I inhibited activation of NLRP3 and AIM2 inflammasomes, leading to suppression
32
of interleukin-1β secretion in vitro. Further, C.I regulates the phosphorylation of apoptosis-
33
associated speck-like protein containing a CARD (ASC), which could be the main
34
contribution to attenuate these inflammasomes activation. C.I also suppressed secretion of
35
pro-inflammatory cytokines and neutrophils recruitment in MSU-induced murine peritonitis
36
model.
37
Conclusions: This study provides scientific evidence substantiating the traditional use of C.
38
indicum in the treatment of inflammatory diseases, including gout, which is induced by
39
physiologically analogous cause to MSU-induced peritonitis.
AC C
EP
TE D
M AN U
SC
RI PT
21
40 41
Keywords: Chrysanthemum indicum; inflammasome; ASC phosphorylation; MSU-induced
42
peritonitis; NLRP3
ACCEPTED MANUSCRIPT 1. Introduction
44
Inflammasomes are large cytosolic multiprotein complexes that interact with diverse
45
pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns
46
(DAMPs), leading to inflammatory responses in stimulated immune cells (Davis et al., 2011;
47
Martinon et al., 2002; Martinon et al., 2002). There are several receptors that interact with
48
PAMPs and DAMPs, such as nucleotide-binding oligomerization domain (NOD)-like
49
receptors (NLRs) family consisting of NLRP1, NLRP3, and NLRC4, and the HIN-200
50
family’s absent in melanoma 2 (AIM2). These receptors react to specific stimulus sources:
51
NLRP1 responds to Bacillus anthracis lethal factor; NLRP3 to various stimuli, including
52
crystalline, intracellular ATP, pore-forming toxin, and several microbial pathogens; and
53
NLRC4 to specific bacterial proteins, such as flagellin and AIM2 with double stranded DNAs
54
(dsDNAs) (Broz and Dixit, 2016; Duncan and Canna, 2018; Duncan and Canna, 2018;
55
Fernandes-Alnemri et al., 2009; Fernandes-Alnemri et al., 2009; Man et al., 2016; Man et al.,
56
2016; Mariathasan et al., 2004; Mariathasan et al., 2004). Commonly when the
57
inflammasomes are being activated by stimuli, activated inflammasome cleaves pro caspase-
58
1 to caspase-1. In turn, caspase-1 regulates the maturation of pro-inflammatory cytokines,
59
such as interleukin-1β (IL-1β) and interleukin-18 (IL-18), and induces lytic cell death termed
60
as pyroptosis (Broz and Dixit, 2016; Miao et al., 2011; Miao et al., 2011; Zhou et al., 2011;
61
Zhou et al., 2011). Excessive amount of IL-1β can cause various inflammatory diseases
62
(Strowig et al., 2012).
63
NLRP3 inflammasome is the most studied among the inflammasomes. Continuous and
64
excessive activation of NLRP3 inflammasomes leads to the outbreak of acute and chronic
65
inflammatory diseases, including type 2 diabetes (Vandanmagsar et al., 2011; Wen et al.,
66
2011; Wen et al., 2011), gout disease (Martinon et al., 2006), Alzheimer’s disease (Heneka et
AC C
EP
TE D
M AN U
SC
RI PT
43
ACCEPTED MANUSCRIPT al., 2013), bronchitis (Pauwels et al., 2011; Simpson et al., 2014; Simpson et al., 2014),
68
atherosclerosis (Duewell et al., 2010), and multiple sclerosis (Shaw et al., 2010).
69
AIM2 is one of the cytosolic sensors that recognizes dsDNA of microbial or host origin. The
70
assembly of AIM2 forms a multiprotein complex structure, termed as AIM2 inflammasome.
71
The excessive activation of AIM2 inflammasome by cytoplasmic self DNA causes the
72
development of psoriasis, dermatitis, arthritis, and other autoimmune diseases (Man et al.,
73
2016).
74
A common feature of the activation of NLRP3 and AIM2 inflammasomes is the need for
75
phosphorylation of the adapter ASC during the formation of the inflammasome complex.
76
ASC is the adaptor protein that interacts with the PYD of both NLRP3 and AIM2 and the
77
CARD of pro caspase-1 (Hara et al., 2013).
78
Chrysanthemum indicum (C. indicum), a wild herb belonging to the chrysanthemum family
79
has long been used as a traditional medicine to treat various inflammatory diseases such as
80
inflammation, hypertension, and respiratory diseases in China, Japan, and Korea (Cheng et al.,
81
2005). Previous reports have suggested that the extract of C. indicum exhibits anti-gout
82
(Dong et al., 2017; Kong et al., 2000; Kong et al., 2000; Park and Cho, 2016; Park and Cho,
83
2016), anti-inflammatory (Cheng et al., 2005; Wu et al., 2013; Wu et al., 2013; Xue et al.,
84
2018; Xue et al., 2018), anti-oxidant (Dong et al., 2017; Dong et al., 2017; Kong et al., 2000),
85
and anti-diabetic (Cha et al., 2018) effects.
86
Although it has been reported that C. indicum exerts an anti-inflammatory effect, the detailed
87
mechanism of its inhibitory effect, including anti-inflammasome activity, are unknown.
88
Therefore, this study aimed to investigate and elucidate the inhibitory mechanisms of C.
89
indicum on inflammasome activation.
AC C
EP
TE D
M AN U
SC
RI PT
67
ACCEPTED MANUSCRIPT 90
2. Materials and methods
92
2.1. Reagents and antibodies
93
ATP and LPS were purchased from Sigma-Aldrich (St. Louis, MO, USA). ELISA kits for IL-
94
6, Nigericin, silica crystal, z-VAD, and Lipofectamine 2000 were purchased from Invivogen
95
(San Diego, CA, USA). Antibody against β-actin was purchased from Santa Cruz
96
Biotechnology (San Diego, CA, USA). ELISA kit for IL-1β and antibody against IL-1β (AF-
97
401-NA) were obtained from R&D systems (Minneapolis, MN, USA). Antibodies against
98
NLRP3 (Cryo-2), ASC (AL177), and caspase-1 (Casper-1) were obtained from Adipogen
99
(San Diego, CA, USA). Antibody against ASC phospho-specific (Tyr144) was obtained from
100
ECM Bioscience (Versailles, KY). Antibody against phosphorylated c-Jun N-terminal kinase
101
(JNK) (G9) was obtained from Cell Signaling Technology (Danvers, MA, USA). Cocktails of
102
the protease and phosphatase inhibitor were obtained from Thermo (Rockford IL, USA).
TE D
M AN U
SC
RI PT
91
103
2.2. Plant material and extraction
105
The methanol extract of flowers, leaves, and stems of C. indicum (C.I) was purchased from
106
the plant extract bank at the Korea Research Institute of Bioscience and Biotechnology
107
(KRIBB) in Daejeon, Korea. KRIBB collected C. indicum from Korea during the month of
108
December in 2003. A voucher specimen was deposited at the herbarium of the Plant Extract
109
Bank at KRIBB for future reference (KRIBB 023-004).
AC C
EP
104
110 111
2.3. Animals
ACCEPTED MANUSCRIPT Female C57BL/6 mice (22–25 g, 6 weeks old) were purchased from Orient Bio Co., Korea.
113
Mice were housed in groups of five under standard conditions (temperature: 22 ± 2 °C,
114
humidity: 55 ± 5%, 12 h light/dark cycle) with food and distilled water. All experiments were
115
performed under the guidelines of the Konkuk University Animal Care Committee, Republic
116
of Korea (Permit No, KU18201).
RI PT
112
117
2.4. Monosodium urate (MSU)-induced peritonitis mouse model and FACS analysis
119
The protocol for the MSU-induced peritonitis mouse model was followed as described in our
120
previous report (Han et al., 2016). The obtained peritoneal cells were stained with Ly6G (Gr-
121
1, clone 1A8-Ly6g) and F4/80 (clone BM8) (eBioscience San Diego, CA, USA), and
122
analyzed using a FACS Calibur (Becton Dickinson San Diego, CA, USA). The number of
123
neutrophils was evaluated by multiplying the total number of cells by the Ly6G+/F4/80- cell
124
ratio. The supernatant of peritoneal lavage fluid was used to evaluate levels of IL-1β and IL-
125
6.
M AN U
TE D
EP
126
SC
118
2.5 Analysis of inflammasome activation
128
BMDMs were first primed with LPS (100 ng/mL) for 3 h, then the medium was replaced
129
with Opti-MEM, and the cells were pre-incubated for 1 h with C.I and inhibitors, zVAD (20
130
µM) the pan-caspase or KCl (150 mM) to block potassium efflux. The employed
131
concentrations of those inhibitors were referred in previous report (Sun et al., 2015). The
132
cells were then stimulated with ATP (5 mM) or nigericin (10 µM) for 1 h; silica crystals (150
133
µg/mL) for 3 h; and with flagellin (1.5 µg/mL) (3 h) or poly(dA:dT) (2 µg/mL) (1 h).
134
AC C
127
ACCEPTED MANUSCRIPT 2.6. Analysis of ASC and JNK phosphorylation
136
The immunoblotting of phosphorylated ASC was performed as described previously (Hoyt et
137
al., 2016; Kwak et al., 2018; Kwak et al., 2018). BMDMs were seeded in 6-well plates and
138
primed with LPS (100 ng/mL) for 3 h in RPMI, then incubated with nigericin (5 µM) and 1
139
mM sodium orthovanadate (Na3VO4) with or without C.I for 1 h in Opti-MEM. Cell lysates
140
were prepared with RIPA buffer (150 mM NaCl, 0.1% SDS, 50 mM Tris, 1.0% NP-40, and
141
0.5% DOC, pH 8). The phosphorylation of ASC in lysates was detected by immunoblotting
142
using the antiphospho-ASC antibody (Tyr144). To examine the role of C.I on JNK
143
phosphorylation, BMDMs were seeded in 12-well plates and primed with LPS (100 ng/mL)
144
for 3 h in RPMI, then incubated with nigericin (10 µM), poly(dA:dT) (2 µg/mL) with or
145
without C.I for 20 min, 40 min, and 60 min in Opti-MEM. Cell lysates were prepared with
146
RIPA buffer. The phosphorylation of JNK in lysates was detected by immunoblotting using
147
the antiphospho-JNK antibody.
TE D
M AN U
SC
RI PT
135
148
2.7. Other experimental methods
150
The experimental protocols for preparation of BMDMs, cell culture and stimulation, LDH
151
assay (Koh and Choi, 1987), MTT assay (Slater et al., 1963), and ELISA assay were followed
152
as described previously (Han et al., 2015; Shim et al., 2015; Shim et al., 2015). The protocols
153
for the separation of Triton X-100 soluble and insoluble fractions, ASC oligomerization, and
154
HPLC fingerprinting analysis were followed as described previously (Shim et al., 2013; Sun
155
et al., 2015; Sun et al., 2015).
AC C
EP
149
156 157
2.8. Statistical analyses
ACCEPTED MANUSCRIPT All values are expressed as the mean ± S.E.M. (n = 3). The statistical analysis was performed
159
using the student’s t test for two groups or a one-way ANOVA (Tukey post-hoc) for three or
160
more multiple groups (GraphPad Software San Diego, CA, USA). For all results, a P value <
161
0.05 was considered statistically significant.
RI PT
158
162
3. Results
164
3.1. C.I inhibits activation of NLRP3 and AIM2 inflammasomes but not of NLRC4
165
inflammasome
166
BMDMs were treated with various concentrations of C.I (12.5-100 µg/mL) for 24 h. MTT
167
and LDH assays were conducted to determine the cytotoxic effect of C.I. As shown in Fig.
168
1A and B, C.I did not show any cytotoxicity up to 100 µg/mL concentration in BMDMs.
169
Therefore, in this study, we used non-cytotoxic concentrations of C.I: 25, 50, and 100 µg/mL,
170
for further experiments.
171
To investigate whether C.I exerts inhibitory effect on inflammasome activation, we treated
172
LPS-primed BMDMs with C.I, and then, stimulated them with different inducers for
173
inflammasome activation. BMDMs were treated with nigericin, ATP, and silica crystals to
174
induce NLRP3 inflammasome, with p(dA:dT) to induce AIM2 inflammasome activation, and
175
with flagellin to induce NLRC4 inflammasome activation. As shown in Fig. 1C to 1F, C.I
176
suppressed NLRP3 and AIM2 inflammasome-mediated secretion of cleaved IL-1β and
177
caspase-1 in a dose-dependent manner without affecting the expression of inflammasome
178
components. C.I also inhibited NLRP3 and AIM2 inflammasome-induced pyroptosis (Fig.
179
1H). However, C.I did not suppress NLRC4 inflammasome-mediated secretion of cleaved IL-
180
1β and caspase-1 (Fig. 1G). Taken together, these data suggested that C.I exerted inhibitory
AC C
EP
TE D
M AN U
SC
163
ACCEPTED MANUSCRIPT 181
effect on both NLRP3 and AIM2 inflammasomes but not on NLRC4 inflammasome
182
activation.
183
RI PT
Fig. 1
AC C
EP
TE D
M AN U
SC
184
185 186 187
ACCEPTED MANUSCRIPT 3.2 C.I inhibits both ASC speck formation and translocation of ASC
189
The adaptor ASC acts as the bridge between caspase-1 and pyrin domain on both NLRP3 and
190
AIM2 inflammasomes to form inflammasome complex, which forms insoluble specks (Case
191
et al., 2009; Masumoto et al., 1999; Masumoto et al., 1999). Activated ASC constitutes ASC
192
oligomerization and forms insoluble speck, which is one of the essential processes for the
193
activation of some inflammasomes (Hara et al., 2013). To investigate whether C.I could
194
inhibit ASC speck formation, LPS-primed BMDMs were treated with inflammasome
195
inducers, such as nigericin or poly(dA:dT), to induce NLRP3 and AIM2 inflammasome
196
activation, respectively. ASC specks were detected by immunocytochemistry. C.I inhibited
197
ASC speck formation during NLRP3 and AIM2 inflammasome activation (Fig. 2A-B).
198
Further, to examine the effect of C.I on ASC oligomerization, LPS-primed BMDMs were
199
treated with nigericin or poly(dA:dT) to induce ASC oligomerization, followed by
200
inflammasome activation. As shown in Fig. 2C, C.I significantly inhibited formation of
201
nigericin or poly(dA:dT)-induced ASC oligomers, dimers as well as monomers. When the
202
NLRP3 inflammasome was activated, ASC translocated from soluble fraction to insoluble
203
fraction under Triton X-100 detergent lysis condition (Han et al., 2016; Shim et al., 2016;
204
Shim et al., 2016; Sun et al., 2015; Sun et al., 2015). We investigated whether C.I could
205
inhibit translocation of inflammasome complex components into Triton X-100 insoluble
206
fraction. As shown in Fig. 2D, C.I inhibited secretion of IL-1β and cleaved caspase-1 in
207
supernatant from NLRP3 or AIM2 inflammasome activated BMDMs. The levels of
208
components in soluble fractions were not changed, but the level of ASC was decreased in
209
insoluble fractions of NLRP3 or AIM2 inflammasome-activated BMDMs.
AC C
EP
TE D
M AN U
SC
RI PT
188
ACCEPTED MANUSCRIPT Fig. 2
212
EP
211
TE D
M AN U
SC
RI PT
210
3.3 C.I inhibits phosphorylation of ASC
214
Phosphorylation of ASC plays an important role in formation of inflammasome complex. In
215
previous reports, ASC phosphorylation was detected using sodium orthovanadate, a
216
phosphatase inhibitor, in J774A.1 cells (Hoyt et al., 2016; Kwak et al., 2018; Kwak et al.,
217
2018). In this study, BMDMs were primed with LPS (37 ng/mL) for 8 h and treated with
218
sodium orthovanadate for 3 h to conserve phosphorylation status. C.I decreased ASC
219
phosphorylation and inhibited IL-1β secretion (Fig. 3A). Furthermore, we investigated
220
whether C.I can inhibit ASC phosphorylation during the inflammasome activation. LPS
AC C
213
ACCEPTED MANUSCRIPT primed BMDMs were co-treated with nigericin and sodium orthovanadate for 1 h. As shown
222
in Fig. 3B, C.I inhibited phosphorylation of ASC in activated NLRP3 inflammasomes.
223
Previous studies with JNK knockout condition validated that phosphorylation of JNK occurs
224
upstream of ASC phosphorylation (Hara et al., 2013). In these reports, C.I inhibited ASC
225
phosphorylation during NLRP3 inflammasome activation. Therefore, we examined whether
226
C.I could also inhibit JNK phosphorylation. LPS primed BMDMs were pretreated with either
227
C.I or SP600125, the JNK inhibitor, and subsequently, with nigericin or poly(dA:dT). Both
228
C.I and SP600125 suppressed secretion of matured IL-1β and cleaved caspase-1 in the
229
supernatant, but C.I could not attenuate phosphorylation of JNK, unlike SP600125, during
230
NLRP3 and AIM2 inflammasome activation in cell lysates (Fig. 3C and D).These results
231
suggested that C.I inhibits ASC phosphorylation independently of JNK phosphorylation.
232
Fig. 3
AC C
EP
TE D
M AN U
SC
RI PT
221
234
AC C
233
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
235
3.4 C.I attenuates MSU induced peritonitis model
236
Following the in vitro findings, which indicated that C.I inhibits NLRP3 inflammasome
237
activation, we further investigated the biological effect of C.I in NLRP3 inflammasome
238
activation in vivo. MSU induced mouse peritonitis model has been widely used for NLRP3
239
inflammasome as an in vivo model (Martinon et al., 2006; Zhou et al., 2010; Zhou et al.,
240
2010). We analyzed the recruitment of neutrophils into the peritoneal cavity and amount of
ACCEPTED MANUSCRIPT IL-1β levels in peritoneal lavage fluid as a barometer of stimulant induced inflammation. As
242
shown in Fig. 4A, C.I treatment caused no significant effect on IL-6 that is secreted
243
independently of the NLRP3 inflammasome activation. However, a significant decrease in
244
IL-1β production in the peritoneal lavage fluid was observed after the administration of C.I
245
via both intraperitoneal and oral routes, which indicates the potential inhibition of NLRP3
246
inflammasome activation (Fig. 4B). Moreover, C.I was able to decrease the recruitment of
247
MSU-induced total cells and Ly6G+/F4/80- neutrophils in peritonium in both intraperitoneal
248
(I.P) injected model and oral administered model (Fig. 4C-E). These results suggested that
249
C.I has a protective effect on MSU induced peritonitis via the inhibition of NLRP3
250
inflammasome activation and concurrent IL-1β secretion.
AC C
EP
TE D
M AN U
SC
RI PT
241
ACCEPTED MANUSCRIPT Fig. 4
253
AC C
252
EP
TE D
M AN U
SC
RI PT
251
254
3.5 HPLC fingerprinting analysis of C.I
255
A previous phytochemical study on C. indicum reported that the extract contained several
256
compounds, such as luteolin, acacetin-7-O-rutinoside, luteolin-7-O-glucoside, apigenin-7-O-
257
glucoside, 1,5-dicaffeoylqunic acid, and chlorogenic acid (Choi et al., 2016). Similarly, our
258
HPLC fingerprinting analysis of C.I revealed that the major peak corresponded to is 1,5-
ACCEPTED MANUSCRIPT dicaffeolyquinic acid and minor peaks to luteolin and chlorogenic acid (Fig. 5). Previous
260
studies have reported the inhibitory effect of luteolin and chlorogenic acid on NLRP3
261
inflammasome activation (Shi et al., 2018; Shi et al., 2018; Zhang et al., 2018). 1,5-
262
dicaffeolyquinic acid did not show any inhibitory effect on inflammasomes activation in our
263
study (data not shown).
264
Fig. 5
265 266
EP
TE D
M AN U
SC
RI PT
259
4. Discussion
268
In this investigation, C.I attenuated the secretion of cleaved IL-1β and caspase-1 in NLRP3
269
and AIM2 inflammasome activated BMDMs, but not in NLRC4 inflammasome activated
270
BMDMs. When the NLRP3 and AIM2 inflammasomes are activated, ASC translocates from
271
Triton X-100 detergent soluble fraction to Triton X-100 insoluble fraction, and forms ASC
272
oligomerization, which is observed as ASC specks (Hara et al., 2013). It suggested that ASC
273
plays a key role in NLRP3 and AIM2 inflammasome activation. In our study, C.I suppressed
274
ASC speck formation and the level of protein in Triton X-100 insoluble fraction. These
AC C
267
ACCEPTED MANUSCRIPT results suggested that C.I attenuates NLRP3 and AIM2 inflammasomes by attenuation of
276
ASC speck formation and translocation.
277
In previous studies and our report, it was proposed that ASC phosphorylation is necessary for
278
inflammasome complex formation (Hara et al., 2013; Hoyt et al., 2016; Hoyt et al., 2016;
279
Kwak et al., 2018; Kwak et al., 2018). In this investigation, we detected a decrease in ASC
280
phosphorylation in C.I pretreated group. ASC phosphorylation can be detected not only in the
281
artificial condition but also in inflammasome activated condition. Therefore, we designed a
282
novel protocol by co-treatment of cells through inflammasome activation and treatment with
283
sodium orthovanadate. This method was applied for the first time. As a result, the decreased
284
amount of ASC phosphorylation in C.I pretreated group compared to nigericin treated group
285
could be clearly detected.
286
JNK phosphorylation is known to be upstream of ASC phosphorylation (Hara et al., 2013).
287
C.I attenuates ASC phosphorylation but does not regulate the phosphorylation of JNK.
288
Therefore, we speculated that either unknown kinases could be involved between JNK and
289
ASC phosphorylation or C.I could directly regulate the ASC phosphorylation by unknown
290
mechanism.
291
MSU crystals are well known as danger molecules which are developed after the release of
292
uric acid in dying cells (Bomalaski et al., 1986). Phagocytized MSU in macrophages
293
stimulate NLRP3 inflammasome to activate pro-caspase-1. Moreover, they are also well-
294
known stimuli for NLRP3 inflammasome, and their accumulation in joint tissue causes gout
295
arthritis via NLRP3 inflammasome (Martinon et al., 2006). The recognition of MSU crystal
296
by innate immune cells, including monocytes and resident macrophages, plays an important
297
role in gout and leads to excessive release of inflammatory cytokines, such as IL-1β (Chen et
298
al., 2006). In our present study, the in vitro results were consistent with those of in vivo
AC C
EP
TE D
M AN U
SC
RI PT
275
ACCEPTED MANUSCRIPT 299
analysis. C.I inhibited the release of IL-1β level in the fluid of peritoneal cavity of MSU-
300
induced peritonitis mouse, suggesting the potential availability of C.I on treatment of gout
301
arthritis.
RI PT
302
5. Conclusion
304
This research is the first to demonstrate scientific evidence of inhibitory effect of C. indicum
305
on inflammasome activation, as well as validating the potential of the traditional medicine of
306
C. indicum in the treatment of inflammatory disorders, such as gout, which is induced by
307
physiologically analogous cause to MSU-induced peritonitis.
308
M AN U
SC
303
Conflict of interest statement
310
The authors have declared that there is no conflict of interest.
311
TE D
309
Author Contributions
313
All authors discussed the research process, results, and implications, and all authors
314
commented on the manuscript. Sang-Hyeun Yu: Experimental design and execution, data
315
analysis and manuscript writing. Xiao Sun: Experimental design, data analysis, and
316
manuscript writing. Mahbuba Akther, Jun-Hyuk Han, Tae-Yeon Kim: In vivo experiments.
317
Jun Jiang: In vitro experiments. Myong-Ki Kim: Execution of fingerprinting analysis. Tae-
318
Bong Kang: Data analysis, discussion, and manuscript review. Kwang-Ho Lee: Supervised
319
the project and contributed to the experimental design, data analysis, and manuscript writing.
320
AC C
EP
312
ACCEPTED MANUSCRIPT Acknowledgements
322
This paper was supported by Konkuk University in 2018.
323
References
RI PT
321
Bomalaski, J.S., Lluberas, G., Schumacher Jr, H.R., 1986. Monosodium urate crystals in the
325
knee joints of patients with asymptomatic nontophaceous gout. Arthritis. Rheum. 29, 1480-
326
1484.
327
Broz, P., Dixit, V.M., 2016. Inflammasomes: mechanism of assembly, regulation and
328
signalling. Nat. Rev. Immunol. 16, 407.
329
Case, C.L., Shin, S., Roy, C.R., 2009. Asc and Ipaf Inflammasomes direct distinct pathways
330
for caspase-1 activation in response to Legionella pneumophila. Infec. Immun. 77, 1981-1991.
331
Cha, J., Nepali, S., Lee, H., Hwang, S., Choi, S., Yeon, J., Song, B., Kim, D., Lee, Y., 2018.
332
Chrysanthemum indicum L. ethanol extract reduces high-fat diet-induced obesity in mice.
333
Exp. Ther. Med. 15, 5070-5076.
334
Chen, C.J., Shi, Y., Hearn, A., Fitzgerald, K., Golenbock, D., Reed, G., Akira, S., Rock, K.L.,
335
2006. MyD88-dependent IL-1 receptor signaling is essential for gouty inflammation
336
stimulated by monosodium urate crystals. J. Clin. Invest. 116, 2262-2271.
337
Cheng, W., Li, J., You, T., Hu, C., 2005. Anti-inflammatory and immunomodulatory
338
activities of the extracts from the inflorescence of Chrysanthemum indicum Linne. J.
339
Ethnopharmacol. 101, 334-337.
AC C
EP
TE D
M AN U
SC
324
ACCEPTED MANUSCRIPT Choi, K.T., Kim, J.H., Cho, H.T., Lim, S.S., Kwak, S.S., Kim, Y.J., 2016. Dermatologic
341
evaluation of cosmetic formulations containing Chrysanthemum indicum extract. J. Cosmet.
342
Dermatol. 15, 162-168.
343
Davis, B.K., Wen, H., Ting, J.P., 2011. The inflammasome NLRs in immunity, inflammation,
344
and associated diseases. Annu. Rev. Immunol. 29, 707-735.
345
Dong, M., Yu, D., Abdullah Al-Dhabi, N., Duraipandiyan, V., 2017. The Impacts of
346
Chrysanthemum indicum Extract on Oxidative Stress and Inflammatory Responses in
347
Adjuvant-Induced Arthritic Rats. Evid. Based Complement. Alternat. Med. 2017, .
348
Duewell, P., Kono, H., Rayner, K.J., Sirois, C.M., Vladimer, G., Bauernfeind, F.G., Abela,
349
G.S., Franchi, L., Nunez, G., Schnurr, M., 2010. NLRP3 inflammasomes are required for
350
atherogenesis and activated by cholesterol crystals. Nature. 464, 1357.
351
Duncan, J.A., Canna, S.W., 2018. The NLRC 4 Inflammasome. Immunol. Rev. 281, 115-123.
352
Fernandes-Alnemri, T., Yu, J., Datta, P., Wu, J., Alnemri, E.S., 2009. AIM2 activates the
353
inflammasome and cell death in response to cytoplasmic DNA. Nature. 458, 509.
354
Han, J., Shim, D., Shin, W., Heo, K., Kwak, S., Sim, E., Jeong, J., Kang, T., Lee, K., 2015.
355
Anti-inflammatory effect of emodin via attenuation of NLRP3 inflammasome activation. Int.
356
J. Mol. Sci. 16, 8102-8109.
357
Han, J., Shim, D., Shin, W., Kim, M., Shim, E., Sun, X., Koppula, S., Kim, T., Kang, T., Lee,
358
K., 2016. Juniperus rigida Sieb. extract inhibits inflammatory responses via attenuation of
359
TRIF-dependent signaling and inflammasome activation. J. Ethnopharmacol. 190, 91-99.
AC C
EP
TE D
M AN U
SC
RI PT
340
ACCEPTED MANUSCRIPT Hara, H., Tsuchiya, K., Kawamura, I., Fang, R., Hernandez-Cuellar, E., Shen, Y., Mizuguchi,
361
J., Schweighoffer, E., Tybulewicz, V., Mitsuyama, M., 2013. Phosphorylation of the adaptor
362
ASC acts as a molecular switch that controls the formation of speck-like aggregates and
363
inflammasome activity. Nat. Immunol. 14, 1247.
364
Heneka, M.T., Kummer, M.P., Stutz, A., Delekate, A., Schwartz, S., Vieira-Saecker, A.,
365
Griep, A., Axt, D., Remus, A., Tzeng, T., 2013. NLRP3 is activated in Alzheimer’s disease
366
and contributes to pathology in APP/PS1 mice. Nature. 493, 674.
367
Hoyt, L.R., Ather, J.L., Randall, M.J., DePuccio, D.P., Landry, C.C., Wewers, M.D., Gavrilin,
368
M.A., Poynter, M.E., 2016. Ethanol and Other Short-Chain Alcohols Inhibit NLRP3
369
Inflammasome Activation through Protein Tyrosine Phosphatase Stimulation. J. Immunol.
370
197, 1322-1334.
371
Koh, J.Y., Choi, D.W., 1987. Quantitative determination of glutamate mediated cortical
372
neuronal injury in cell culture by lactate dehydrogenase efflux assay. J Neurosci Methods. 20,
373
83-90.
374
Kong, L., Cai, Y., Huang, W., Cheng, C.H., Tan, R., 2000. Inhibition of xanthine oxidase by
375
some Chinese medicinal plants used to treat gout. J. Ethnopharmacol. 73, 199-207.
376
Kwak, S., Koppula, S., In, E., Sun, X., Kim, Y., Kim, M., Lee, K., Kang, T., 2018. Artemisia
377
Extract Suppresses NLRP3 and AIM2 Inflammasome Activation by Inhibition of ASC
378
Phosphorylation. Mediators. Inflamm. 2018, .
379
Man, S.M., Karki, R., Kanneganti, T., 2016. AIM2 inflammasome in infection, cancer, and
380
autoimmunity: role in DNA sensing, inflammation, and innate immunity. Eur. J. Immunol. 46,
381
269-280.
AC C
EP
TE D
M AN U
SC
RI PT
360
ACCEPTED MANUSCRIPT Mariathasan, S., Newton, K., Monack, D.M., Vucic, D., French, D.M., Lee, W.P., Roose-
383
Girma, M., Erickson, S., Dixit, V.M., 2004. Differential activation of the inflammasome by
384
caspase-1 adaptors ASC and Ipaf. Nature. 430, 213.
385
Martinon, F., Burns, K., Tschopp, J., 2002. The inflammasome: a molecular platform
386
triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell. 10, 417-
387
426.
388
Martinon, F., Pétrilli, V., Mayor, A., Tardivel, A., Tschopp, J., 2006. Gout-associated uric
389
acid crystals activate the NALP3 inflammasome. Nature. 440, 237.
390
Masumoto, J., Taniguchi, S., Ayukawa, K., Sarvotham, H., Kishino, T., Niikawa, N., Hidaka,
391
E., Katsuyama, T., Higuchi, T., Sagara, J., 1999. ASC, a novel 22-kDa protein, aggregates
392
during apoptosis of human promyelocytic leukemia HL-60 cells. J. Biol. Chem. 274, 33835-
393
33838.
394
Miao, E.A., Rajan, J.V., Aderem, A., 2011. Caspase- 1- induced pyroptotic cell death.
395
Immunol. Rev. 243, 206-214.
396
Pauwels, N.S., Bracke, K.R., Dupont, L.L., Van Pottelberge, G.R., Provoost, S., Vanden
397
Berghe, T., Vandenabeele, P., Lambrecht, B.N., Joos, G.F., Brusselle, G.G., 2011. Role of
398
IL-1alpha and the Nlrp3/caspase-1/IL-1beta axis in cigarette smoke-induced pulmonary
399
inflammation and COPD. Eur. Respir. J. 38, 1019-1028.
400
Shaw, P.J., Lukens, J.R., Burns, S., Chi, H., McGargill, M.A., Kanneganti, T.D., 2010.
401
Cutting edge: critical role for PYCARD/ASC in the development of experimental
402
autoimmune encephalomyelitis. J. Immunol. 184, 4610-4614.
AC C
EP
TE D
M AN U
SC
RI PT
382
ACCEPTED MANUSCRIPT Shi, A., Shi, H., Wang, Y., Liu, X., Cheng, Y., Li, H., Zhao, H., Wang, S., Dong, L., 2018.
404
Activation of Nrf2 pathway and inhibition of NLRP3 inflammasome activation contribute to
405
the protective effect of chlorogenic acid on acute liver injury. Int. Immunopharmacol. 54,
406
125-130.
407
Shim, D., Han, J., Ji, Y., Shin, W., Koppula, S., Kim, M., Kim, T., Park, P., Kang, T., Lee, K.,
408
2016. Cichorium intybus Linn. extract prevents type 2 diabetes through inhibition of NLRP3
409
inflammasome activation. J. Med. Food. 19, 310-317.
410
Shim, D., Han, J., Sun, X., Jang, C., Koppula, S., Kim, T., Kang, T., Lee, K., 2013.
411
Lysimachia clethroides Duby extract attenuates inflammatory response in Raw 264.7
412
macrophages stimulated with lipopolysaccharide and in acute lung injury mouse model. J.
413
Ethnopharmacol. 150, 1007-1015.
414
Shim, D., Shin, H.J., Han, J., Ji, Y., Jang, C., Koppula, S., Kang, T., Lee, K., 2015. A novel
415
synthetic derivative of melatonin, 5-hydroxy-2’-isobutyl-streptochlorin (HIS), inhibits
416
inflammatory responses via regulation of TRIF-dependent signaling and inflammasome
417
activation. Toxicol. Appl. Pharmacol. 284, 227-235.
418
Simpson, J.L., Phipps, S., Baines, K.J., Oreo, K.M., Gunawardhana, L., Gibson, P.G., 2014.
419
Elevated expression of the NLRP3 inflammasome in neutrophilic asthma. Euro. Resp. J. 43,
420
1067-1076.
421
Slater, T., Sawyer, B., Sträuli, U., 1963. Studies on succinate-tetrazolium reductase systems:
422
III. Points of coupling of four different tetrazolium salts III. Points of coupling of four
423
different tetrazolium salts. Biochimi Biophys Acta. 77, 383-393.
AC C
EP
TE D
M AN U
SC
RI PT
403
ACCEPTED MANUSCRIPT Strowig, T., Henao-Mejia, J., Elinav, E., Flavell, R., 2012. Inflammasomes in health and
425
disease. Nature. 481, 278.
426
Sun, X., Shim, D., Han, J., Shin, W., Sim, E., Kim, M., Heo, K., Kim, Y., Koppula, S., Kim,
427
T., 2015. Anti-inflammatory effect of Impatiens textori Miq. extract via inhibition of NLRP3
428
inflammasome activation in in vitro and in vivo experimental models. J. Ethnopharmacol.
429
170, 81-87.
430
Vandanmagsar, B., Youm, Y., Ravussin, A., Galgani, J.E., Stadler, K., Mynatt, R.L.,
431
Ravussin, E., Stephens, J.M., Dixit, V.D., 2011. The NLRP3 inflammasome instigates
432
obesity-induced inflammation and insulin resistance. Nat. Med. 17, 179.
433
Wen, H., Gris, D., Lei, Y., Jha, S., Zhang, L., Huang, M.T., Brickey, W.J., Ting, J.P., 2011.
434
Fatty acid–induced NLRP3-ASC inflammasome activation interferes with insulin signaling.
435
Nat. Immunol. 12, 408.
436
Wu, X.L., Li, C.W., Chen, H.M., Su, Z.Q., Zhao, X.N., Chen, J.N., Lai, X.P., Zhang, X.J., Su,
437
Z.R., 2013. Anti-Inflammatory Effect of Supercritical-Carbon Dioxide Fluid Extract from
438
Flowers and Buds of Chrysanthemum indicum Linnen. Evid. Based Complement. Alternat
439
Med. 2013, 413237.
440
Xue, G., Li, X., Chen, C., Chen, K., Wang, X., Gu, Y., Luo, J., Kong, L., 2018. Highly
441
oxidized guaianolide sesquiterpenoids with potential anti-inflammatory activity from
442
chrysanthemum indicum. J. Nat. Prod. 81, 378-386.
443
Zhang, B., Li, Z., Xu, W., Xiang, C., Ma, Y., 2018. Luteolin alleviates NLRP3
444
inflammasome activation and directs macrophage polarization in lipopolysaccharide-
445
stimulated RAW264. 7 cells. Am. J Transl. Res. 10, 265.
AC C
EP
TE D
M AN U
SC
RI PT
424
ACCEPTED MANUSCRIPT Zhou, R., Tardivel, A., Thorens, B., Choi, I., Tschopp, J., 2010. Thioredoxin-interacting
447
protein links oxidative stress to inflammasome activation. Nat. Immunol. 11, 136.
448
Zhou, R., Yazdi, A.S., Menu, P., Tschopp, J., 2011. A role for mitochondria in NLRP3
449
inflammasome activation. Nature. 469, 221.
450
AC C
EP
TE D
M AN U
SC
451
RI PT
446
ACCEPTED MANUSCRIPT Figure captions
453
Fig. 1. C.I specifically inhibited activation of NLRP3 and AIM2 inflammasomes. BMDMs
454
were treated with the indicated concentrations of C.I for 24 h, and their viability and
455
cytotoxicity were determined by MTT assay (A) and LDH release assay (B), respectively.
456
LPS primed-BMDMs were treated with various doses of C.I for 1 h, and then, stimulated
457
with nigericin for 1 h (C), ATP for 1 h (D), silica crystals (Silica) for 3 h (E), poly(dA:dT) for
458
1 h (F), and flagellin for 3 h (G). Supernatants (SN) were used to detect the levels of cleaved
459
IL-1β and caspase-1. Cell lysates (Lys) were used to detect inflammasome components. (H)
460
LPS primed BMDMs were treated with C.I at the indicated concentrations for 1 h, and then,
461
treated with inflammasome activators; supernatants were collected for LDH determination.
462
The results are expressed as mean ± S.E.M (n = 3). ns: non-significant; **p < 0.01 and ***p
463
< 0.001 compared with control cells.
TE D
464
M AN U
SC
RI PT
452
Fig. 2. C.I inhibited both ASC speck formation and translocation of ASC into Triton X-100
466
insoluble fraction. (A) ASC specks were observed by immunofluorescent microscope. (B)
467
The proportion of cells containing ASC specks are represented as histograms. (C) ASC
468
oligomerization of NLRP3 and AIM2 in Triton X-100 insoluble (Pel) fractions from cell
469
lysates was determined by western bolt analysis. Cleaved IL-1β was detected from culture
470
supernatant (SN). Total ASC and β-actin levels in total cell lysates (Lys) were used as internal
471
control. (D) LPS primed BMDMs were treated for 1 h with various doses of C.I, and then,
472
stimulated with NLRP3 or AIM2 inflammasome stimulators. Inflammasome components in
473
Triton X-100 soluble (Lys) or insoluble (Pel) fractions of cell lysates were detected by
474
western blot using indicated antibodies. The results are expressed as mean ± S.E.M (n = 3).
475
*p < 0.05 and ***p < 0.001 compared with nigericin or poly(dA:dT)-treated cells.
AC C
EP
465
ACCEPTED MANUSCRIPT 476
Fig. 3. C.I inhibits ASC phosphorylation. (A) LPS primed BMDMs were pretreated with C.I
478
for 1 h, and then, treated with sodium orthovanadate (Na3VO4) for 3 h. IL-1β level was
479
detected by ELISA and phosphorylated ASC was detected by immunoblot. (B) LPS primed
480
BMDMs were pretreated with C.I, followed by co-treatment with nigericin and sodium
481
orthovanadate for 1 h. Phosphorylated-ASC was detected by immunoblot. LPS primed
482
BMDMs were pretreated with C.I for 1 h or SP600125 for 30 min, and then, stimulated with
483
nigericin (C) or poly(dA:dT) (D) for indicated times. Supernatants (SN) were used to detect
484
levels of cleaved IL-1β and caspase-1. Cell lysates (Lys) were used to detect p-JNK and total
485
JNK (t-JNK).
M AN U
SC
RI PT
477
486
Fig. 4. C.I attenuates MSU induced peritonitis model. C.I was intraperitoneally (i.p) injected
488
into mice 12 h and 2 h before i.p injection with MSU, or C.I was orally administered (oral)
489
into mice once a day for seven days before i.p injection with MSU. Peritoneal lavage fluid
490
was harvested 6 h after MSU injection. Concentrations of IL-6 (A) and IL-1β (B) were
491
measured by ELISA. (C) Total cells were quantified by microscope. (D) Neutrophils were
492
determined by multiplying the cell numbers by the percentage of Ly6G+/F4/80- cells from
493
total cells. (E) Ly6G+/F4/80- neutrophils were quantified by flow cytometry. * p < 0.05, ** p
494
< 0.01, and *** p < 0.001 compared to the MSU-injected group.
EP
AC C
495
TE D
487
496
Fig. 5. HPLC fingerprinting analysis of C.I. The components of C.I were determined using an
497
HPLC system.