Hesperidin Methylchalcone Suppresses Experimental Gout Arthritis in

Jun 1, 2018 - Gout arthritis is a painful inflammatory disease induced by monosodium urate (MSU) crystals. We evaluate the therapeutic potential of th...
0 downloads 0 Views 4MB Size
Subscriber access provided by Kaohsiung Medical University

Bioactive Constituents, Metabolites, and Functions

Hesperidin methylchalcone suppresses experimental gout arthritis in mice by inhibiting NF-kB activation Kenji W Ruiz-Miyazawa, Felipe Almeida Pinho-Ribeiro, Sergio M Borghi, Larissa StaurengoFerrari, Victor Fattori, Flávio Amaral, Mauro Martins Teixeira, José Carlos Faria AlvesFilho, Thiago Mattar Cunha, Fernando Q Cunha, Rubia Casagrande, and Waldiceu A Verri J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.8b00959 • Publication Date (Web): 01 Jun 2018 Downloaded from http://pubs.acs.org on June 1, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 48

Journal of Agricultural and Food Chemistry

1

Hesperidin methylchalcone suppresses experimental gout arthritis in mice by inhibiting NF-

2

κB activation

3 4

Kenji W. Ruiz-Miyazawa†, Felipe A. Pinho-Ribeiro†, Sergio M. Borghi†, Larissa Staurengo-

5

Ferrari†, Victor Fattori†, Flavio A. Amaral‡, Mauro M. Teixeira‡, Jose C. Alves-Filho§, Thiago M.

6

Cunha§, Fernando Q. Cunha§, Rubia Casagranded, and Waldiceu A. Verri, Jr †,*

7 8



9

Garcia Cid, Km 380, PR445, 86057-970, Cx. Postal 10.011, Londrina, Paraná, Brazil.

Departamento de Ciências Patológicas, Universidade Estadual de Londrina-UEL, Rod. Celso

10



11

Imunofarmacologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.

12

§

13

Bandeirantes s/n, 14050-490, Ribeirão Preto, São Paulo, Brazil.

14

¥

15

Robert Koch, 60, Hospital Universitário, 86038-350, Londrina, Paraná, Brazil.

Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Laboratório de

Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida

Departamento de Ciências Farmacêuticas, Universidade Estadual de Londrina-UEL, Avenida

16 17

*Corresponding author: Waldiceu A. Verri Jr - Department of Pathology, Biological Sciences

18

Center, Londrina State University. Rod. Celso Garcia Cid, Pr 445, Km380, Londrina, Paraná,

19

Brazil, Cx. Postal 10.011, CEP 86057-970, Tel: + 55 43 33714979, FAX: +55 43 33715828, e-mail

20

addresses: [email protected]; and [email protected].

21 22 23 24 25

1 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

26

ABSTRACT

27

Gout arthritis is a painful inflammatory disease induced by monosodium urate (MSU) crystals. In

28

this work, we evaluate the therapeutic potential of the flavonoid hesperidin methylchalcone (HMC)

29

in a mouse model of gout arthritis induced by intra-articular injection of MSU (100 µg/10 µL).

30

Orally given HMC (3-30 mg/kg, 100 µL) reduced in a dose-dependent manner the MSU-induced

31

hyperalgesia (44%, p < 0.05), edema (54%, p < 0.05) and leukocyte infiltration (70%, p < 0.05).

32

HMC (30 mg/kg) inhibited MSU-induced infiltration of LysM-eGFP+ cells (81%, p < 0.05),

33

synovitis (76%, p < 0.05), and oxidative stress (increased GSH, FRAP and ABTS by 62%, 78% and

34

73%, respectively; and reduced O2- and NO by 89% and 48%, p < 0.05), and modulated cytokine

35

production (reduced IL-1β, TNF-α, IL-6 and IL-10 by 35%, 72%, 37% and 46%, respectively, and

36

increased TGF-β by 90%, p < 0.05). HMC also inhibited MSU-induced NF-κB activation (41%,

37

p < 0.05), gp91phox (66%, p < 0.05) and NLRP3 inflammasome components mRNA expression in

38

vivo (72%, 77%, 71% and 73% for NLRP3, ASC, pro-caspase-1 and pro-IL-1 β, respectively,

39

p < 0.05), and induced Nrf2/HO-1 mRNA expression (3.9 and 5.1 fold increase, respectively,

40

p < 0.05). HMC (30, 100 and 300 µM) did not inhibit IL-1β secretion by macrophages primed by

41

LPS and challenged with MSU (450 µg/mL), demonstrating that the anti-inflammatory effect of

42

HMC in gout arthritis depends on inhibiting NF-κB, but not on direct inhibition of inflammasome.

43

The pharmacological effects of HMC indicate its therapeutic potential for the treatment of gout.

44 45

KEYWORDS: Gout arthritis, cytokines, NLRP3 inflammasome, NF-κB, oxidative stress.

46 47 48 49 50

2 ACS Paragon Plus Environment

Page 2 of 48

Page 3 of 48

Journal of Agricultural and Food Chemistry

51

1. INTRODUCTION

52 53

Gout is an arthritic disease characterized by articular and peri-articular deposition of

54

monosodium urate (MSU) crystals. Deposition of MSU crystals affects mainly distal joints because

55

low temperatures facilitate crystal precipitation, causing intense pain and articular inflammation.1,2

56

The incidence of gout is associated with increasing of age, affecting mainly middle-aged or older

57

patients, reaching a plateau in men after 70 years old.3 In women, gout is more commonly observed

58

after menopause due to the reduction in estrogen levels since this hormone regulates the elimination

59

of uric acid by the kidneys.4 Current treatments for gout arthritis include colchicine, corticosteroids

60

NSAIDs (and non-steroidal anti-inflammatory drugs).5,6 Side effects of these drugs include gastritis,

61

hormonal imbalance as well as bleeding and renal dysfunction, which support the need of

62

developing novel pharmacological approaches with reduced side effects.7

63

In gout arthritis, there is an intense inflammatory response and immune cell activation. This

64

is because MSU crystals induce cytokine production by tissue resident cells, including macrophage-

65

like synoviocytes, and by recruited cells, including monocytes and neutrophils.1,6,7 Evidence

66

indicates that MSU crystals activate macrophages via CD14 (cluster of differentiation 14) and

67

TLR4 (toll-like receptor 4), which triggers the production of the pro-inflammatory mediators tumor

68

necrosis factor-alpha (TNF-α), interleukin (IL)-1β, IL-6, IL-8 and monocyte chemotactic factors.8,9

69

These cytokines increase the production of reactive species such as superoxide anion and nitric

70

oxide (NO) by NADPH (nicotinamide adenine dinucleotide phosphate) oxidase and iNOS

71

(inducible nitric oxide synthase), respectively.10 These reactive oxygen species (ROS) contribute to

72

tissue lesion observed in gout arthritis.11,12

73

Importantly, both ROS and MSU are well-known activators of NLRP3 (NACHT, LRR and

74

PYD domains-containing protein 3) inflammasome.1,13,14 Macrophages phagocytize MSU crystals,

75

which causes the rupture of the phagolysosome membrane and the release of cathepsin G into the

3 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

76

cytoplasm. In turn, cathepsin G activates the NLR receptor NLRP3 that recruits the adaptive

77

molecule ASC (apoptosis-associated speck-like protein containing CARD), which interacts with

78

pro-caspase-1 resulting in its activation to cleave pro-IL-1β to generate its mature form IL-1β.1,13–15

79

Then, released IL-1β induces inflammation and further activates the transcription factor NF-

80

κB.1,13,15 This activity is reciprocal since NF-κB signaling stimulates the expression of more pro-IL-

81

1β, as well as inflammasome components NLRP3, ASC, pro-caspase-1.15,16 As a consequence,

82

enhanced NF-κB activity together with MSU-induced activation of NLRP3 induces pronounced IL-

83

1β release that orchestrates gout arthritis. Proving the importance of this concept, the treatment with

84

IL-1 receptor antagonist (IL-1ra) known as Anakinra reduces acute gout arthritis flares in clinical

85

practice.7 Therefore, NLRP3 inflammasome activation and NF-κB activation are central

86

mechanisms in the development of gout arthritis.13,17 This NF-κB and NLRP3 inflammasome

87

system can be investigated in vitro in cultured macrophages as demonstrated in the seminal

88

manuscript that unveiled this essential pathologic mechanism in gout arthritis shaping our current

89

understanding of this disease.13 In this in vitro macrophage system, lipopolysaccharide (LPS) is

90

signal 1 to trigger NF-κB activation and induction of the expression of NLRP3 inflammasome

91

components, and MSU is signal 2 that activates NLRP3 inflammasome.13,18

92

Hesperidin, or 3,5,7-trihydroxy flavanone 7-rhamnoglucoside, is a flavonoid belonging to

93

the class of flavanones, which is found in vegetables and fruits such as oranges and grape fruit,

94

especially in citrus fruits.19 In general, flavonoids present several biological properties and have

95

antioxidant, anti-allergic, anti-tumoral, antimicrobial, anti-inflammatory and analgesic effects.20

96

Several studies demonstrated that hesperidin reduces oxidative stress and inflammation.21,22

97

However, the bioactivity of hesperidin is profoundly affected due to its poor water solubility and

98

low intestinal absorption when compared with other flavonoids. Methylation under alkaline

99

conditions produces hesperidin methylchalcone (HMC), or (E)-1-[4-[[6-O-(6-Deoxy-α-L-

100

mannopyranosyl)-ß-D-glucopyranosyl]oxy]-2-hydroxy-6-methoxyphenyl]-3-(3-hydroxy-44 ACS Paragon Plus Environment

Page 4 of 48

Page 5 of 48

Journal of Agricultural and Food Chemistry

101

methoxyphenyl)-2-propen-1-one, that presents greater bioavailability, metabolic stability and tissue

102

distribution than hesperidin.23

103

HMC induces vasodilation, vascular permeability and stabilizes prostaglandin E.24 In fact,

104

HMC is clinically used as a vasoprotective agent similarly to other drugs used to treat vascular

105

disorders such as Cyclo 3 Fort and Cirkan.25,26 Recently, our group showed that HMC presents

106

analgesic, antioxidant, and anti-inflammatory activity in mouse models of inflammatory pain. These

107

effects have been associated with inhibition of cytokine production and NF-κB activation.22 In the

108

current study, we investigate the therapeutic potential of HMC in a murine model of gout arthritis

109

and its effects on pro-inflammatory and pro-oxidative pathways known to contribute to gout

110

disease.

111 112 113 114 115 116 117 118 119 120 121 122 123 124 125

5 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

126

2. MATERIALS AND METHODS

127 128

2.1. Chemicals. NaCl 0.9% was supplied by Fresenius Kabi Brasil Ltda. (Aquiraz, CE,

129

Brazil). NaOH was obtained from Labsynth Ltda. (Diadema, SP, Brazil). Hesperidin

130

methylchalcone (HMC, PubChem CID: 6436550, MW: 624.592 g/mol, ≥95% purity) from Santa

131

Cruz Biotechnology (Santa Cruz, CA, USA). ELISA Ready-SET-Go! Kits from eBioscience (San

132

Diego, CA, USA). PathScan® kits from Cell Signaling Technology (Beverly, MA, USA). ABTS

133

[2,2-azinobis (3-ethylbenzothiazoline-6-sulfonate)], NBT (nitroblue tetrazolium), 2,4,6-tripiridil-s-

134

triazine (TPTZ), ferric chloride hexahydrate, 6-hydroxy- 2,5,7,8-tetramethylchroman-2-11

135

carboxylic (trolox) and monosodium urate crystals (MSU) from Millipore Sigma (St. Louis, MO,

136

USA), and quercetin (PubChem CID: 5280343, MW: 302.238 g/mol, 95% purity) was from Acros

137

(Pittsburg, PA, USA).

138 139

2.2. Animals. Swiss mice (male, 25-30 g) were obtained from the animal facility of

140

Universidade Estadual de Londrina (Londrina, PR, Brazil), and LysM-eGFP+ mice (male, 20-25 g)

141

from animal facility of Ribeirão Preto Medical School, University of São Paulo (Ribeirão Preto, SP,

142

Brazil). The heterogeneous Swiss mice strain is currently used by our laboratory to study the

143

molecular and cellular mechanism related to inflammatory diseases and inflammatory pain12,22 and

144

was used for all experimental analysis in the present study, with exception of the experiment

145

evaluating the participation of LysM+ leukocytes in the model, in which LysM-eGFP+ mice were

146

used. LysM-eGFP+ strain was generated through its background C57BL/6 mouse and present eGFP

147

(enhanced green fluorescent protein) expression controlled by the lysozyme M promoter (LysM),

148

enzyme that is found predominantly in neutrophils granules and secreted upon cell activation.27

149

LysM-eGFP+ mice were used to corroborate the data on leukocyte recruitment using total and

150

differential cell counts. The difference in the background of mice (Swiss and C56BL/6) is a likely

6 ACS Paragon Plus Environment

Page 6 of 48

Page 7 of 48

Journal of Agricultural and Food Chemistry

151

explanation for the difference in the intensity of cellular recruitment observed in the results section.

152

Animals were housed in standard clear plastic cages with water and food ad libitum, in a 12/12h

153

light/dark cycle and controlled temperature (21ºC). Behavioral measurements were acquired

154

between 9 a.m. and 5 p.m. in a temperature controlled room (21ºC). Euthanasia was performed with

155

deep isoflurane anesthesia (5% in oxygen, Abbott Park, IL, USA) followed by cervical dislocation

156

and decapitation. All procedures were previously approved by institutional Ethics Committee on

157

Animal Use (CEUA) of Universidade Estadual de Londrina (process n. 14600.2013.73). Every

158

effort was made to avoid animal stress and discomfort and reduce the number of mice used. We did

159

not observe unexpected death of mice during the study.

160 161

2.3. Preparation of MSU crystal. We prepared the MSU crystals following the method

162

described previously.12 Monosodium urate (800 mg) was dissolved in boiling milli-Q water (155

163

ml) and NaOH (5 ml). The solution was then adjusted to the pH 7.2 and gradually cooled at room

164

temperature. After centrifugation (3,000 g, 2 min, 4◦C), the aqueous portion was evaporated and

165

crystals sterilized at 180◦C for 2 hours. Crystals were collected and kept sterile until use. Presence

166

of endotoxin in the MSU crystals was assessed using a ToxinSensor™ Single Test Kit (GenSricpt)

167

following manufacturer’s instructions. This is a qualitative in vitro end-point endotoxin test used for

168

pre-clinical studies and supplies a series of kits with different sensitivities of which we used the

169

highest sensitivity testing tube (0.015 EU/mL). Each kit contains a lysate of Limulus Polyphemus

170

(LAL) that detect the labeled concentration (EU/mL) of the FDA Reference Standard Endotoxin.

171

Briefly, 200 µL of sample was added to tubes containing LAL reagent and incubated in a water bath

172

(60 min, 37°C). The presence of endotoxin results in the formation of a viscous gel. Entodotoxin

173

free water was used as negative control. We tested titrated concentrations of MSU diluted in

174

endotoxin free water (10, 5, 2, 1 and 0.5 mg/mL). The test was positive for 200 µL of a 10 mg/mL

175

solution (2000 µg of MSU crystals), and negative for 200 µL of a 5 mg/ml solution (1000 µg of

7 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

176

MSU crystals) and solutions with lower concentrations of MSU crystals. These results indicate that

177

the endotoxin concentration in 200 µL of a 10 mg/mL solution of MSU is between 0.015 and 0.03

178

EU/mL. The in vivo dose of MSU used in this study was 100 µg/ 10 µL and in vitro concentration

179

of MSU was 450 µg/mL. Therefore, the selected dose and concentration of MSU did not present

180

detectable endotoxin levels.

181 182

2.4. Induction of articular joint inflammation. MSU (100 µg/10µL, i.a.) injection of the

183

right knee was performed to induce joint inflammation in mice. Sterile saline was administered in

184

the control group.12 To reduce suffering and stress, all i.a. injections were performed under

185

anesthesia (isoflurane 5% in oxygen, Abbott Laboratories, IL, USA).

186 187

2.5. Electronic pressure-meter test. Flexion-induced pain was performed using the

188

electronic pressure-meter test as previously described.12 Briefly, tibiotarsal joint flexion was

189

induced by application of pressure in the plantar surface using a non-nociceptive tip probe (area of

190

4.15 mm2 ). The pressure necessary to evoke flexion-elicited paw withdrawal response was

191

automatically recorded in grams (n = 6 mice/group/experiment, experiments were performed in

192

twice).

193 194

2.6. Edema. A Mitutoyo caliper (Suzano, SP, Brazil) was used to measure thickness of knee

195

joint before (time-point zero) and after the i.a. injection of MSU or saline at different time-points (n

196

= 6/group/experiment, experiments were performed twice). Increases in articular thickness were

197

calculated as the individual differences between the measurements after injection and the starting

198

value (time-point zero), and edema presented as ∆ reaction in mm.22

199

8 ACS Paragon Plus Environment

Page 8 of 48

Page 9 of 48

Journal of Agricultural and Food Chemistry

200

2.7. Total and differential cell counts. Knee joint cavity was washed with 3.3 µL of PBS +

201

1 mM EDTA, three times (one knee joint wash per sample, n = 6/group/experiment, experiments

202

were performed twice), Samples were diluted in Turk`s solution and used for total leukocyte

203

quantification in a Neubauer chamber. Cytocentrifuge stained slides (Rosenfeld) and Cytospin 4

204

(Shandon, Pittsburg, PA, USA) were used for differential cell counts. Widefield light microscope

205

was used for quantification and results presented as mean ± SEM of cells (x103) per cavity.12

206 207

2.8. Immunofluorescence assay. Articular fluids of LysM-eGFP+ mice were collected in

208

sterile slides 15 hours after MSU i.a. injection into the knee joints using the same procedures

209

presented in sub-section 2.7., and processed for immunofluorescence assay (one knee joint wash per

210

sample, n = 6/group/experiment, experiments were performed twice). DAPI fluorescent stain was

211

added to slides for localization of nucleus in each sample. The representative images and

212

quantitative analysis were performed using a confocal microscope (SP8, Leica Microsystems,

213

Mannheim, Germany). The intensity of fluorescence was quantified in randomly selected fields of

214

different groups by a blind evaluator. Result is presented as the percentage of GFP fluorescent

215

intensity.

216 217

2.9. Histopathological analysis. Knee joints were dissected 15 hours after MSU or saline

218

injection (one whole knee joint per sample, n = 6/group/experiment, experiments were performed

219

twice) and fixed in 10% buffered formalin in PBS. After fixation, decalcification of samples was

220

performed by incubation in EDTA for 10 days. Decalcified samples were embedded in paraffin,

221

sectioned, and stained with hematoxylin and eosin (H&E) for histopathological analysis.

222

Conventional morphological analysis was performed in 10 aleatory fields by an investigator blinded

223

to the experimental groups.

224

9 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

225

2.10. Measurement of reduced glutathione (GSH) levels. Knee joints were dissected and

226

samples collected 15 h after MSU injection (one whole knee joint per sample, n =

227

6/group/experiment, and experiments were performed twice) and stored at -80 ºC for 48 h or more.

228

Samples were homogenized in 0.02 M EDTA (200 µL), and trichloroacetic acid (25 µL, 50% v/v)

229

added the homogenate. Samples were vortexed three times over 15 min, centrifuged (15 min, 1500

230

g, 4 ºC), and supernatant collected. Supernatants were mixed with TRIS buffer (200 µL, 0.2 M, pH

231

8.2) and DTNB (10 µL, 0.01 M) and incubated for 5 min at room temperature before absorbance

232

measurement at 412 nm (Multiskan GO, Thermo Scientific). Samples were normalized to

233

absorbance of wells with reagent only. GSH values were calculated using a standard GSH curve and

234

expressed as GSH/mg of protein.22

235 236

2.11. ABTS and FRAP assays. Samples from knee joints were dissected 15 h after injection

237

of MSU or saline and used to evaluate the antioxidant capacity by ABTS and FRAP microplate-

238

adapted assays as described before.22 After dissection, samples were homogenized in 500 µL of ice-

239

cold KCl buffer (1.15% w/v), centrifuged (200 g, 10 min, 4 °C), and supernatants collected (one

240

whole knee joint per sample, n = 6/group/experiment, and experiments were performed twice). For

241

ABTS assay, 10 µL of supernatant were mixed with 200 µL of ABTS solution, incubated for 6 min

242

at 25 °C, and absorbance determined (730 nm). For FRAP assay, 10 µL of supernatant were mixed

243

with 150 µL of fresh FRAP reagent, incubated for 30 min at 37 °C, and absorbance determined at

244

595 nm (Multiskan GO Thermo Scientific). Antioxidant capacity of the samples was compared with

245

a standard curve of antioxidant Trolox (0.02 – 20 nmol).

246 247

2.12. Production of superoxide anion. Knee joint homogenates (10 mg/mL) were prepared

248

in 1.15% KCl, and superoxide anion (O2-) production measured using the microplate-adapted

249

nitroblue tetrazolium (NBT) reduction assay (one whole knee joint per sample, n =

10 ACS Paragon Plus Environment

Page 10 of 48

Page 11 of 48

Journal of Agricultural and Food Chemistry

250

6/group/experiment, experiments were performed twice). Samples were collected 15 h after MSU

251

injection.22 Absorbance was measured at 600 nm, and results normalized by the tissue weight.

252 253

2.13. Production of Nitrite. Knee joint was dissected 15 h after MSU or saline injection

254

(one whole knee joint per sample, n = 6/group/experiment, experiments were performed twice),

255

homogenized in 500 µL of saline, and the concentration of nitrite (NO2-) determined using the

256

Griess reaction.28 Homogenates (100 µL) were incubated with Griess reagent (100 µL) for 5 min

257

(25 °C), and the absorbance at 550 nm compared to a standard curve of NaNO2 solution. Tissue

258

weight was used for normalization, and the results expressed as µmol of NO2- /mg.

259 260

2.14. RT-qPCR. Samples of articular tissue were homogenized in Trizol reagent and used

261

for RT-qPCR (real time and quantitative polymerase chain reaction) as previously described.12. SV

262

Total RNA Isolation System (Promega Corporation, Madison, WI, USA) was used for RNA

263

extraction (one whole knee joint per sample, n = 6/group/experiment, experiments were performed

264

twice). All reactions were performed in triplicate. Cycling conditions: 2 min at 50° C, 2 min at

265

95°C, and 40 cycles of 15 s at 95° C and 30s at 60° C (LightCycler Nano Instrument, Roche,

266

Mississauga, ON, USA) using the Platinum SYBR Green qPCR SuperMix UDG (Invitrogen, USA).

267

The primers used were GAPDH forward: CAT ACC AGG AAA TGA GCT TG, reverse: ATG

268

ACA TCA AGA AGG TGG TG; Nrf2, forward: TCA CAC GAG ATG AGC TTA GGG CAA,

269

reverse: TAC AGT TCT GGG CGG CGA CTT TAT; gp91phox, forward: AGC TAT GAG GTG

270

GTG ATG TTA GTG G, reverse: CAC AAT ATT TGT ACC AGA CAG ACT TGA G; Nlrp3,

271

forward: AGC TAT GAG GTG GTG ATG TTA GTG G, reverse: CAC AAT ATT TGT ACC

272

AGA CAG ACT TGA G; HO-1, forward: CCC AAA ACT GGC CTG TAA AA, reverse: CGT

273

GGT CAG TCA ACA TGG AT; pro-caspase-1: forward: TGG TCT TGT GAC TTG GAG GA,

274

reverse: TGG CTT CTT ATT GGC ACG AT; pro-IL-1β, forward: GAA ATG CCA CCT TTT

11 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

275

GAC AGT G, reverse: TGG ATG CTC TCA TCA GGA CAG; ASC, forward: ATG GGG CGG

276

GCA CGA GAT G, reverse: GCT CTG CTC CAG GTC CAT CAC. mRNA data was normalized

277

by GAPDH expression in each sample.

278 279

2.15. Cytokine measurement. Knee joint was dissected 15 h after saline or MSU injection

280

and homogenized in 500µL of buffer containing protease inhibitor (1 mM phenylmethanesulfonyl

281

fluoride, Sigma Aldrich) (one whole knee joint per sample, n = 6/group/experiment, experiments

282

were performed twice. Levels of TNF-α, TGF-β, IL-1β, IL-6, and IL-10 were measured by ELISA

283

using eBioscience kits and manufacture instructions, and the results expressed in picograms

284

(pg)/100 mg of tissue22

285 286

2.16. Activation of NF-κB. Knee joints were dissected 15 h after saline or MSU injection

287

(one whole knee joint per sample, n = 6/group/experiment, experiments were performed twice) and

288

homogenized at 4°C in lysis buffer (Cell Signaling). Samples were centrifuged (14000 rpm, 10 min,

289

4 °C) and supernatants collected. Total and phosphorylated NF-κB (p65 subunit) levels were

290

determined by ELISA PathScan® kits (Cell Signaling). Absorbance was measured at 450 nm and

291

results presented as total p65/phospho-p65 ratio as described previously.12

292 293

2.17. Isolation and culture of bone marrow-derived macrophages for inflammasome

294

activation assay. Mouse bones (femora and tibiae) were flushed with RPMI 1640 media and bone

295

marrow cells suspension cultured in supplemented RPMI 1640 medium (10% FBS + 15% L929 cell

296

conditioned medium). At day 7, bone marrow-derived macrophages (BMDMs) were harvested and

297

seeded in 96-well plates (1.5x105 cells per well). BMDMs were pre-treated with lipopolysaccharide

298

(LPS, 500 ng/mL) from Escherichia coli (Santa Cruz Biotechnology) and stimulated with 450

299

µg/mL of MSU 3 h later to induce activation of NLRP3 inflammasome as previously described.13

12 ACS Paragon Plus Environment

Page 12 of 48

Page 13 of 48

Journal of Agricultural and Food Chemistry

300

BMDMs were treated with HMC (30, 100 or 300 µM), or Quercetin (30 µM), 30 min before MSU

301

stimulation. After 5 h of MSU application, cell culture supernatants were collected and IL-1β

302

concentration quantitated by ELISA. Lactate dehydrogenase (LDH) release in the supernatant was

303

used as a marker of cellular viability (n = 6 wells/group/experiment, experiments were performed

304

twice).

305 306

2.18. Experimental procedures. HMC or saline (vehicle) was administered orally (3- 30

307

mg/kg) 1 h before stimulus with MSU (100 µg/10 µL, i.a.). Edema and mechanical hyperalgesia

308

were measured at different time points after MSU or saline injection. Total and differential

309

quantification of articular leukocytes was evaluated 15 h after MSU injection. The dose of 30 mg/kg

310

of HMC was used in subsequent experiments. Mice received HMC (30 mg/kg) or saline, and

311

stimulated with MSU (100 µg/10 µL, i.a. injection) for the determination of LysM-eGFP+

312

neutrophil infiltration in the knee joint lavage, histopathological analysis, cytokine production

313

(TNF-α, TGF-β, IL-1β, IL-6, and IL-10), oxidative stress (GSH, FRAP, ABTS, superoxide anion,

314

and NO assays), NF-κB activation (total NFκB p65/phosphorylated NFκB p65 ratio), and gp91phox,

315

HO-1 (heme-oxigenase-1), Nrf2 (nuclear factor (erythroid-derived 2)-like 2)), NLRP3 (NACHT,

316

LRR and PYD domains-containing protein 3), ASC (apoptosis-associated speck-like protein

317

containing a CARD), pro-caspase-1 and pro-IL-1β mRNA expression in the knee joint samples

318

collected 15 h after i.a. stimulus with MSU crystals. In vitro analyses using LPS-primed BMDMs

319

treated with HMC (30, 100 or 300 µM) were performed 5 hours after subsequent stimulation with

320

MSU crystals (450 µg/mL) for the determination of mature IL-1β secretion. Figure 1 summarizes

321

these in vivo (A) and in vitro (B) experimental procedures. Unlike humans, mice express the

322

peroxisomal enzyme uricase that degrades urate. In fact, evidence demonstrates that humans have

323

higher levels of urate than mice (≈240-360 µM vs ≈30-50 µM, respectively).17,29 Uricase efficiently

324

oxidizes urate to allantoin, reducing the levels of uric acid in the mouse. Consequently, mice do not

13 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

325

develop gout arthritis under natural conditions, and therefore, the inflammatory response has a

326

shorter duration in mice than in humans.17,29 This is an explanation for the time point of evaluation

327

at 15h, which was standardized in previous studies of our group.30,31 Furthermore, this is also a

328

limitation of the mouse model of gout arthritis, which without the expression of uricase could

329

present a prolonged inflammatory response.17,29

330 331

2.19. Data analyses. Results are presented as means ± standard error mean (SEM) of

332

measurements (n = 6/group/experiment, two independent experiments). Two-way ANOVA was

333

used for statistical comparison of two or more groups at multiple time points (Fig. 2). One-way

334

ANOVA was used for statistical comparison of three or more groups at a single time point (Fig. 3-

335

10). Tukey’s post hoc test was used. Analyzed factors include time, treatments (groups), and time

336

versus treatment interaction. Statistical differences were considered when p < 0.05. GraphPad Prism

337

v5.0 was used for statistical analysis.

338 339 340 341 342 343 344 345 346 347 348 349

14 ACS Paragon Plus Environment

Page 14 of 48

Page 15 of 48

350

Journal of Agricultural and Food Chemistry

3. RESULTS

351 352

3.1. HMC reduces mechanical hyperalgesia and articular edema induced by MSU.

353

Intra-articular injection of MSU crystals caused mechanical hyperalgesia as expected. At the dose

354

of 3 mg/kg, HMC showed no protective effects on hyperalgesia (Figure 2A). At the medium dose

355

(10 mg/kg), HMC was able to reduce significantly the hyperalgesia at 7h and 15 h after MSU

356

injection when compared to groups that received vehicle or the low dose of HMC (Figure 2A). At

357

the higher dose (30 mg/kg), HMC inhibited hyperalgesia between 5-15 h (up to 44%) after MSU

358

injection. The higher dose of HMC was more efficient than the medium dose at 15 h time point

359

(Figure 2A). In the same treatment schedule, the low dose of HMC (3 mg/kg) had no effect on

360

articular edema, while the medium dose (10 mg/kg) of HMC significantly inhibited MSU-induced

361

edema between 7-15 h (Figure 2B). In parallel with its effect on hyperalgesia, the higher dose of

362

HMC (30 mg/kg) was also more efficient than the other two doses in reducing articular edema (5-15

363

h, 54%) (Figure 2B). Thus, HMC dose-dependently inhibits MSU-induced hyperalgesia and edema.

364 365

3.2. HMC inhibits intra-articular recruitment of leukocytes induced by MSU. After

366

intra-articular injection of MSU, a significant increase of total leukocytes (Figure 3A), mononuclear

367

(Figure 3B) and neutrophil (Figure 3C) counts was observed in the knee joint exudate. HMC

368

presented the same profile of inhibition of MSU-induced intra-articular leukocytes (total leukocytes,

369

mononuclear cells, and neutrophils) (Figure 3A-C) in which the lower dose (3 mg/kg) presented no

370

effect, medium dose (10 mg/kg) induced a tendency (not significant) of reduction, and the higher

371

dose (30 mg/kg) induced a reduction of 60% in total leukocytes (Figure 3A), 70% in mononuclear

372

cells (Figure 3B), and 56% in neutrophils (Figure 3C). Thus, we selected the higher dose of HMC

373

(30 mg/kg) for the next tests. In a separate protocol, we also performed experiments using samples

374

of articular washes of LysM-eGFP+ mice. LysM is a reliable marker of neutrophil population.32 In

15 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

375

fact, this gene-targeted mouse expressing eGFP is considered an efficient tool to determine the

376

neutrophils/monocytes counts in models of diseases.27,32 In this sense, our primary aim in using this

377

specific tool was to determine the dynamics of neutrophils in experimental gout and whether

378

treatment with HMC would be able to reduce the neutrophilic infiltration, and not compare the

379

response obtained in the two different strains used in the present study (Swiss vs LysM-eGFP+).

380

MSU induced a robust infiltration of LysM+ neutrophils when compared to control animals, which

381

was inhibited by HMC treatment (Figure 3D-M).

382 383

3.3. HMC reduces MSU-induced knee joint synovitis. Histopathological analysis show

384

that saline group presented regular histological appearance (Figure 4A). In turn, MSU induced

385

significant joint synovitis with marked increase of inflammatory cells (Figure 4B), which was

386

decrease by the treatment with HMC by 76% (Figure 4C) as observed by inflammatory cell counts

387

per field in experimental groups (Figure 4D).

388 389

3.4. HMC inhibits MSU-induced depletion of endogenous antioxidant apparatus and

390

oxidative stress. MSU induced oxidative stress in the knee joint as observed by a significant

391

decrease in antioxidant defense in the knee joint, which was prevented by HMC treatment as

392

observed by GSH (62%, Figure 5A), FRAP (78%, Figure 5B) and ABTS (73%, Figure 5C) assays.

393

Corroborating the increase of antioxidant defenses, HMC was able to inhibit MSU-induced

394

superoxide anion (89%, Figure 5D) and NO (48%, Figure 5E) production. Thus, HMC inhibited

395

MSU-induced oxidative stress.

396 397

3.5. HMC inhibits the expression of gp91phox and increases expression of Nfr2 and HO-

398

1 in the knee joint after MSU injection. MSU enhanced the expression of gp91phox mRNA, and

399

this increase was significantly inhibited by HMC in 66% (Figure 6A). While no differences in Nrf2

16 ACS Paragon Plus Environment

Page 16 of 48

Page 17 of 48

Journal of Agricultural and Food Chemistry

400

and HO-1 mRNA expression were observed after MSU injection, HMC enhanced their expression

401

by approximately 4 and 5 fold, respectively (Figure 6B and 6C, respectively). Thus, HMC inhibited

402

the MSU-induced expression of a gene involved in promoting the production of superoxide anion,

403

and induced the expression of genes involved in promoting antioxidant effects.

404

3.6. HMC modulates MSU-induced cytokine production in the knee joint. Following

405 406

the intra-articular injection of MSU, increased levels of the pro-inflammatory cytokines TNFα, IL-

407

1β and IL-6 were observed, and this was inhibited by HMC treatment (35%, 72% and 37%,

408

respectively) (Figure 7A-C). HMC also efficiently reduced the levels of IL-10 after MSU injection

409

(46%, Figure 7D). Nevertheless, HMC induced approximately 2 fold increase of MSU-induced

410

TGF-β production, which is also an anti-inflammatory cytokine in gout arthritis (Figure 7E). Thus,

411

HMC inhibited pro-inflammatory cytokines production and also modulated in a different manner

412

the levels the anti-inflammatory cytokines, reducing IL-10 and enhancing TGF-β.

413

3.7. HMC inhibits the activity of NF-κB in the knee joint after MSU injection. Using

414 415

ELISA kits, we observed that MSU decreased the total NF-κB p65 subunit/phosphorylated NF-κB

416

p65 subunit ratio indicating NF-κB activation, which was inhibited by HMC treatment (41%)

417

(Figure 8). Thus, HMC inhibited the activation of NF-κB induced by MSU injection in the knee

418

joint.

419 420

3.8. HMC inhibits MSU-induced increase of inflammasome components expression in

421

the knee joint. MSU stimulus increased mRNA expression of inflammasome components

422

(NLRP3, ASC, pro-caspase-1, and pro-IL-1β) compared to saline control group (Figure 9A-D,

423

respectively). In turn, HMC treatment inhibited the expression of all components of inflammasome

424

(NLRP3 by 72%, ASC by 77%, pro-caspase-1 by 71%, and pro-IL-1β by 73%) (Figure 9A-D,

17 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

425

respectively). Thus, HMC inhibited the MSU-induced increase of inflammasome platform

426

components mRNA expression.

427 428

3.9. HMC does not inhibit the release of mature IL-1β by BMDMs induced by MSU.

429

Stimulation of BMDMs with LPS (priming, signal 1) followed by MSU (signal 2) resulted in

430

increased levels of IL-1β in the supernatant, and this effect was not inhibited by HMC treatment

431

(30-300 µM; Figure 10). On the other hand, treatment of these cells with the positive control

432

quercetin (30 µM) inhibited IL-1β secretion induced by MSU stimulation (Figure 10). The

433

quercetin result is a control demonstrating that this system works properly.18 None of the treatments

434

affected the LDH levels in the culture supernatant, which indicates that cell viability was not

435

affected (data not shown). Isolated stimulation of BMDMs with LPS or MSU was not able to

436

promote increases in supernatant levels of mature IL-1β (data not shown). Thus, HMC could not

437

inhibit the activity of inflammasome in BMDMs cells stimulated with MSU in vitro, indicating that

438

directly targeting inflammasome activation is not a mechanism of action of HMC in gout arthritis.

439 440 441 442 443 444 445 446 447 448 449

18 ACS Paragon Plus Environment

Page 18 of 48

Page 19 of 48

450

Journal of Agricultural and Food Chemistry

4. DISCUSSION

451 452

HMC is a safe drug with therapeutic applications for vascular diseases. HMC improves

453

vascular tonus and reduces symptoms of acute hemorrhoid and other venous diseases.33,34 Further,

454

evidence shows that HMC has anti-inflammatory and analgesic properties.10,22,35 In this sense,

455

repurpusing of HMC for treatment of pain and inflammation seems reasonable. The present data

456

support the potential of HMC to inhibit inflammation and pain in an animal model of gout arthritis

457

by blocking NF-kB activation without directly affecting inflammasome activation. The lack of

458

effect of HMC over inflammasome activation indicates that HMC would present an additive effect

459

with the current IL-1ra therapy since these molecules act by distinct though complementary

460

mechanisms.

461

Although HMC inhibitory effects upon inflammatory pain have already been demonstrated

462

by our group using classical models of pain such as formalin and carrageenan22, this MSU intra-

463

articular injection model differs from previously used models since represents specifically the

464

physiopathological mechanisms and therapeutic targets of gout arthritis. Oral treatment with HMC

465

dose-dependently inhibited mechanical hyperalgesia, joint edema, and recruitment of leukocytes

466

induced by intra-articular MSU. Thus, HMC consistently inhibited MSU clinical inflammatory

467

signs. HMC also inhibited MSU-induced neutrophil recruitment and synovitis that are important

468

parameters indicating reduction of tissue lesion. Activation of tissue resident macrophages and the

469

recruitment of other leukocytes (e.g. neutrophils) represent an important mechanism of

470

pathogenesis in acute gout. Leukocytes actively contribute to the gout disease36 by releasing varied

471

mediators including ROS, proteolytic enzymes and pro-inflammatory cytokines (e.g. TNFα, IL-1β),

472

which contribute to cartilage degradation and joint damage.11,12,37 Here, the treatment with HMC

473

reduced the accumulation of neutrohils into the joint, as identified by morphology and

474

immunofluorescence using LysM-eGFP+ mice. Macrophages also express LysM, but to a lesser

19 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

475

extent than neutrophils.27 Therefore, the HMC inhibition of leukocyte infiltration, joint edema, pain

476

and synovitis demonstrates the therapeutic potential of this flavonoid.

477

Active NADPH oxidase in phagocytes produces large quantities of superoxide anion

478

generating various ROS that target proteins, lipids and nucleic acids.14 Nitric oxide (NO) is also

479

produced and contributes to arthritis by reacting with superoxide anion generating peroxynitrite that

480

causes tissue lesion.38 Synovial tissue from gout arthritis patients exhibits an increase of NO

481

production, and MSU crystals increases NO production by monocytes/macrophages of gout

482

patients.39 The excessive production of ROS causes depletes GSH and promotes oxidation of

483

cysteine in proteins, resulting in oxidative stress-induced lesioning of tissues. ROS also activate

484

NF-κB and NLRP3 inflammasome signaling pathways, which are crucial in gout arthritis

485

pathogenesis.14 Cytokines and oxidative stress contribute to edema40, leukocyte recruitment41, and

486

further pro-inflammatory cytokines production and pain.42 HMC inhibited MSU-induced oxidative

487

stress and re-established endogenous antioxidant levels and activity. The antioxidant effects of

488

HMC were not solely explained by its structural antioxidant chemical groups.20 HMC also induced

489

Nrf2 mRNA expression, an important transcription factor that induces HO-1 and GSH43 lining up

490

with the results of enhanced HO-1 mRNA expression and GSH levels by HMC treatment. Hence,

491

HMC inhibited gp91phox mRNA expression (NADPH oxidase) and the formation of its product

492

superoxide anion. Therefore, HMC has antioxidant chemical structures20 and up-regulates

493

endogenous antioxidant and anti-inflammatory mechanisms.

494

Pro-inflammatory cytokines present a pivotal role in gout.37 HMC inhibited MSU-induced

495

pro-inflammatory cytokines production (TNFα, IL-1β and IL-6). These cytokines induce the

496

activation of NADPH oxidase and the production of superoxide anion, NO and other ROS.10

497

Furthermore, these cytokines induce pain, edema and leukocyte recruitment.44,45 Therefore, the

498

HMC inhibition of MSU-induced cytokine production accounted to reducing oxidative stress, pain,

499

edema and leukocyte recruitment. The production of the anti-inflammatory cytokine IL-10 is

20 ACS Paragon Plus Environment

Page 20 of 48

Page 21 of 48

Journal of Agricultural and Food Chemistry

500

concomitant with the production of pro-inflammatory cytokines, which helps to limit their

501

production and activity.46 Flavonoids have been shown to induce IL-10 production, which explains

502

in part the anti-inflammatory actions of these molecules.47 However, HMC did not induce an

503

increase of IL-10, but rather reduced its production. It is likely that as HMC inhibited the pro-

504

inflammatory cytokines production, IL-10 release was also reduced since its limiting actions over

505

inflammatory cytokines were not necessary.35 Furthermore, these results show that not all

506

flavonoids act by the same mechanisms. MSU-induced knee joint inflammation was also

507

accompanied by a slight non-significant increase of TGF-β production. HMC treatment enhanced

508

MSU-induced TGF-β production. TGF-β increases the phagocytosis of apoptotic neutrophils by

509

monocytes/macrophages. This clearance of cell debris is very important for resolution of

510

inflammation.48 Therefore, the increase of TGF-β production in response to HMC administration

511

may be a contributing mechanism to its anti-inflammatory and analgesic effect.

512

NF-κB and NLRP3 inflammasome signaling pathways have pivotal roles in gout.13,49

513

Activation of the transcription factor NF-κB increases the expression of cytokines and enzymes that

514

produce ROS.12,22,50 Cytokines and ROS, in turn, further activate NF-κB, and this reciprocal circle

515

amplifies the inflammatory response.51 HMC inhibited the pro-inflammatory cytokines production

516

and the oxidative stress induced by MSU. Accordingly, HMC inhibited MSU-induced NF-κB

517

activation in the knee joint, thus, HMC targets an essential transcription factor in the inflammatory

518

response explaining the prominent effect of this flavonoid in reducing MSU-induced knee joint

519

inflammation and pain. Activation of NF-κB also induces the expression of the components of

520

NLRP3 inflammasome (NLRP3, ASC, pro-caspase-1, and pro-IL-1β).16,52 In agreement with the

521

HMC inhibition of NF-κB activation, this flavonoid reduced NLRP3, ASC, pro-caspase-1, and pro-

522

IL-1β mRNA expression induced by MSU in vivo. This result is important since it demonstrates

523

that HMC reduces the up-regulation of an inflammatory platform that is essential in gout

524

pathogenesis. 21 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Flavonoids such as quercetin inhibit ASC oligomerization and the activation of

525 526

inflammasome, which contributes to reduce inflammasome-dependent inflammation in a model of

527

vasculitis18 and gout arthritis.12 Therefore, we reasoned that it was rational to evaluate whether

528

HMC would also inhibit inflammasome activation. The flavonoid quercetin decreased secretion of

529

IL-1β by MSU-stimulated, LPS-primed BMDMs, corroborating previous data18 and that this model

530

was working properly in our experimental conditions. However, HMC did not affect MSU-induced

531

inflammasome-dependent IL-1β secretion. Therefore, inhibiting MSU-induced NLRP3 activation is

532

not a mechanism of action of HMC. Other studies with various inflammasome activators are

533

necessary to investigate the extent of inflammasome inhibition by flavonoids, but our results do

534

suggest that not all flavonoids inhibit inflammasome activation, and therefore, not all flavonoids

535

have the same mechanism of action. Further studies addressing structure-relationship of flavonoids

536

and inflammasome, and also applying other inflammasome activators will be necessary to

537

understand these differences in greater detail.

538

In conclusion, our study demonstrates that HMC inhibits MSU-induced acute knee joint

539

inflammation and pain. The mechanism of action of HMC is not dependent on inhibiting MSU

540

crystals-induced inflammasome activation, but rather it depends on inhibiting NF-κB activation and

541

induces Nrf2/HO-1 pathway. As a consequence of these effects, HMC inhibits the recruitment of

542

cells and production of ROS and cytokines, effects that explain its anti-inflammatory and analgesic

543

effects.

544 545 546 547 548 549

22 ACS Paragon Plus Environment

Page 22 of 48

Page 23 of 48

Journal of Agricultural and Food Chemistry

550

AUTHOR INFORMATION

551

Corresponding author

552

*Phone: + 55 43 33714979. E-mail: [email protected]; [email protected].

553

Authors contributions

554

R.C. and W.A.V.J. designed the study. K.W.R.M., F.A.P.R., S.M.B., L.S.F., V.F., F.A.A., M.M.T.,

555

J.C.A.F., T.M.C., F.Q.C., R.C. and W.A.V.J. planned experiments and analyze the data. K.W.R.M.,

556

F.A.P.R., S.M.B., L.S.F., and V.F. performed the experiments. F.A.A., M.M.T., J.C.A.F., T.M.C.

557

and F.Q.C. provided essential materials. K.W.R.M., F.A.P.R., S.M.B., R.C. and W.A.V.J. wrote the

558

manuscript. W.A.V.J. supervised the study. All authors read and approved the manuscript.

559 560

Acknowledgment

561

This work was supported by grants from Coordenadoria de Aperfeiçoamento de Pessoal de Nível

562

Superior (CAPES), Financiadora de Estudo e Projetos–Apoio à Infraestrutura (CT-INFRA 01/2011;

563

process 01.13.0049.00), Central Multiusuária de Laboratórios de Pesquisa da UEL (CMLP-UEL),

564

São Paulo Research Foundation (FAPESP), Center for Research on Inflammatory Diseases (CRID),

565

Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Pesquisa para o Sistema

566

Único de Saúde (PPSUS) grant supported by Ministério da Ciência, Tecnologia e Inovação (MCTI),

567

Secretaria da Ciência, Tecnologia e Ensino Superior (SETI), Decit/SCTIE/MS through CNPq with

568

the support of Fundação Araucária and Secretaria da Saúde do Estado do Paraná (SESA-PR), and

569

Parana State Government (Brazil).

570 571

Notes

572

There are no competing financial interests to be declared.

573 574

23 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

575

References

576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622

(1) (2)

(3) (4) (5)

(6) (7) (8)

(9)

(10)

(11)

(12)

(13) (14) (15) (16)

(17) (18)

Martinon, F.; Glimcher, L. H. Gout: new insights into an old disease. J. Clin. Invest. 2006, 116 (8), 2073–2075. Zhu, Y.; Pandya, B. J.; Choi, H. K. Prevalence of gout and hyperuricemia in the US general population: the national health and nutrition examination survey 2007-2008. Arthritis Rheum. 2011, 63 (10), 3136–3141. Kuo, C.-F.; Grainge, M. J.; Zhang, W.; Doherty, M. Global epidemiology of gout: prevalence, incidence and risk factors. Nat. Rev. Rheumatol. 2015, 11 (11), 649–662. Hak, A. E.; Curhan, G. C.; Grodstein, F.; Choi, H. K. Menopause, postmenopausal hormone use and risk of incident gout. Ann. Rheum. Dis. 2010, 69 (7), 1305–1309. Paschke, S.; Weidner, A. F.; Paust, T.; Marti, O.; Beil, M.; Ben-Chetrit, E. Technical Advance: Inhibition of neutrophil chemotaxis by colchicine is modulated through viscoelastic properties of subcellular compartments. J. Leukoc. Biol. 2013, 94 (5), 1091– 1096. Cronstein, B. N.; Terkeltaub, R. The inflammatory process of gout and its treatment. Arthritis Res. Ther. 2006, 8 Suppl 1 (Suppl 1), S3. Neogi, T. Clinical practice. Gout. N. Engl. J. Med. 2011, 364 (5), 443–452. Guerne, P. A.; Terkeltaub, R.; Zuraw, B.; Lotz, M. Inflammatory microcrystals stimulate interleukin-6 production and secretion by human monocytes and synoviocytes. Arthritis Rheum. 1989, 32 (11), 1443–1452. di Giovine, F. S.; Malawista, S. E.; Thornton, E.; Duff, G. W. Urate crystals stimulate production of tumor necrosis factor alpha from human blood monocytes and synovial cells. Cytokine mRNA and protein kinetics, and cellular distribution. J. Clin. Invest. 1991, 87 (4), 1375–1381. Manchope, M. F.; Calixto-Campos, C.; Coelho-Silva, L.; Zarpelon, A. C.; Pinho-Ribeiro, F. A.; Georgetti, S. R.; Baracat, M. M.; Casagrande, R.; Verri, W. A. Naringenin inhibits superoxide anion-induced inflammatory pain: role of oxidative stress, cytokines, Nrf-2 and the NO−cGMP−PKG−KATP channel signaling pathway. PLoS One 2016, 11 (4), e0153015. Zamudio-Cuevas, Y.; Hernández-Díaz, C.; Pineda, C.; Reginato, A. M.; Cerna-Cortés, J. F.; Ventura-Ríos, L.; López-Reyes, A. Molecular basis of oxidative stress in gouty arthropathy. Clin. Rheumatol. 2015, 34 (10), 1667–1672. Ruiz-Miyazawa, K. W.; Staurengo-Ferrari, L.; Mizokami, S. S.; Domiciano, T. P.; Vicentini, F. T. M. C.; Camilios-Neto, D.; Pavanelli, W. R.; Pinge-Filho, P.; Amaral, F. A.; Teixeira, M. M.; et al. Quercetin inhibits gout arthritis in mice: induction of an opioid-dependent regulation of inflammasome. Inflammopharmacology 2017, 1–16. Martinon, F.; Pétrilli, V.; Mayor, A.; Tardivel, A.; Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006, 440 (7081), 237–241. Martinon, F. Signaling by ROS drives inflammasome activation. Eur. J. Immunol. 2010, 40 (3), 616–619. Schroder, K.; Tschopp, J. The Inflammasomes. Cell 2010, 140 (6), 821–832. Bauernfeind, F. G.; Horvath, G.; Stutz, A.; Alnemri, E. S.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B. G.; Fitzgerald, K. A.; et al. Cutting edge: NFkappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J. Immunol. 2009, 183 (2), 787–791. Rock, K. L.; Kataoka, H.; Lai, J.-J. Uric acid as a danger signal in gout and its comorbidities. Nat. Rev. Rheumatol. 2013, 9 (1), 13–23. Domiciano, T. P.; Wakita, D.; Jones, H. D.; Crother, T. R.; Verri, W. A.; Arditi, M.; 24 ACS Paragon Plus Environment

Page 24 of 48

Page 25 of 48

623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672

Journal of Agricultural and Food Chemistry

(19) (20)

(21)

(22)

(23) (24) (25) (26)

(27)

(28)

(29) (30)

(31)

(32)

(33)

Shimada, K. Quercetin inhibits inflammasome activation by interfering with asc oligomerization and prevents interleukin-1 mediated mouse vasculitis. Sci. Rep. 2017, 7, 41539. Garg, A.; Garg, S.; Zaneveld, L. J.; Singla, A. K. Chemistry and pharmacology of the Citrus bioflavonoid hesperidin. Phytother. Res. 2001, 15 (8), 655–669. Verri, W. A.; Vicentini, F. T. M. C.; Baracat, M. M.; Georgetti, S. R.; Cardoso, R. D. R.; Cunha, T. M.; Ferreira, S. H.; Cunha, F. Q.; Fonseca, M. J. V.; Casagrande, R. Flavonoids as anti-inflammatory and analgesic drugs: mechanisms of action and perspectives in the development of pharmaceutical forms. In Studies in Natural Products Chemistry; Elsevier, Ed.; Studies in Natural Products Chemistry; Elsevier: Amsterdam, 2012; Vol. 36, pp 297– 330. Li, R.; Cai, L.; Xie, X.; Peng, L.; Wu, T.; Li, J. 7,3′-dimethoxy hesperetin inhibits inflammation by inducing synovial apoptosis in rats with adjuvant-induced arthritis. Immunopharmacol. Immunotoxicol. 2013, 35 (1), 1–8. Pinho-Ribeiro, F. A.; Hohmann, M. S. N. N.; Borghi, S. M.; Zarpelon, A. C.; Guazelli, C. F. S. S.; Manchope, M. F.; Casagrande, R.; Verri, W. A. Protective effects of the flavonoid hesperidin methyl chalcone in inflammation and pain in mice: Role of TRPV1, oxidative stress, cytokines and NF-κB. Chem. Biol. Interact. 2015, 228, 88–99. Walle, T. Methylation of dietary flavones greatly improves their hepatic metabolic stability and intestinal absorption. Mol. Pharm. 2007, 4 (6), 826–832. Chen, Q. M.; Feng, G. H. Vasodilating action of methylhesperidin. Zhongguo Yao Li Xue Bao 1987, 8 (4), 344–348. Stoianova, V. [Cyclo 3 fort--alternative in chronic venous insufficiency]. Akusherstvo i Ginekol. 2006, 45 Suppl 3, 78–80. Allaert, F. A.; Hugue, C.; Cazaubon, M.; Renaudin, J. M.; Clavel, T.; Escourrou, P. Correlation between improvement in functional signs and plethysmographic parameters during venoactive treatment (Cyclo 3 Fort). Int. Angiol. 2011, 30 (3), 272–277. Wenzel, P.; Knorr, M.; Kossmann, S.; Stratmann, J.; Hausding, M.; Schuhmacher, S.; Karbach, S. H.; Schwenk, M.; Yogev, N.; Schulz, E.; et al. Lysozyme m-positive monocytes mediate angiotensin II-induced arterial hypertension and vascular dysfunction. Circulation 2011, 124 (12), 1370–1381. Lima-Junior, D. S.; Costa, D. L.; Carregaro, V.; Cunha, L. D.; Silva, A. L. N.; Mineo, T. W. P.; Gutierrez, F. R. S.; Bellio, M.; Bortoluci, K. R.; Flavell, R. A.; et al. Inflammasomederived IL-1β production induces nitric oxide-mediated resistance to Leishmania. Nat. Med. 2013, 19 (7), 909–915. So, A.; Thorens, B. Uric acid transport and disease. J. Clin. Invest. 2010, 120 (6), 1791– 1799. Amaral, F. A.; Costa, V. V; Tavares, L. D.; Sachs, D.; Coelho, F. M.; Fagundes, C. T.; Soriani, F. M.; Silveira, T. N.; Cunha, L. D.; Zamboni, D. S.; et al. NLRP3 inflammasomemediated neutrophil recruitment and hypernociception depend on leukotriene B4 in a murine model of gout. Arthritis Rheum. 2012, 64 (2), 474–484. Vieira, A. T.; Macia, L.; Galvão, I.; Martins, F. S.; Canesso, M. C. C.; Amaral, F. A.; Garcia, C. C.; Maslowski, K. M.; De Leon, E.; Shim, D.; et al. A Role for Gut Microbiota and the Metabolite-Sensing Receptor GPR43 in a Murine Model of Gout. Arthritis Rheumatol. 2015, 67 (6), 1646–1656. Clausen, B. E.; Burkhardt, C.; Reith, W.; Renkawitz, R.; Förster, I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 1999, 8 (4), 265–277. Guex, J. J.; Enriquez Vega, D. M. E.; Avril, L.; Boussetta, S.; Taïeb, C. Assessment of quality of life in Mexican patients suffering from chronic venous disorder - impact of oral 25 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722

(34)

(35)

(36)

(37) (38)

(39)

(40)

(41)

(42)

(43)

(44) (45)

(46) (47)

(48)

Ruscus aculeatus-hesperidin-methyl-chalcone-ascorbic acid treatment - “Quality study.” Phlebology 2009, 24 (4), 157–165. Guex, J. J.; Avril, L.; Enrici, E.; Enriquez, E.; Lis, C.; Taïeb, C. Quality of life improvement in Latin American patients suffering from chronic venous disorder using a combination of Ruscus aculeatus and hesperidin methyl-chalcone and ascorbic acid (quality study). Int. Angiol. 2010, 29 (6), 525–532. Martinez, R. M.; Pinho-Ribeiro, F. A.; Steffen, V. S.; Caviglione, C. V; Pala, D.; Baracat, M. M.; Georgetti, S. R.; Verri, W. A.; Casagrande, R. Topical formulation containing hesperidin methyl chalcone inhibits skin oxidative stress and inflammation induced by ultraviolet B irradiation. Photochem. Photobiol. Sci. 2016, 15 (4), 554–563. Terkeltaub, R.; Baird, S.; Sears, P.; Santiago, R.; Boisvert, W. The murine homolog of the interleukin-8 receptor CXCR-2 is essential for the occurrence of neutrophilic inflammation in the air pouch model of acute urate crystal-induced gouty synovitis. Arthritis Rheum. 1998, 41 (5), 900–909. Busso, N.; So, A. Mechanisms of inflammation in gout. Arthritis Res. Ther. 2010, 12 (2), 206. Farrell, A. J.; Blake, D. R.; Palmer, R. M.; Moncada, S. Increased concentrations of nitrite in synovial fluid and serum samples suggest increased nitric oxide synthesis in rheumatic diseases. Ann. Rheum. Dis. 1992, 51 (11), 1219–1222. Chen, L.; Hsieh, M.-S.; Ho, H.-C.; Liu, Y.-H.; Chou, D.-T.; Tsai, S.-H. Stimulation of inducible nitric oxide synthase by monosodium urate crystals in macrophages and expression of iNOS in gouty arthritis. Nitric Oxide 2004, 11 (3), 228–236. Serafim, K. G. G.; Navarro, S. A.; Zarpelon, A. C.; Pinho-Ribeiro, F. A.; Fattori, V.; Cunha, T. M.; Alves-Filho, J. C.; Cunha, F. Q.; Casagrande, R.; Verri, W. A. Bosentan, a mixed endothelin receptor antagonist, inhibits superoxide anion-induced pain and inflammation in mice. Naunyn. Schmiedebergs. Arch. Pharmacol. 2015, 388 (11), 1211–1221. Hattori, H.; Subramanian, K. K.; Sakai, J.; Jia, Y.; Li, Y.; Porter, T. F.; Loison, F.; Sarraj, B.; Kasorn, A.; Jo, H.; et al. Small-molecule screen identifies reactive oxygen species as key regulators of neutrophil chemotaxis. Proc. Natl. Acad. Sci. U. S. A. 2010, 107 (8), 3546– 3551. Wang, Z.-Q. Q.; Porreca, F.; Cuzzocrea, S.; Galen, K.; Lightfoot, R.; Masini, E.; Muscoli, C.; Mollace, V.; Ndengele, M.; Ischiropoulos, H.; et al. A newly identified role for superoxide in inflammatory pain. J. Pharmacol. Exp. Ther. 2004, 309 (3), 869–878. Bryan, H. K.; Olayanju, A.; Goldring, C. E.; Park, B. K. The Nrf2 cell defence pathway: Keap1-dependent and -independent mechanisms of regulation. Biochem. Pharmacol. 2013, 85 (6), 705–717. Zhang, J.-M.; An, J. Cytokines, inflammation, and pain. Int. Anesthesiol. Clin. 2007, 45 (2), 27–37. Cunha, T. M.; Verri, W. A.; Valério, D. A.; Guerrero, A. T.; Nogueira, L. G.; Vieira, S. M.; Souza, D. G.; Teixeira, M. M.; Poole, S.; Ferreira, S. H.; et al. Role of cytokines in mediating mechanical hypernociception in a model of delayed-type hypersensitivity in mice. Eur. J. Pain 2008, 12 (8), 1059–1068. Iyer, S. S.; Cheng, G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit. Rev. Immunol. 2012, 32 (1), 23–63. Leyva-López, N.; Gutierrez-Grijalva, E. P.; Ambriz-Perez, D. L.; Basilio Heredia, J. Flavonoids as cytokine modulators: A possible therapy for inflammation-related diseases. International Journal of Molecular Sciences. Multidisciplinary Digital Publishing Institute (MDPI) June 9, 2016, p 921. Huynh, M.-L. N.; Fadok, V. A.; Henson, P. M. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J. Clin. 26 ACS Paragon Plus Environment

Page 26 of 48

Page 27 of 48

723 724 725 726 727 728 729 730 731 732 733 734 735 736 737

Journal of Agricultural and Food Chemistry

(49)

(50) (51)

(52)

Invest. 2002, 109 (1), 41–50. Jaramillo, M.; Naccache, P. H.; Olivier, M. Monosodium urate crystals synergize with IFNgamma to generate macrophage nitric oxide: involvement of extracellular signal-regulated kinase 1/2 and NF-kappa B. J. Immunol. 2004, 172 (9), 5734–5742. Niederberger, E.; Geisslinger, G. The IKK-NF-kappaB pathway: a source for novel molecular drug targets in pain therapy? FASEB J. 2008, 22 (10), 3432–3442. Bowie, A.; O’Neill, L. A. Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem. Pharmacol. 2000, 59 (1), 13–23. Dhanasekar, C.; Rasool, M. Morin, a dietary bioflavonol suppresses monosodium urate crystal-induced inflammation in an animal model of acute gouty arthritis with reference to NLRP3 inflammasome, hypo-xanthine phospho-ribosyl transferase, and inflammatory mediators. Eur. J. Pharmacol. 2016, 786, 116–127.

738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754

27 ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

755

Figures caption

756 757

Figure 1. Schematic representation of in vivo (a) and in vitro (b) protocols used in the study.

758 759

Figure 2. HMC inhibits MSU-induced articular mechanical hyperalgesia and edema. Mice were

760

treated with HMC (3, 10 or 30 mg/kg/saline, p.o.) or vehicle (saline) 1 h before MSU (100 µg/10

761

µL) i.a injection. Mechanical hyperalgesia (A) and edema (B) were assessed 1-15 h after MSU

762

administration using an electronic pressure test and a caliper, respectively. Results are presented as

763

means ± SEM of 6 mice per group per experiment and are representative of 2 independent

764

experiments. *p < 0.05 compared to saline group; #p < 0.05 compared to MSU + vehicle group; ƒp