Polyphenols from Blossoms of Citrus aurantium ... - ACS Publications

Then the lysis was centrifuged at 12 000 g for 20 min at 4 °C and the supernatants were collected as whole cell lysates. Furthermore, the cytoplasmic...
3 downloads 13 Views 686KB Size
Subscriber access provided by PEPPERDINE UNIV

Article

Polyphenols from blossoms of Citrus aurantium L. var. amara Engl. show significant anti-complement and anti-inflammatory effects Chun-Yan Shen, Jian-Guo Jiang, Chun-Ling Huang, Wei Zhu, and Chao-Yang Zheng J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.7b03759 • Publication Date (Web): 24 Sep 2017 Downloaded from http://pubs.acs.org on September 28, 2017

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 free 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 accessible to all readers and 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.

Journal of Agricultural and Food Chemistry 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 24

Journal of Agricultural and Food Chemistry

1

Polyphenols from blossoms of Citrus aurantium L. var. amara Engl.

2

show significant anti-complement and anti-inflammatory effects

3

4

Chun-Yan Shen †, Jian-Guo Jiang *†, Chun-Ling Huang § ⊗, Wei Zhu § , Chao-Yang Zheng *§

5



6

510640, China

7

§

8

510120, China

9



College of Food and Bioengineering, South China University of Technology, Guangzhou,

The second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou

Sci-tech Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou 510120, China

10 11

*Author (Jian-Guo Jiang) for correspondence (e-mail: [email protected]; phone +86-20-

12

87113849; fax: +86-20-87113843)

13

*Author (Chao-Yang Zheng) for correspondence ([email protected]; phone +86-20-

14

39318571; fax: +86-20-39318571)

15 16

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

17

Abstract

18

Citrus aurantium L. var. amara Engl. (CAVA) was traditionally used as a digestant or expectorant

19

in China. Crude polyphenols (CAVAP-W) extracted from blossoms of CAVA were mainly

20

composed of eriocitrin/neoeriocitrin, eriocitrin/neoeriocitrin, rhoifolin, hesperidin, naringin,

21

rutin, veronicastroside, neohesperidin and hesperetin by LC-MS analysis. CAVAP-W showed

22

significant anti-complement and anti-inflammatory effects. Due to the close relationship between

23

anti-complement and anti-inflammatory activity, the anti-inflammatory effect was further

24

investigated and the results showed that CAVAP-W significantly suppressed production of

25

interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β), and mRNA

26

expression of inducible nitric oxide synthase (iNOS), IL-6, TNF-α, IL-1β and cyclooxygenase-2

27

(COX-2) in lipopolysaccharides-stimulated RAW264.7 cells. Furthermore, CAVAP-W inhibited

28

mitogen-activated protein kinase (MAPK) phosphorylation and NF-κB activation through

29

suppressing nuclear translocation of nuclear factor-kappa B (NF-κB) P65, degradation and

30

phosphorylation of IκBα, phosphorylation of IκKBɑ/ß, c-Jun N-terminal kinase (JNK) and P38,

31

and activation of COX-2, thereby exerting the anti-inflammatory effects.

32 33

Key words: Citrus aurantium L. var. amara Engl.; polyphenolys; LC-MS; anti-complement; anti-

34

inflammatory

35

2

ACS Paragon Plus Environment

Page 2 of 24

Page 3 of 24

Journal of Agricultural and Food Chemistry

36

INTRODUCTION

37

Complement system, playing a critical role in the host defense, had various beneficial

38

biological activities, such as protecting against infectious agents and opsonizing anti-genic

39

particles. However, excessive activation of complement could also induce severe tissue damage

40

and various inflammation associated diseases. Inflammation was a complex process regulated by

41

many pro-inflammatory mediators and their excessive production might lead to various acute and

42

chronic inflammatory diseases including atherosclerosis, rheumatoid arthritis and even cancer.1-3

43

There is growing acceptance that active ingredients extracted from natural products usually

44

exert various beneficial effects such as anti-aging, immunomodulatory, anti-inflammatory and

45

anti-cancer, with few side effects.4-7 Specifically, polyphenols are a major class of phytochemicals

46

found in natural products and receiving much attention owing to their great contributions to

47

human health. Especially, a growing number of published data focused on the anti-inflammatory

48

effects of Citrus polyphenols.8-11 For example, Kang et al. found that the flavonoids extracted

49

from Korea Citrus aurantium L. displayed significant anti-inflammatory effects on LPS-induced

50

RAW264.7 cells by suppressing mitogen-activated protein kinase (MAPK) and nuclear factor-

51

kappa B (NF-κB) signaling pathways.10

52

Citrus aurantium L. var. amara Engl. (CAVA), a member of genus Citrus (Rutaceae), was

53

widely used as edible and medicinal resource nowadays in China. It was traditionally used as a

54

digestant and expectorant in China. Also, blossoms of CAVA were claimed to be a popular kind

55

of beverage. Our published reports

56

oil, flavonoids and polysaccharides extracted from blossoms of CAVA possessed various

57

pharmacological activities.

12-14

showed that many active ingredients including essential

58

Nevertheless, information about possible bioactivities of its polyphenols was rather limited.

59

Therefore, this study was undertaken to investigate the anti-complement and anti-inflammatory

60

activities of polyphenols from CAVA as well as explore the underlying molecular mechanism.

61

Simultaneously, high performance liquid chromatography-mass spectrometry (LC-MS) analysis of

62

CAVAP-W was also performed in order to speculate the specific constituents responsible for the

63

observed bioactivity.

3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

64

MATERIALS AND METHODS

65

Materals and Chemicals

66

Blossoms of Citrus aurantium L. var. amara Engl. were collected in the year of 2016 from

67

Zhejiang Province in China. The blossoms were washed, drained, freeze-dried and powdered with

68

a grinder and then packed into sealed containers for further use. Dulbecco’s modified eagle

69

medium (DMEM), fetal bovine serum (FBS), Trizol Reagent and Penicillin G/streptomycin were

70

purchased from GIBCO (Grand Island, NY). 3-(4, 5-Dimethyl-2-thiazolyl)-2, 5-diphenyl-2H-

71

tetrazolium bromide (MTT) and lipopolysaccharides (LPS) were obtained from Sigma-Aldrich (St.

72

Louis, MO, USA). NF-κB activation-nuclear translocation assay kit and the nuclear and

73

cytoplasmic protein extraction kit were purchased from Beyotime Biotech (Guangzhou, China).

74

ELISA kits of mouse interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α) and interleukin-1β

75

(IL-1β) were purchased from Cusabio Biothch CO., Ltd. (Wuhan, China). The antibodies to

76

GAPDH, phospho-SAPK/JNK, phospho-P38 MAPK, NF-κB P65, IκBα, phospho-IκKα/ß and

77

cyclooxygenase-2 (COX-2) were all purchased from Cell Signaling Technology (Beverly, MA,

78

USA). Acetonitrile was HPLC-grade and all other reagents used in this research were of analytical

79

grade.

80

Extraction of Polyphenols

81

Processed powders (5.0 g) of CAVA blossoms were extracted at 90 oC for 1 h with 100 mL of

82

distilled water. The resulting extracts were filtered through filter papers and evaporated under

83

reduced pressure using a rotary vacuum evaporator at 40 oC. The dried samples were eventually

84

obtained and named as CAVAP-W.

85

Determination of Total Polyphenols Contents

86

The dried samples (CAVAP-W) obtained in 2.2 were used for determining total phenolic

87

contents according to the published method.15 Aliquots (0.1, 0.2, 0.4, 0.6, 0.8 and 1.0 mL) of the

88

sample solutions were taken in separated test tubes and made up to the volume of 1 mL with

89

distilled water. Then 0.5 mL of Folin–Ciocalteu reagent and 2.5 mL of saturated sodium carbonate

90

solution were added sequentially to each tube. After vigorous vortex, the tubes containing the

91

reaction mixture were placed in the dark for 40 min. Eventually, the absorbance at 725 nm was

4

ACS Paragon Plus Environment

Page 4 of 24

Page 5 of 24

Journal of Agricultural and Food Chemistry

92

recorded against the blank. Then the total phenolic contents were calculated as gallic acid

93

equivalents according to the calibration curve.

94

LC-MS Analysis

95 96

In order to determine the compounds in CAVAP-W, LC-MS analysis was conducted using Agilent Technology 6130 Quadrupole LC/MS equipment based on previous reports.16-18

97

The chromatographic separation of CAVAP-W was carried out by HPLC using a Waters

98

AQUITY UPLC CSH C18 column (2.1×100 mm, 1.7 µm). The mobile phases were 0.1% aqueous

99

formic acid (eluent A) and acetonitrile (eluent B). The eluent gradient applied for solvent B was as

100

follows: 0.01-8 min, 2 %-20% B; 8-13 min, 20%-25% B; 13-15 min, 25%-25% B; 15-22 min,

101

25%-50% B; 22-25 min, 50%-95% B; 25-28 min, 95%-2% B; 28-30 min, 2% B, at a flow rate of

102

0.3 mL/min. The column temperature was 40 oC and the injection volume was 10 µL. MS

103

operation parameters were set as follows: capillary voltage, 4000 V; nebulizer pressure, 40 psi;

104

drying gas flow rate, 10 L/min; gas temperature, 500 oC. And the electrospray mass spectra

105

ranging from m/z 100 to 1000 were taken.

106

Anti-complement Activity Assay

107

In the current research, distilled water was employed to resuspend CAVAP-W. anti-

108

complement activity of CAVAP-W was investigated using the classical pathway according to Seo

109

et al. with slight modifications.19 Normal human serum was used as the complement source and

110

heparin sodium was used as the positive control.

111

Anti-inflammatory Activity Assay

112

Cell Culture and Treatment

113

Murine macrophage RAW264.7 cell line, human hepatocyte cells LO2 were purchased from

114

cell bank of the Chinese Academy of Sciences in Shanghai of China and cultured in DMEM

115

supplemented with 10% FBS at 37 oC in a 5% CO2 humidified atmosphere. CAVAP-W was

116

dissolved in dimethyl sulfoxide (DMSO), filtered through 0.25 µm filter membrane, and stored at -

117

20 °C for future use. During the experiment, the samples were diluted in DMEM to various

118

working concentrations varying from 15.625 to 500 µg/mL.

119

Assay of Cell Viability

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

120

Aliquot of 100 µL cells (LO2 and RAW264.7) were seeded into 96-well plates at a

121

concentration of 7×104 cells/mL and incubated for 24 h to make the cells adherent. Then the

122

supernates were removed and fresh medium containing different concentrations of CAVAP-W

123

(31.25, 62.5, 125, 250 and 500 µg/mL) were added. After incubation for 24 h, the supernates were

124

discarded and 200 µL of MTT solution (0.5 mg/mL) diluted in fresh DMEM without FBS was

125

added. 4 h later, MTT solutions were removed and the adherent cells were washed twice with PBS.

126

Then 150 µL of DMSO was carefully added into each well to complete dissolve the purple

127

formazan crystals. Finally, the absorbance of 570 nm was detected using a microplate reader.

128

Determinaton of NO Production and Cytokines Secretion

129

Production of NO was determined based on the accumulation of nitrite in the culture

130

supernatants using the protocol described by our previous report.7 Briefly, 1 mL of RAW264.7

131

cells at a density of 3×105 cells/mL were seeded into each well of 24-well plates and stimulated

132

with culture medium or LPS (1 µg/mL) or LPS in the presence of different concentrations of

133

CAVAP-W (15.625, 31.25, 62.5, 125, 250 and 500 µg/mL) for 24 h. Then the culture supernatants

134

were collected and incubated with equal volume of Griess reagents for 10 min in the dark.

135

Eventually, nitrite concentration was determined based on the absorbance at 550 nm and the

136

sodium nitrite standard curve. Furthermore, the cytokines (IL-6, TNF-α and IL-1β) released to the

137

culture medium, were also determined by ELISA using commercial reagent kits following the

138

manufacturer’s instructions. DXM (dexamethasone) is a steroid that can inhibit the expression of

139

several immune mediators and widely used in clinical trials for inflammation-related diseases.20

140

Therefore, we choose DXM as a positive control when evaluating the anti-inflammatory potential

141

of CAVAP-W.

142

RT-PCR Analysis

143

RT-PCR analysis was conducted according to Lu et al with slight modification.21 In brief,

144

RAW264.7 cells were seeded into 6-well plates at 5×105 cells/well and stimulated with tested

145

samples for 3 h. Then LPS (1 µg/mL) was added and the cells were incubated for another 12 h. 12

146

h later, RAW264.7 cells were washed with cold PBS twice and the total RNA was isolated using

147

Trizol reagent. Thereafter, total RNA was quantified with Nano Drop spectrophotometer (Nano

148

Drop Technologies, Wilmington, DE) and reverse-transcribed into cDNA with cDNA synthesis

6

ACS Paragon Plus Environment

Page 6 of 24

Page 7 of 24

Journal of Agricultural and Food Chemistry

149

kits. The resulting cDNA was further amplified by PCR with Fast Star DNA Master SYBR Green

150

I

151

TTTGTCAAGCTCATTTCCTGGTATG-3, reverse, 5-TGGGATAGGGCCTCTCTTGC-3),

152

inducible nitric oxide synthase (iNOS) (forward, 5-CGGCAA ACATGACTTCAGGC-3, reverse,

153

5-GCACATCAAAGCGGCCATAG-3), IL-6 (forward, 5-TACTCGGCAAACCTAGTGCG-3,

154

reverse,

155

GGGGATTATGGCTCAGGGTC-3,

156

(forward,

157

TGTCCATTGAGGTGGAGAGCTTTCAGC-3)

158

ATCCTTGCTGTTCC-3,

159

initiated at 50 oC for 2 min, followed by 95 oC for 10 min, 40 cycles of 95 oC for 15 s, 60 oC for 1

160

min, 95 oC for 15 s, 60 oC for 1 min and 95 oC for 15 s. The results were expressed as the ratio of

161

optimal density normalized to GAPDH.

162

Detection of NF-κB Localization by Immunofluorescence

kit

and

5

the

gene-specific

primers

were

as

below:

–GTGTCCCAACATTCATATTGTCAGT-3), reverse,

TNF-α

(forward,

(forward,

5-CGAGGCTCCAGTGAATTCGG-3),

5-TGAAGGGCTGCTTCCAAACCTTTGACC-3,

reverse,

GAPDH

and

COX-2

reverse, (forward,

5-

5IL-1β 5-

5-CAGCAA

5-TGGGCAAAGAATGCAAACATC-3). Cycling was

163

The detection of NF-κB nuclear translocation was performed based on the published research

164

using the NF-κB Activation-Nuclear Translocation Assay Kit (Beyotime Biotech, China).22

165

Initially, RAW264.7 cells adherent on glass coverslips were stimulated with LPS for 3 h and then

166

CAVAP-W (500 µg/mL) for another 12 h. When incubation time was reached, cells were fixed

167

for15 min and blocked for 1 h at room temperature. Then the primary rabbit anti-NF-κB P65

168

antibody and the secondary Cy3-labeled antibody were added successively. Afterwards,

169

RAW264.7 cells were stained with 2-(4-amidinophenyl)-6-indolecarbamidine dihydrochloride

170

(DAPI) at 2 µM for 5 min. Each step above was followed by washing three times with washing

171

buffer for 5 min each. Finally, RAW264.7 cells were preserved in anti-fade mounting medium and

172

observed with a fluorescence microscope immediately.

173

Western Blot Analysis

174

RAW264.7 cells treated with CAVAP-W at concentrations of 125, 250 and 500 µg/mL were

175

scraped into ice-cold PBS and centrifuged at 2,000 g for 5 min. Then the resulting pellets were

176

incubated with lysis buffer on ice for 40 min. Vigorous vortex was conducted every 10 min to

177

make complete lysis. Then the lysis was centrifuged at 12,000 g for 20 min at 4 °C and the 7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

178

supernatants were collected as whole cell lysates. Furthermore, the cytoplasmic and nuclear

179

proteins were extracted and isolated using the Nuclear and Cytoplasmic Protein Extraction Kit

180

(Beyotime, China) based on the manufacturer’s instructions. The concentrations of the proteins

181

obtained above were determined using a BCA kit. 40 µg of proteins were separated using 10-12%

182

sodium dodecyl sulfate polyacrylamide gel electrophoresis and then transferred to PVDF

183

membranes. Then the blots were blocked with 5% BSA diluted in 1×TBST (0.1% Tween-20 in

184

1×TBS) at 4 oC overnight, followed with incubation with various primary antibodies and

185

horseradish peroxidase-linked secondary antibody. 1×TBST was used to wash the blots after each

186

procedure above. The bands were visualized using ECL reagents and quantified with Image J

187

software (NIH, USA).

188

Statistical Analysis

189

All tests were performed in triplicate and the results were presented as mean±standard

190

deviation (SD). The results were analyzed by one-way ANOVA followed by the Tukey post hoc

191

test (p < 0.05). Statistical analysis was carried out using Statistical Package for the Social Sciences

192

20.0 software and p < 0.05 was considered significant.

193

RESULTS

194

LC-MS Analysis of CAVAP-W

195

The total phenolic content of CAVAP-W was determined to be 3.7%. Fig 1 displayed the LC-

196

MS chromatogram and the identification data. Nine compounds including eriocitrin/neoeriocitrin,

197

eriocitrin/neoeriocitrin, rhoifolin, hesperidin, naringin, rutin, veronicastroside, neohesperidin and

198

hesperetin were identified in CAVAP-W. Specifically, eriocitrin and neoeriocitrin were isomeric

199

compounds. Generally speaking, we may not be able to identify and distinguish eriocitrin and

200

neoeriocitrin based on the mass spectrum. Naringin and hesperidin were also detected in Korea C.

201

aurantium 10 and C. aurantium var. bigaradia 23 according to the published literatures. In fact, all

202

the nine compounds were identified for the first time in the analyzed sample of CAVAP-W for the

203

fact that there were no published literatures on the study of polyphenols from blossoms of CAVA.

204

Inhibitory Effects on Hemolysis and LPS-induced NO Production

8

ACS Paragon Plus Environment

Page 8 of 24

Page 9 of 24

Journal of Agricultural and Food Chemistry

205

The potential cytotoxicity of CAVAP-W on LO2 and RAW264.7 cells was evaluated by

206

MTT assay. The results (data not shown) demonstrated that CAVAP-W displayed no cellular

207

toxicity (cell viability all exceeded 90%) at concentrations up to 500 µg/mL. Therefore, 15.625,

208

31.25, 62.5, 125, 250 and 500 µg/mL were chosen as working concentrations for further research.

209

As shown in Fig 2A, CAVAP-W showed the highest hemolysis inhibition ratio of 58.91% at the

210

maximum concentration of 500 µg/mL, close to that of the positive control heparin sodium

211

(89.11%) at the same concentration (Fig 2A). Of note, at low concentration of 15.625 µg/mL, the

212

hemolysis inhibition activity of CAVAP-W was 63.75 %, much higher than that of the positive

213

control heparin sodium (19.7%).

214

Furthermore, CAVAP-W significantly suppressed the production of NO at 500 µg/mL and

215

the inhibition activity was close to that of the positive control DXM (50 µg/mL) (Fig 2B).

216

Additionally, effects of CAVAP-W on morphological changes in LPS-stimulated RAW264.7 cells

217

were also evaluated in the current research. As shown in Fig 3, LPS-induced RAW264.7 cells

218

increased in size and became irregular in shape, which was in accordance with the large quantities

219

of NO in response to inflammatory stimulus of LPS. However, the cells possessed relatively

220

smooth surfaces after treated with CAVAP-W at different concentrations of 125, 250 and 500

221

µg/mL.

222

Anti-inflammatory Activity Assay

223

Inhibitory Effect of CAVAP-W on Cytokine Secretion

224

In order to further evaluate the anti-inflammatory effect of CAVAP-W, the secretion of

225

cytokines including IL-6, TNF-ɑ and IL-1ß were investigated. Obviously, CAVAP-W displayed

226

the strongest inhibition effect on expression of IL-6 in a concentration-dependent manner (Fig 4A).

227

For example, the suppression effect of CAVAP-W (125 µg/mL) on expression of IL-6 was greater

228

than the positive control DXM (50 µg/mL). At concentrations of 31.25, 125, 250 and 500 µg/mL,

229

CAVAP-W showed significant inhibitory effects on TNF-ɑ production (Fig 4B). However, when

230

RAW264.7 cells were stimulated with CAVAP-W at 62.5 µg/mL, the accumulation of TNF-ɑ

231

(28281.82 pg/mL) even exceeded that of LPS (26781.82 pg/mL), indicating that the concentration

232

of CAVAP-W was closely related to the secretion of TNF-ɑ. Simultaneously, CAVAP-W varying

233

from 31.25 to 500 µg/mL showed significant inhibition effects on IL-1ß production without a 9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

234

concentration-dependent manner (Fig 4C). In support, published data also found that polyphenols

235

from blueberries could mediate the balances in pro-inflammatory cytokines of IL-1 and IL-6, thus

236

exhibiting anti-inflammatory activity.24

237

Inhibitory Effect of CAVAP-W on mRNA Expression of iNOS, IL-6, TNF-ɑ and

238

IL-1ß

239

The inhibitory effect of CAVAP-W on expression of iNOS on transcription level was

240

demonstrated in Fig 4D. The expression of iNOS mRNA was the lowest in normal RAW264.7

241

cells while sharply increased in LPS-induced cells. When CAVAP-W was added to the media, the

242

expression of iNOS mRNA was significantly suppressed, indicating that CAVAP-W might inhibit

243

NO production through suppressing iNOS mRNA expression. Consistently, Hwang et al. also

244

reported that NO production was closely associated with the gene expression of iNOS.25

245

Consistent with the data in 3.4.1, CAVAP-W also potently inhibited expression of IL-6 mRNA

246

(Fig 4E) and IL-1ß mRNA (Fig 4G). The mRNA level of TNF-α was not significantly affected by

247

CAVAP-W at 62.5-250 µg/mL. CAVAP-W only suppressed the expression of TNF-ɑ mRNA at

248

its maximum concentration of 500 µg/mL (Fig 4G). According to Fig 4B and F, CAVAP-W

249

showed higher inhibitory effect on TNF-α gene expression than that on TNF-α production. One

250

possible explanation would be that TNF-ɑ acted through two transmembrane receptors including

251

TNFR1 and TNFR2.26 Another reason could be that the production of some other pro-

252

inflammatory mediators, such as IL-6 and IL-1ß, could also regulate the secretion of TNF-ɑ from

253

activated macrophages.27-30

254

Inhibitory Effect of CAVAP-W on COX-2 Activation

255

COX-2 is an inducible enzyme and undetectable in most normal tissues. However, it could be

256

abundantly expressed in activated macrophages stimulated with LPS.31 Therefore, COX-2 could

257

serve as target for developing novel therapeutic options. As shown in Fig 4H, CAVAP-W

258

produced a concentration-dependent decrease in expression of COX-2 mRNA. Subsequently, this

259

was further confirmed by examining the expression of COX-2 protein using western blot method

260

(Fig 6A-1 and A-2). CAVAP-W at 125-500 µg/mL potently suppressed the expression of COX-2

261

protein in a concentration-dependent manner. Consistently, rheosmin isolated from pine needle

10

ACS Paragon Plus Environment

Page 10 of 24

Page 11 of 24

Journal of Agricultural and Food Chemistry

262

also significantly decreased COX-2 expression in RAW264.7 cells stimulated with LPS through

263

inhibiting activation of NF-kB pathway.32

264

Inhibitory Effect of CAVAP-W on NF-κB Activation

265

Immunocytochemical analysis suggested that CAVAP-W dramatically inhibited the

266

translocation of NF-kB subunit P65 into the nucleus in LPS-stimulated macrophages compared

267

with control cells (Fig 5). Consistent with immunocytochemical analysis, western blot analysis

268

further showed that CAVAP-W treatment enhanced the cytosolic P65 (Fig 6C-1 and C-2),

269

resulting in a decrease in the nuclear P65 (Fig 6D-1 and D-2). Also, CAVAP-W dramatically

270

inhibited the phosphorylation of P65 (Fig 6B-1 and B-2). In addition, CAVAP-W significantly

271

inhibited the proteolytic degradation of IκBɑ (Fig 6E-1 and E-2), phosphorylation of IκBɑ (Fig

272

6F-1 and F-2) and phosphorylation of IκKɑ/ß (Fig 6G-1 and G-2). For example, the expression of

273

IκBɑ in RAW264.7 cells was sharply improved when administrated with CAVAP-W at 500

274

µg/mL, close to that in control cells. In support, several phytochemicals showed anti-inflammation

275

potential by inhibiting activation and translocation of NF-kB from the cytoplasm to the nucleus.33-

276

36

277

Inhibitory Effect of CAVAP-W on MAPKs Phosphorylation

278

In order to further investigate the mechanism of CAVAP-W in regulation of inflammatory

279

response, the inhibition effect of CAVAP-W on MAPKs signaling pathways were also detected.

280

As shown in Fig 6E-1 and E-2, CAVAP-W significantly suppressed the phosphorylation of JNK.

281

The expression level of phospho-P38 was down-regulated when administrated with LPS,

282

consistent with published report.21 However, the expression level of phospho-P38 was up-

283

regulated when pre-treated with CAVAP-W at 125-500 µg/mL and was close to that of the control

284

cells without samples (Fig 6F-1 and F-2). These results indicated that MAPKs signaling pathway

285

was also blocked.

286

DISCUSSION

287

The crude polyphenols of CAVA (CAVAP-W) were extracted with distilled water and the

288

yield of polyphenols was determined to be 3.7% with Folin–Ciocalteu reagent. Consistent with our

289

results, Karimi also reported that the total phenolic content of boiling water extracts from Citrus

11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

290

aurantium bloom was 3.93%.37 Then LC-MS analysis demonstrated that CAVAP-W was mainly

291

composed of eriocitrin/neoeriocitrin, eriocitrin/neoeriocitrin, rhoifolin, hesperidin, naringin, rutin,

292

veronicastroside, neohesperidin and hesperetin. LC-MS method, a powerful technique for the

293

characterization and structural elucidation in the identification of polyphenols were employed in

294

many published reports.38, 39

295

Furthermore, we investigated the anti-complement and anti-inflammatory effects of CAVAP-

296

W. CAVAP-W displayed comparable hemolysis and NO inhibition rate to the positive control of

297

heparin sodium and DXM, respectively. Complement was one of the major mediators of the

298

inflammatory response. Agents with anti-inflammatory activities were usually found to display

299

significant anti-complement effects. Numerous published data reported that the complement

300

system was closely related to the inflammatory process.40,

301

demonstrated that the anti-inflammatory activity of triterpenes was probably owing to their anti-

302

complementary activity.42 Consistent with published reports,40-42 it could be inferred that the

303

potent inhibitory effect of CAVAP-W on NO production (Fig 2B) might be also closely related to

304

that on hemolysis (Fig 2A). Therefore, further study on CAVAP-W was conducted to reveal the

305

underlying molecular mechanism of its anti-inflammatory effects.

41

Specifically, previous study

306

ELISA and RT-PCR analysis further indicated that CAVAP-W potently inhibited the

307

secretion of IL-6, TNF-α and IL-1β, as well as the mRNA expression of IL-6, TNF-α, IL-1β and

308

COX-2 on transcription levels. A growing number of published reports demonstrated that NF-κB

309

was an important factor in the regulation of iNOS, IL-6, TNF-ɑ and IL-1ß.43, 44 For example, the

310

NF-κB-binding element at -85 bp of murine iNOS promoter played a critical role in iNOS

311

expression and NO production.45 So the immunocytochemical analysis and western blot analysis

312

were performed to investigate whether NF-κB was involved in regulation of inflammatory

313

response of CAVAP-W. The results showed that CAVAP-W significantly blocked NF-κB

314

activation by inhibiting the nuclear translocation of nuclear factor-kappa B (NF-κB) P65,

315

degradation and phosphorylation of IκBα, phosphorylation of IκBα and IκKBɑ/ß. Furthermore,

316

CAVAP-W administration also inhibited MAPK signaling pathway by suppressing

317

phosphorylation of JNK as well as P38. In fact, MAPK signaling pathway was closely associated

318

with NF-κB pathway and they worked together to regulate the inflammatory responses. For 12

ACS Paragon Plus Environment

Page 12 of 24

Page 13 of 24

Journal of Agricultural and Food Chemistry

319

example, polyozellin isolated from Polyozellus multiplex dramatically inhibited NO accumulation

320

and iNOS gene expression by suppressing activation of NF-κB and MAPK signaling pathways.36

321

Numerous bioactivity investigations on polyphenols extracted from natural products using cell

322

lines, at concentrations in the low-µM-to-mM range, showed that most of these active ingredients

323

appear as phase II metabolites in the circulatory system after dietary intake, and their presence in

324

plasma after dietary intake rarely exceeds nM concentrations.46 However, there were also a

325

growing number of published data demonstrating that modest long-term intakes of polyphenols

326

are beneficial to the treatment of various diseases.47, 48 Furthermore, various methods such as

327

physical activity49 and enzymatic modification50 were reported to be employed to improve the

328

bioavailability of polyphenols. Specifically, Nielsen et al. proved that the bioavailability of

329

hesperidin could be increased by changing the absorption site, which was achieved via enzymatic

330

modification to hesperetin-7-glucoside.50 Therefore, new methods could be used in future

331

interventions to ensure or improve the bioavailability of polyphenols thereby fully exploiting their

332

beneficial properties. The future work will be focused on the purification and characterization of

333

CAVAP-W in order to find the specific compounds responsible for its bioactivities.

334

Funding

335

This research was supported by Science and Technology Project of Guangzhou City

336

(201604020150), Science and Technology Project of Guangdong (2014A020212682), Special

337

Research of Traditional Chinese Medicine Hospital of Guangdong Province (YN2014ZH02).

338

Notes

339

The authors declare no competing financial interest.

340

REFERENCES

341 342 343 344 345 346 347 348 349

1. Reddy, D.-B.; Reddanna, P. Chebulagic acid (CA) attenuates LPS-induced inflammation by suppressing NF-kappa B and MAPK activation in RAW 264.7 macrophages. Biochemical and Biophysical Research Communications. 2009, 381, 112-117. 2. McCulloch, C.-A.; Downey, G.-P.; El-Gabalawy, H. Signalling platforms that modulate the inflammatory response: new targets for drug development. Nature Reviews Drug Discovery. 2006, 5, 864-876. 3. Hayashida, K.; Parks, W.-C.; Park, P.-W. Syndecan-1 shedding facilitates the resolution of neutrophilic inflammation by removing sequestered CXC chemokines. Blood. 2009, 114, 30333043.

13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405

4. Shen, C.-Y.; Jiang, J.-G.; Yang, L.; Wang, D.-W.; Zhu, W. Anti-ageing active ingredients from herbs and nutraceuticals used in traditional Chinese medicine: pharmacological mechanisms and implications for drug discovery. British journal of pharmacology. 2017, 174, 1395-1425. 5. Shen, C.-Y.; Zhang, W.-L.; Jiang, J.-G. Immune-enhancing activity of polysaccharides from Hibiscus sabdariffa Linn. via MAPK and NF-kappa B signaling pathways in RAW264.7 cells. Journal of Functional Foods. 2017, 34, 118-129. 6. Gogoi, B.; Gogoi, D.; Silla, Y.; Kakoti, B.-B.; Bhau, B.-S. Network pharmacology-based virtual screening of natural products from Clerodendrum species for identification of novel anticancer therapeutics. Molecular bioSystems. 2017, 13, 406-416. 7. Shen, C.-Y.; Zhang, T.-T.; Zhang, W.-L.; Jiang, J.-G. Anti-inflammatory activities of essential oil isolated from the calyx of Hibiscus sabdariffa L. Food & Function. 2016, 7, 44514459. 8. Chen, X.-M.; Tait, A.-R.; Kitts, D.-D. Flavonoid composition of orange peel and its association with antioxidant and anti-inflammatory activities. Food Chemistry. 2017, 218, 15-21. 9. Khan, R.-A.; Mallick, N.; Feroz, Z. Anti-inflammatory effects of Citrus sinensis L., Citrus paradisi L. and their combinations. Pakistan Journal of Pharmaceutical Sciences. 2016, 29, 843-852. 10. Kang, S.-R.; Il Park, K.; Park, H.-S.; Lee, D.-H.; Kim, J.-A.; Nagappan, A.; Kim, E.-H.; Lee, W.-S.; Shin, S.-C.; Park, M.-K.; Han, D.-Y.; Kim, G.-S. Anti-inflammatory effect of flavonoids isolated from Korea Citrus aurantium L. on lipopolysaccharide-induced mouse macrophage RAW 264.7 cells by blocking of nuclear factor-kappa B (NF-kappa B) and mitogenactivated protein kinase (MAPK) signalling pathways. Food Chemistry. 2011, 129, 1721-1728. 11. Mahmoud, M.-F.; Hamdan, D.-I.; Wink, M.; El-Shazly, A.-M. Hepatoprotective effect of limonin, a natural limonoid from the seed of Citrus aurantium var. bigaradia, on Dgalactosamine-induced liver injury in rats. Naunyn-Schmiedebergs Archives of Pharmacology. 2014, 387, 251-261. 12. Shen, C.-Y.; Wang, T.-X; Zhang, X.-M; Jiang, J.-G. Various Antioxidant Effects Were Attributed to Different Components in the Dried Blossoms of Citrus aurantium L. var. amara Engl. Journal of Agricultural and Food Chemistry.2017, 65, 6087-6092. 13. Shen, C.-Y.; Jiang, J.-G.; Li, M.-Q.; Zheng, C.-Y.; Zhu, W. Structural characterization and immunomodulatory activity of novel polysaccharides from Citrus aurantium Linn. variant amara Engl. Journal of Functional Foods. 2017, 35, 352-362. 14. Shen, C.-Y.; Yang, L.; Jiang, J.-G.; Zheng, C.-Y.; Zhu, W. Immune enhancement effects and extraction optimization of polysaccharides from Citrus aurantium L. var. amara Engl. Food & Function. 2017, 8, 796-807. 15. Makkar, H.P.-S.; Becker, K.; Abel, H.; Pawelzik, E. Nutrient contents, rumen protein degradability and antinutritional factors in some colour- and white-flowering cultivars of Vicia faba beans. Journal of the Science of Food and Agriculture. 1997, 75, 511-520. 16. Kelebek, H. Sugars, organic acids, phenolic compositions and antioxidant activity of Grapefruit (Citrus paradisi) cultivars grown in Turkey. Industrial Crops and Products. 2010, 32, 269-274. 17. de Souza, M.-P.; Bataglion, G.-A.; da Silva, F.M.-A.; de Almeida, R.-A.; Paz, W.H.-P.; Nobre, T.-A.; Marinho, J.V.-N.; Salvador, M.-J.; Fidelis, C.H.-V.; Acho, L.D.-R.; de Souza, A.D.L.; Nunomura, R.C.-S.; Eberlin, M.-N.; Lima, E.-S.; Koolen, H.H.-F. Phenolic and aroma compositions of pitomba fruit (Talisia esculenta Radlk.) assessed by LC-MS/MS and HSSPME/GC-MS. Food Research International. 2016, 83, 87-94. 18. Ibanez, C.; Simo, C.; Garcia-Canas, V.; Gomez-Martinez, A.; Ferragut, J.-A.; Cifuentes, A. CE/LC-MS multiplatform for broad metabolomic analysis of dietary polyphenols effect on colon cancer cells proliferation. Electrophoresis. 2012, 33, 2328-2336. 19. Seo, H.-W.; Hung, T.-M.; Na, M.; Jung, H.-J.; Kim, J.-C.; Choi, J.-S.; Kim, J.-H.; Lee, H.-K.; Lee, I.; Bae, K.; Hattori, M.; Min, B.-S. Steroids and triterpenes from the fruit bodies of Ganoderma lucidum and their anti-complement activity. Archives of Pharmacal Research. 2009, 32, 1573-1579. 20. Ramesh, G.; Martinez, A.-N.; Martin, D.-S.; Philipp, M.-T. Effects of dexamethasone and meloxicam on Borrelia burgdorferi-induced inflammation in glial and neuronal cells of the central nervous system. Journal of Neuroinflammation. 2017, 14.

14

ACS Paragon Plus Environment

Page 14 of 24

Page 15 of 24

Journal of Agricultural and Food Chemistry

406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462

21. Lu, C.-L.; Zhu, W.; Wang, M.; Hu, M.-M.; Chen, W.-L.; Xu, X.-J.; Lu, C.-J. Polysaccharides from Smilax glabra inhibit the pro-inflammatory mediators via ERK1/2 and JNK pathways in LPS-induced RAW264.7 cells. Carbohydrate Polymers. 2015, 122, 428-436. 22. Hu, F.; Yang, S.; Zhao, D.; Zhu, S.; Wang, Y.; Li, J. Moderate extracellular acidification inhibits capsaicin-induced cell death through regulating calcium mobilization, NF-kappa B translocation and ROS production in synoviocytes. Biochemical and Biophysical Research Communications. 2012, 424, 196-200. 23. Hamdana, D.-I.; Mahmoud, M.-F.; Wink, M.; Ei-Shazly, A.-M. Effect of hesperidin and neohesperidin from bittersweet orange (Citrus aurantium var. bigaradia) peel on indomethacininduced peptic ulcers in rats. Environmental Toxicology and Pharmacology. 2014, 37, 907-915. 24. Cheng, A.; Yan, H.; Han, C.; Wang, W.; Tian, Y.; Chen, X. Polyphenols from blueberries modulate inflammation cytokines in LPS-induced RAW264.7 macrophages. International Journal of Biological Macromolecules. 2014, 69, 382-387. 25. Hwang, P.-A.; Chien, S.-Y.; Chan, Y.-L.; Lu, M.-K.; Wu, C.-H.; Kong, Z.-L.; Wu, C.-J. Inhibition of Lipopolysaccharide (LPS)-Induced Inflammatory Responses by Sargassum hemiphyllum Sulfated Polysaccharide Extract in RAW 264.7 Macrophage Cells. Journal of Agricultural and Food Chemistry. 2011, 59, 2062-2068. 26. Parameswaran, N.; Patial, S. Tumor Necrosis Factor-a Signaling in Macrophages. Critical Reviews in Eukaryotic Gene Expression. 2010, 20, 87-103. 27. Lee, J.; Yang, G.; Lee, K.; Lee, M.-H.; Eom, J.-W.; Ham, I.; Choi, H.-Y. Antiinflammatory effect of Prunus yedoensis through inhibition of nuclear factor-kappa B in macrophages. Bmc Complementary and Alternative Medicine. 2013, 13. 28. Lee, D.-S.; Ko, W.; Tran Hong, Q.; Kim, K.-S.; Sohn, J.-H.; Jang, J.-H.; Ahn, J.-S.; Kim, Y.-C.; Oh, H. Penicillinolide A: A New Anti-Inflammatory Metabolite from the Marine Fungus Penicillium sp SF-5292. Marine Drugs. 2013, 11, 4510-4526. 29. Cuoi, Y.-H.; Jin, G.-Y.; Li, G.-Z.; Yan, G.-H. Cornuside Suppresses LipopolysaccharideInduced Inflammatory Mediators by Inhibiting Nuclear Factor-Kappa B Activation in RAW 264.7 Macrophages. Biological & Pharmaceutical Bulletin. 2011, 34, 959-966. 30. Liu, Z.; Jiang, T.; Wang, X.; Wang, Y. Fluocinolone acetonide partially restores the mineralization of LPS-stimulated dental pulp cells through inhibition of NF-kappa B pathway and activation of AP-1 pathway. British Journal of Pharmacology. 2013, 170, 1262-1271. 31. Yoon, W.-J.; Ham, Y.-M.; Yoo, B.-S.; Moon, J.-Y.; Koh, J.; Hyun, C.-G. Oenothera laciniata inhibits lipopolysaccharide induced production of nitric oxide, prostaglandin E-2, and proinflammatory cytokines in RAW264.7 macrophages. Journal of Bioscience and Bioengineering. 2009, 107, 429-438. 32. Jeong, J.-B.; Jeong, H.-J. Rheosmin, a naturally occurring phenolic compound inhibits LPS-induced iNOS and COX-2 expression in RAW264.7 cells by blocking NF-kappa B activation pathway. Food and Chemical Toxicology. 2010, 48, 2148-2153. 33. Won, J.-H.; Kim, J.-Y.; Yun, K.-J.; Lee, J.-H.; Back, N.-I.; Chung, H.-G.; Chung, S.-A.; Jeong, T.-S.; Choi, M.-S.; Lee, K.-T. Gigantol isolated from the whole plants of Cymbidium goeringii inhibits the LPS-induced iNOS and COX-2 expression via NF-kappa B inactivation in RAW 264.7 macrophages cells. Planta Medica. 2006, 72, 1181-1187. 34. Lin, C.-M.; Huang, S.-T.; Liang, Y.-C.; Lin, M.-S.; Shih, C.-M.; Chang, Y.-C.; Chen, T.Y.; Chen, C.-T. Isovitexin suppresses lipopolysaccharide-mediated inducible nitric oxide synthase through inhibition of NF-kappa B in mouse macrophages. Planta Medica. 2005, 71, 748-753. 35. Ko, H.-C.; Kuo, Y.-H.; Wei, B.-L.; Chiou, W.-F. Laxifolone a suppresses LPS/IFNgamma-induced NO synthesis by attenuating NF-kappa B translocation: Role of NF-kappa B p105 level. Planta Medica. 2005, 71, 514-519. 36. Jin, X.-Y.; Lee, S.-H.; Kim, J.-Y.; Zhao, Y.-Z.; Park, E.-J.; Lee, B.-S.; Nan, J.-X.; Song, K.-S.; Ko, G.; Sohn, D.-H. Polyozellin inhibits nitric oxide production by down-regulating LPSinduced activity of NF-kappa B and SAPK/JNK in RAW 264.7 cells. Planta Medica. 2006, 72, 857-859. 37. Karimi, E.; Oskoueian, E.; Hendra, R.; Oskoueian, A.; Jaafar, H.Z.-E. Phenolic Compounds Characterization and Biological Activities of Citrus aurantium Bloom. Molecules. 2012, 17, 1203-1218. 38. Lucci, P.; Saurina, J.; Nunez, O. Trends in LC-MS and LC-HRMS analysis and characterization of polyphenols in food. Trac-Trends in Analytical Chemistry 2017, 88, 1-24.

15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497

39. Antonia Alvarez-Fernandez, M.; Cerezo, A.-B.; Canete-Rodriguez, A.-M.; Troncoso, A.M.; Carmen Garcia-Parrilla, M. Composition of Nonanthocyanin Polyphenols in AlcoholicFermented Strawberry Products Using LC-MS (QTRAP), High-Resolution MS (UHPLCOrbitrap-MS), LC-DAD, and Antioxidant Activity. Journal of Agricultural and Food Chemistry. 2015, 63, 2041-2051. 40. Kapil, A.; Moza, N. Anticomplementary activity of boswellic acids--an inhibitor of C3convertase of the classical complement pathway. International journal of immunopharmacology 1992, 14, 1139-43. 41. Frank, M.-M.; Fries, L.-F. The role of complement in inflammation and phagocytosis. Immunology today. 1991, 12, 322-6. 42. Geetha, T.; Varalakshmi, P. Anticomplement activity of triterpenes from Crataeva nurvala stem bark in adjuvant arthritis in rats. General Pharmacology. 1999, 32, 495-497. 43. Arend, W.-P.; Dayer, J.-M. Inhibition of the production and effects of interleukin-1 and tumor necrosis factor alpha in rheumatoid arthritis. Arthritis and rheumatism. 1995, 38, 151-60. 44. Koch, A.-E. Chemokines and Their Receptors in Rheumatoid Arthritis Future Targets? Arthritis and Rheumatism. 2005, 52, 710-721. 45. Xie, Q.; Nathan, C. The high-output nitric oxide pathway: role and regulation. Journal of leukocyte biology. 1994, 56, 576-82. 46. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.-E.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (Poly)phenolics in Human Health: Structures, Bioavailability, and Evidence of Protective Effects Against Chronic Diseases. Antioxidants & Redox Signaling. 2013, 18, 18181892. 47. Reshef, N.; Hayari, Y.; Goren, C.; Boaz, M.; Madar, Z.; Knobler, H. Antihypertensive effect of sweetie fruit in patients with stage I hypertension. American Journal of Hypertension. 2005, 18, 1360-1363. 48. Morand, C.; Dubray, C.; Milenkovic, D.; Lioger, D.; Martin, J.-F.; Scalbert, A.; Mazur, A. Hesperidin contributes to the vascular protective effects of orange juice: a randomized crossover study in healthy volunteers. American Journal of Clinical Nutrition. 2011, 93, 73-80. 49. Medina, S.; Dominguez-Perles, R.; Garcia-Viguera, C.; Cejuela-Anta, R.; Martinez-Sanz, J. M.; Ferreres, F.; Gil-Izquierdo, A. Physical activity increases the bioavailability of flavanones after dietary aronia-citrus juice intake in triathletes. Food Chemistry. 2012, 135, 2133-2137. 50. Nielsen, I.L.-F.; Chee, W.S.-S.; Poulsen, L.; Offord-Cavin, E.; Rasmussen, S.-E.; Frederiksen, H.; Enslen, M.; Barron, D.; Horcajada, M.-N.; Williamson, G. Bioavailability is improved by enzymatic modification of the citrus flavonoid hesperidin in humans: A randomized, double-blind, crossover trial. Journal of Nutrition. 2006, 136, 404-408.

498 499

16

ACS Paragon Plus Environment

Page 16 of 24

Page 17 of 24

Journal of Agricultural and Food Chemistry

500 501 502

Figure legends

503

5, 6, 7, 8 and 9 represented neohespeidoside (C27H32O15), neoeriocitrin (C27H32O15), rhoifolin

504

(C27H30O14), hesperidin (C28H34O15), naringin (C27H32O14), rutin (C27H30O16), veronicastroside

505

(C27H30O15), neohesperidin (C29H38O23) and hesperetin (C16H14O6), respectively. Their fragmens

506

at (M-H)- (m/z) were 595.1708, 595.1708, 577.1583, 609.1851, 579.1743, 609.1491,

507

593.1535, 753.2283 and 301.0721.

508

Fig. 2. Inhibitory effects of CAVAP-W on the classical pathway of the complement system (A);

509

Inhibition effect of CAVAP-W on NO production in LPS-induced RAW264.7 cells (B). All

510

experiments were run in triplicate, and data showed mean ± SD values. *p < 0.05 and **p < 0.01

511

compared to control group.

512

Fig. 3. Pictures of RAW264.7 cells by an invert microscope after treated with CAVAP-W at

513

different concentrations.

514

Fig. 4. Effects of CAVAP-W on LPS-induced secretion of IL-6 (A), TNF-α (B) and IL-1ß (C);

515

Effects of CAVAP-W on LPS-induced mRNA expression of iNOS (D), IL-6 (E), TNF-α (F), IL-

516

1ß (G) and COX-2 (H). All experiments were run in triplicate, and data showed mean ± SD values.

517

*p < 0.05 and **p < 0.01 compared to LPS-treated group, while

518

control group.

519

Fig. 5. Analysis of nuclear translocation of NF-κB P65 of CAVAP-W by immunofluorescence

520

analysis.

521

Fig. 6. Effects of CAVAP-W on the COX-2 (A-1, A-2); phospho-P65 (B-1, B-2); cytosol P65 (C-

522

1, C-2); nucleus P65 (D-1; D-2); IκBα (E-1, E-2); phospho-IκBα (F-1, F-2); phospho-IκKα/ß (G-1,

523

G-2); phospho-JNK (H-1, H-2); phospho-P38 (I-1, I-2);. All experiments were run in triplicate,

524

and data showed mean ± SD values. *p < 0.05 and **p < 0.01 compared to LPS-treated group,

525

while ##p < 0.01 compared to the control group.

Fig 1. MS chromatogram of CAVAP-W. The numbers represents the compounds.. Peaks 1, 2, 3, 4,

526

17

ACS Paragon Plus Environment

##

p < 0.01 compared to the

Journal of Agricultural and Food Chemistry

527 528

Fig 1. The total ion chromatogram of CAVAP-W. Peaks 1, 2, 3, 4, 5, 6, 7, 8 and 9 represented

529

eriocitrin/neoeriocitrin (C27H32O15), eriocitrin/neoeriocitrin (C27H32O15), rhoifolin (C27H30O14),

530

hesperidin (C28H34O15), naringin (C27H32O14), rutin (C27H30O16), veronicastroside (C27H30O15),

531

neohesperidin (C29H38O23) and hesperetin (C16H14O6), respectively. Their precursor ions [M-H]-

532

of each individual were at m/z 595.1708, 595.1708, 577.1583, 609.1851, 579.1743, 609.1491,

533

593.1535, 753.2283 and 301.0721, respectively.

534

18

ACS Paragon Plus Environment

Page 18 of 24

Page 19 of 24

Journal of Agricultural and Food Chemistry

535 CAVAP-W Heparin sodium

80 60

∗∗

∗∗

100

∗∗

NO inhibition (%)

Hemolysis inhibition (%)

100

∗∗ ∗∗ ∗



∗∗



∗∗

40 20

∗∗



15.625 31.25 62.5 125 250 500 concentration (µg/mL) A

80

∗∗ ∗∗

60 40 ∗

20

∗∗

0 DXM 15.625 31.25 62.5 125 250 500 CAVAP-W (µg/mL) B

536

Fig. 2. Inhibitory effects of CAVAP-W on the classical pathway of the complement system (A);

537

Inhibition effect of CAVAP-W on NO production in LPS-induced RAW264.7 cells (B). All

538

experiments were run in triplicate, and data showed mean ± SD values. *p < 0.05 and **p < 0.01

539

compared to control group.

540

19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

541 542

Fig. 3. Pictures of RAW264.7 cells by an invert microscope after treated with CAVAP-W at

543

different concentrations.

544

20

ACS Paragon Plus Environment

Page 20 of 24

Page 21 of 24

Journal of Agricultural and Food Chemistry

∗∗ ∗∗

∗∗

∗∗ ∗∗

∗∗

Con LPS DXM 31.25 62.5 125 250 500 CAVAP-W (µg/mL) A

35 30 25 20 15 10 5 0

TNF-α accumulation (pg/mL)

##

35000 30000 25000 20000 15000 10000 5000 0

iNOS mRNA expression

3500 3000 2500 2000 1500 1000 500 0

IL-1β accumulation (pg/mL)

IL-6 accumulation (pg/mL)

545

## ∗∗

∗∗

∗∗

∗∗

∗∗

∗∗

Con LPS DXM 31.25 62.5 125 250 500 CAVAP-W (µg/mL) C

30000 ∗∗ ∗∗

∗∗

∗∗

10000 0

∗∗

Con LPS DXM 62.5 125 250 500 CAVAP-W (µg/mL) E

COX-2 mRNA expression

IL-1β mRNA expression

##

100000 80000

∗∗

∗∗

## ∗∗

∗∗

∗∗

∗∗ ∗∗

Con LPS DXM 62.5 125 250 500 CAVAP-W (µ g/mL ) D ## ∗∗ ∗∗

## ∗∗

3000

∗∗

∗∗

60000

∗∗

2000

40000

∗∗

∗∗

∗∗

1000

20000 0

Con LPS DXM 31.25 62.5 125 250 500 CAVAP-W (µg/mL) B

4000

∗∗

∗∗

∗∗

C o n L P S D X M 6 2 .5 1 2 5 2 5 0 5 0 0 C A V A P -W ( µ g /m L ) F 5000

140000 120000

∗∗

TNF-α mRNA expression

IL-6 mRNA expression

##

20000

∗∗ ∗∗

14 12 10 8 6 4 2 0

50000 40000

7000 6000 5000 4000 3000 2000 1000 0

##

C on LPS D X M 62.5 125 250 500 CA VA P-W ( µ g/mL)

0 C on

LPS D X M 62.5 125 250 500

546

C A V A P-W (µ g/m L ) H G Fig. 4. Effects of CAVAP-W on LPS-induced secretion of IL-6 (A), TNF-α (B) and IL-1ß (C);

547

Effects of CAVAP-W on LPS-induced mRNA expression of iNOS (D), IL-6 (E), TNF-α (F), IL-

548

1ß (G) and COX-2 (H). All experiments were run in triplicate, and data showed mean ± SD values.

549

*p < 0.05 and **p < 0.01 compared to LPS-treated group, while

550

control group.

551

21

ACS Paragon Plus Environment

##

p < 0.01 compared to the

Journal of Agricultural and Food Chemistry

552 553

Fig. 5. Analysis of nuclear translocation of NF-κB P65 of CAVAP-W by immunofluorescence

554

analysis.

22

ACS Paragon Plus Environment

Page 22 of 24

Page 23 of 24

Journal of Agricultural and Food Chemistry

555

B-1

0.3

∗∗

∗∗

0.2

0.4 ∗∗

0.2

∗∗

0.1 0.0

0.0 Con LPS 125 250 500 CAVAP-W (µg/mL) A-2

Con LPS 125 250 500 CAVAP-W (µg/mL)

Relative density of IκBα

∗∗ ∗∗

∗∗

0.4

∗∗

0.0

Con LPS 125 250 500 CAVAP-W (µg/mL) D-2

Con LPS 125 250 500 CAVAP-W (µg/mL) E-2

Relative density of p-IκKα/β

Relative density of p-JNK

##

∗∗ ∗∗

0.4

∗∗

∗∗

0.2

0.2 0.0 Con LPS 125 250 500 CAVAP-W (µ g/mL )

Con LPS 125 250 500 CAVAP-W (µg/mL)C-2

F-1 ##

∗∗ ∗∗ ∗∗

0.0 Con LPS 125 250 500 CAVAP-W (µg/mL) F-2

I-1

H-1 0.6

0.4

0.0

0.2

0.8

∗∗

0.2

0.4

G-1 ##

∗∗

0.6

##

0.1

0.2

0.6

∗∗

0.8

∗∗

0.2

∗∗

0.8

∗∗ ##

0.4

1.0

0.3

##

0.6

0.6

B-2

0.4

0.8

0.8

E-1

D-1 1.0 Relative density of P65

Relative density of P65

∗∗

Relative density of p-IκBα

0.6

##

0.4

##

0.0 C on LPS 125 250 500 C A V A P-W (ug/mL)

Relative density of p-P38

0.8

0.0

C-1 1.0

0.5

Relative density of p-P65

Relative density of COX-2

A-1 1.0

0.3 ∗∗ ∗∗

0.2

∗∗ ##

0.1 0.0

Con LPS 125 250 500

556

CA V AP-W (µ g/mL ) I-2 H-2 G-2 Fig. 6. Effects of CAVAP-W on the COX-2 (A-1, A-2); phospho-P65 (B-1, B-2); cytosol P65 (C-

557

1, C-2); nucleus P65 (D-1; D-2); IκBα (E-1, E-2); phospho-IκBα (F-1, F-2); phospho-IκKα/ß (G-1,

558

G-2); phospho-JNK (H-1, H-2); phospho-P38 (I-1, I-2). All experiments were run in triplicate, and

559

data showed mean ± SD values. *p < 0.05 and **p < 0.01 compared to LPS-treated group, while

560

##

p < 0.01 compared to the control group.

561 23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

562

TOC graphic

563

24

ACS Paragon Plus Environment

Page 24 of 24