• 沒有找到結果。

Anti-inflammatory Activities of Cinnamomum cassia Constituents in vitro and in vivo

N/A
N/A
Protected

Academic year: 2021

Share "Anti-inflammatory Activities of Cinnamomum cassia Constituents in vitro and in vivo"

Copied!
44
0
0

加載中.... (立即查看全文)

全文

(1)

Running Head: Anti-inflammatory activities of Cinnamomum cassia Constituents 1

2

Anti-inflammatory Activities of Cinnamomum cassia Constituents in vitro

3

and in vivo

4 5

Jung-Chun Liaoa, Jeng-Shyan Dengb, Chuan-Sung Chic, d, Wen-Chi Houe, Shyh-Shyun 6

Huanga, Pei-Hsin Shiec, Guan-Jhong Huangc,* 7

8 a

School of Pharmacy, College of Pharmacy, China Medical University, Taichung 404, 9

Taiwan 10

b

Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, 11

Taiwan 12

c

School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College 13

of Pharmacy, China Medical University, Taichung 404, Taiwan. 14

d

Nursing Department, Hsin Sheng College of Medical Care and Management, Taoyuan 325, 15

Taiwan. 16

e

Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei 250, Taiwan. 17 18 * Corresponding author: 19 Guan-Jhong Huang 20

School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of 21

Pharmacy, China Medical University, Taichung 404, Taiwan. 22

Tel: +886- 4- 2205-3366. Ext: 5508. Fax: +886- 4-2208-3362, 23

E-mail address: gjhuang@mail.cmu.edu.tw 24

25 26

(2)

Abstract

27

In this study, we have investigated the anti-inflammatory effects of Cinnamomum 28

cassia constituents (cinnamic aldehyde, cinnamic alcohol, cinnamic acid, and coumarin) 29

using lipopolysaccharide (LPS)-stimulated mouse macrophage (RAW264.7) in vitro and 30

carrageenan (Carr)-induced mouse paw edema model in vivo. When RAW264.7

31

macrophages were treated with cinnamic aldehyde together with LPS, a significant

32

concentration-dependent inhibition of nitric oxide (NO), tumor necrosis factor (TNF-α),

33

and prostaglandin E2 (PGE2) levels productions were detected. Western blotting 34

revealed that cinnamic aldehyde blocked protein expression of inducible nitric oxide

35

synthase (iNOS), cyclooxygenase-2 (COX-2), nuclear transcription factor kappa B

36

(NF-B), and IB, significantly.

37

In the anti-inflammatory test, cinnamic aldehyde decreased the paw edema at the 4th 38

and the 5th h after -carrageenin (Carr) administration, and increased the activities of 39

catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) in the 40

paw tissue. We also demonstrated cinnamic aldehyde significantly attenuated the 41

malondialdehyde (MDA) level and myeloperoxidase (MPO) activity in the edema paw at 42

the 5th h after Carr injection. Cinnamic aldehyde decreased the NO, TNF-α, and PGE2 43

levels on the serum level at the 5th h after Carr injection. Western blotting revealed that 44

cinnamic aldehyde decreased Carr-induced iNOS, COX-2, and NF-B expressions at the 45

5th h in the edema paw. An intraperitoneal (i.p.) injection treatment with cinnamic 46

aldehyde also diminished neutrophil infiltration into sites of inflammation as did 47

indomethacin (Indo). The anti-inflammatory mechanisms of cinnamic aldehyde might be 48

related to the decrease in the level of MDA, MPO, iNOS, and COX-2 via increasing the 49

(3)

activities of CAT, SOD, and GPx in the edema paw through the suppression of NO, 50

TNF-, and PGE2. These findings demonstrated that cinnamic aldehyde has excellent 51

anti-inflammatory activities in vitro and in vivo and thus have great potential to be used 52

as a source for natural health products. 53

54

KEY WORDS: Chinese medicine; Cinnamic aldehyde; Anti-inflammation; NO; TNF-α.

55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72

(4)

INTRODUCTION

73

Inflammation is recognized as a biological process in response to tissue injury. At the 74

injury site, an increase in blood vessel wall permeability followed by migration of 75

immune cells can lead edema formation during inflammation [1]. Inflammation leads to 76

the up-regulation of a series of enzymes and signaling proteins in affected cells and 77

tissues. Inducible nitric oxide synthase (iNOS), a member of the NOS protein family, 78

catalyzes the formation of nitric oxide (NO) from L-arginine [2]. Low concentration of 79

NO produced by iNOS is likely to contribute to the antimicrobial activity of macrophages 80

against certain bacterial pathogens. Lipopolysaccharide (LPS) is an endotoxin and a 81

constituent of the outer membrane of gram-negative bacteria. LPS stimulates innate 82

immunity, by regulating the productions of inflammatory mediators, like, NO, TNF-α, 83

and Interleukin-6 [3]. And in the animal the inflammation model of a carrageenan (Carr) 84

induced edema is usually used to assess the contributionof natural products in resisting 85

the biochemical changes associated with acute inflammation. Carr can induce acute 86

inflammation beginning with infiltration of phagocytes, the production of free radicals as 87

wellas the release of inflammatory mediators [4]. The resulting inflammation has been 88

shown to be associated with a number of chronic diseases, including asthma, rheumatoid 89

arthritis, inflammatory bowel disease, atherosclerosis, and Alzheimer’s disease, and also 90

has a role in various human cancers [5]. 91

Intracellular antioxidant mechanisms against these inflammatory stresses involve 92

antioxidant enzymes, including superoxide dismutase (SOD), catalase (CAT) and 93

glutathione peroxidase (GPx) in tissues. Recently, it has been shown that faulty cellular 94

antioxidant systems cause organisms to develop a series of inflammatory and cancer 95

(5)

diseases [6]. However, it appears that the various roles of enzymatic antioxidants help to 96

protect organisms from excessive generation of oxidative stress in the inflammatory 97

process, which has triggered studies focusing on the role of natural products in 98

suppressing the production of oxidation by increasing enzymatic antioxidants in tissues 99

[7]. 100

Cinnamomum cassia (C. cassia), bark is the outer skin of an evergreen tall tree 101

belonging to the family Lauraceae. It is commonly used as traditional Chinese medicine 102

for treating dyspepsia, gastritis, blood circulation disturbances, and inflammatory 103

diseases. Its extracts contain several active components such as essential oils (cinnamic 104

aldehyde, cinnamic alcohol, cinnamic acid, and coumarin), tannin, mucus and 105

carbohydrates [8]. C. cassia has been shown to have many pharmacological properties, 106

such as antiulcerogenic, anti-inflammatory, antipyretic, antimicrobial, antidiabetic and 107

anti-tumor activity [9, 10]. However, in this paper we examined that cinnamic aldehyde 108

was the most potent anti-inflammatory constituent of C. cassia on LPS-induced in 109

RAW264.7 cells and Carr-induced on paw edema in mice. And we detected the levels of 110

iNOS, COX-2, and NF-B in either RAW264.7 cell or paw edema. Also, the activities of 111

CAT, SOD, and GPx in the paw tissue at the 5th h after Carr injection were measured to 112

understand the relationship between the anti-inflammatory mechanism of cinnamic 113

aldehyde and antioxidant enzymes. 114

115

Materials and methods

116

Chemicals

(6)

LPS (endotoxin from Escherichia coli, serotype 0127:B8), Carr, indomethacin, 118

cinnamic aldehyde ( 98%), cinnamic alcohol ( 98%), cinnamic acid ( 99%), 119

coumarin (99%) (Fig. 1A) and other chemicals were purchased from Sigma Chemical 120

Co. (St. Louis, USA). TNF-α and PGE2 were purchased from Biosource International Inc. 121

(Camarillo, CA, USA). Anti-iNOS, anti-COX-2, anti-NF-B, anti-IB, and anti-β-actin 122

antibody (Santa Cruz, USA) and a protein assay kit (Bio-Rad Laboratories Ltd., Watford, 123

Herts, U.K.) were obtained as indicated. Poly-(vinylidene fluoride) membrane 124

(Immobilon-P) was obtained from Millipore Corp. (Bedford, MA, USA). 125

126

Animals

127

6-8 weeks male imprinting control region (ICR) mice were obtained from the 128

BioLASCO Taiwan Co., Ltd. The animals were kept in plexiglass cages at a constant 129

temperature of 22 ±1°C, and relative humidity of 55 ± 5 % with 12 h dark-light cycle for 130

at least 2 week before the experiment. They were given food and water ad libitum. All 131

experimental procedures were performed according to the National Institutes of Health 132

(NIH) Guide for the Care and Use of Laboratory Animals. In addition, all tests were 133

conducted under the guidelines of the International Association for the Study of Pain. 134

After a 2-week adaptation period, male ICR mice (18-25 g) were randomly assigned 135

to four groups (n=6) of the animals in the study. The control group receives normal saline 136

(i.p.). The other three groups include a Carr-treated, a positive control (Carr + Indo) and 137

cinnamic aldehyde administered groups (Carr + cinnamic aldehyde). 138

(7)

Cell culture

140

A murine macrophage cell line RAW264.7 (BCRC No. 60001) was purchased from

141

the Bioresources Collection and Research Center (BCRC) of the Food Industry Research 142

and Development Institute (Hsinchu, Taiwan). Cells were cultured in plastic dishes 143

containing Dulbecco’s Modified Eagle Medium (DMEM, Sigma, St. Louis, MO, USA) 144

supplemented with 10% fetal bovine serum (FBS, Sigma, USA) in a CO2 incubator (5% 145

CO2 in air) at 37°C and subcultured every 3 days at a dilution of 1:5 using 0.05% 146

trypsin–0.02% EDTA in Ca2+-, Mg2+- free phosphate-buffered saline (DPBS). 147

148

Cell viability

149

Cells (2 x 105) were cultured in 96-well plate containing DMEM supplemented with 150

10% FBS for 1 day to become nearly confluent. Then cells were cultured with cinnamic 151

aldehyde, cinnamic alcohol, cinnamic acid, and coumarin in the presence of 100 ng/mL 152

LPS (lipopolysaccharide) for 24 h. After that, the cells were washed twice with DPBS 153

and incubated with 100 L of 0.5 mg/mL MTT for 2 h at 37°C testing for cell viability 154

{MTT, (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide)}. The medium 155

was then discarded and 100 L dimethyl sulfoxide (DMSO) was added. After 30-min 156

incubation, absorbance at 570 nm was read using a microplate reader. 157

158

Measurement of Nitric oxide/Nitrite

159

NO production was indirectly assessed by measuring the nitrite levels in the cultured

160

media and serum determined by a colorimetric method based on the Griess reaction [4]. 161

(8)

The cells were incubated with cinnamic aldehyde, cinnamic alcohol, cinnamic acid, 162

coumarin (0, 6.25, 12.5, 25, and 50 M) in the presence of LPS (100 ng/mL) at 370C for 163

24 h. Then, cells were dispensed into 96-well plates, and 100 mL of each supernatant was 164

mixed with the same volume of Griess reagent (1% sulfanilamide, 0.1% naphthyl 165

ethylenediamine dihydrochloride and 5% phosphoric acid) and incubated at room 166

temperature for 10 min, the absorbance was measured at 540 nm with a Micro-Reader 167

(Molecular Devices, Orleans Drive, Sunnyvale, CA). Serum samples were diluted four 168

times with distilled water and deproteinized by adding 1/20 volume of zinc sulfate (300 169

g/L) to a final concentration of 15 g/L. After centrifugation at 10,000×g for 5 min at room 170

temperature, 100 μL supernatant was applied to a microtiter plate well, followed by 100 171

μL of Griess reagent. After 10 min of color development at room temperature, the 172

absorbance was measured at 540 nm with a Micro-Reader. By using sodium nitrite to 173

generate a standard curve, the concentration of nitrite was measured by absorbance at 540 174 nm. 175 176 Carr-induced Edema 177

The Carr-induced hind paw edema model was used for determination of

178

anti-inflammatory activity [1]. Animals were i.p. treated with cinnamic aldehyde (1.25, 179

2.5 and 5 mg/kg), Indo or normal saline, 30 min prior to injection of 1% Carr (50 μL) in 180

the plantar side of right hind paws of the mice. The paw volume was measured after Carr 181

injection and at 1, 2, 3, 4, and 5 h intervals after the administration of the edematogenic 182

agent using a plethysmometer (model 7159, Ugo Basile, Varese, Italy). The degree of 183

swelling induced was evaluated by the ratio a/b, where a is the volume of the right hind 184

(9)

paw after Carr treatment, and b is the volume of the right hind paw before Carr treatment. 185

Indo was used as a positive control. After 5 h, the animals were sacrificed and the 186

Carr-induced edema feet were dissected and stored at -80 ºC. Also, blood were 187

withdrawn and kept at -80 ºC. The protein concentration of the sample was determined by 188

the Bradford dye-binding assay (Bio-Rad, Hercules, CA). 189

190

MDA Assay

191

MDA from Carr-induced edema foot was evaluated by the thiobarbituric acid reacting

192

substance (TRARS) method [1]. Briefly, MDA reacted with thiobarbituric acid in the 193

acidic high temperature and formed a red-complex TBARS. The absorbance of TBARS 194

was determined at 532 nm. 195

196

Myeloperoxidase Activity Assay

197

The activity of tissue MPO was assessed at the 5th h after injection of Carr into

198

the mouse right hind paw according to the method of Bani et al. [11] with some

199

modifications. Samples were placed in 0.75 mL of 80 mM phosphate-buffered

200

saline (PBS), pH 5.4, and then homogenized in a motor-driven homogenizer. The

201

homogenate was centrifuged at 12,000 × g at 4 °C for 15 min. Triplicate 0.1 mL of

202

supernatant with 2.9 mL of potassium phosphate buffer (50 mM, pH 6) containing 0.19

203

mg/mL of o-dianisidine chloride and 0.0005% H2O2 was a substrate for myeloperoxidase. 204

Oxidized o-dianisidine formed a soluble chromophore and absorbance (OD460) was 205

determined by spectrophotometry (Molecular Devices, Orleans Drive, Sunnyvale, CA)

(10)

over 2 min. Myeloperoxidase activity ( OD460) was calculated by subtracting the value 207

of OD460 at time 0 min from that at 2 min for each sample. 208

209

Measurement of TNF-α and PGE2 by an Enzyme-Linked Immunosorbent Assay

210

(ELISA). The levels of TNF- and PGE2 were determined using a commercially 211

available ELISA kit (Biosource International Inc., Camarillo, CA) according to the 212

manufacturer’s instruction. TNF- and PGE2were determined from a standard curve. 213

214

Antioxidant Enzyme Activity Measurements

215

The following biochemical parameters were analyzed to check the paw tissues 216

activity of cinnamic aldehyde by the methods given below. 217

Total SOD activity was determined by the inhibition of cytochromec reduction [12].

218

The reduction of cytochrome c was mediated by superoxide anions generated by the 219

xanthine/xanthine oxidase system and monitored at 550 nm. One unit of SOD was 220

defined as the amount of enzyme requiredto inhibit the rate of cytochrome c reduction by 221

50%. Total CAT activity was based on that of Aebi [13]. In brief, the reduction of 10mM 222

H2O2 in 20 mM of phosphate buffer (pH 7.0) was monitored by measuring the absorbance 223

at 240 nm. The activity was calculated using a molar absorption coefficient, and the 224

enzyme activities were defined as nanomoles of dissipating hydrogen peroxide per 225

milligram protein per minute. Total GPx activity in cytosol was determined according to 226

Paglia and Valentine’s method [14]. The enzyme solution was added to a mixture 227

containing hydrogen peroxide and glutathione in 0.1 mM Tris buffer (pH 7.2) and the 228

(11)

absorbance at 340 nm was measured. Activity was evaluated from a calibration curve, 229

and the enzyme activities were defined as nanomoles of NADPH oxidized per milligram 230

protein per minute. 231

232

Protein Lysate Preparation and Western blot Analysis of iNOS, COX-2, IB, and

233

NF-B

234

The stimulated murine macrophage cell line RAW264.7 cells were washed with 235

PBS and lysed in an ice-cold lysis buffer [10% glycerol, 1% Triton X-100, 1mM Na3VO4, 236

1mM EGTA, 10mM NaF, 1mM Na4P2O7, 20 mM Tris buffer (pH 7.9), 100 mM 237

-glycerophosphate, 137 mM NaCl, 5 mM EDTA, and one protease inhibitor cocktail 238

tablet (Roche, Indianapolis, IN, USA)] on ice for 1 h, followed by centrifugation at 239

12,000 rpm for 30 min at 4°C. Soft tissues were removed from individual mice paws and 240

homogenized in a solution containing 10 mM CHAPS, 1 mM phenylmethylsulphonyl 241

fluoride (PMSF), 5 g/mL, aprotinin, 1 M pepstatin and 10 M leupeptin. The 242

homogenates were centrifuged at 12,000g for 20 min, and 30 g of protein from the 243

supernatants was then separated on 10% sodium dodecylsulphate–polyacrylamide gel 244

(SDS-PAGE) and transferred to polyvinylidene difluoride membranes. After transfer, the 245

membrane was blocked for 2 h at room temperature with 5% skim milk in Tris-buffered 246

saline-Tween (TBST; 20 mM Tris, 500 mM NaCl, pH 7.5, 0.1% Tween 20). The 247

membranes were then incubated with mouse monoclonal anti-iNOS, anti-COX-2, 248

anti-IB, or anti-NF-B antibody in 5% skim milk in TBST for 2 h at room temperature. 249

The membranes were washed three times with TBST at room temperature and then 250

incubated with a 1 : 2000 dilution of anti-mouse IgG secondary antibody conjugated to 251

(12)

horseradish peroxidase (Sigma, St Louis, MO, U.S.A.) in 2.5% skim milk in TBST for 1 252

h at room temperature. The membranes were washed three times and the immunoreactive 253

proteins were detected by enhanced chemiluminescence (ECL) using hyperfilm and ECL 254

reagent (Amersham International plc., Buckinghamshire, U.K.). The results of Western 255

blot analysis were quantified by measuring the relative intensity compared to the control 256

using Kodak Molecular Imaging Software (Version 4.0.5, Eastman Kodak Company, 257

Rochester, NY) and represented in the relative intensities. 258

259

Histological Examination. For histological examination, biopsies of paws were taken 5

260

h following the interplanetary injection of Carr. The tissue slices were fixed in a solution 261

(1.85% formaldehyde, 1% acetic acid) for 1 week at room temperature, dehydrated by 262

graded ethanol and embedded in Paraffin (Sherwood Medical). Sections (thickness 5 μm) 263

were deparaffinized with xylene and stained with hematoxylin and eosin (H&E) stain. All 264

samples were observed and photographed with BH-2 Olympus microscopy. Every 3~5 265

tissue slices were randomly chosen from Carr, Indo and cinnamic aldehyde-treated (5 266

mg/kg) groups. Histological examination of these tissue slices revealed an excessive 267

inflammatory response with massive infiltration of neutrophils [ploymorphonuclear 268

leukocytes (PMNs)] by microscopy. The numbers of neutrophils were counted in each 269

scope (400 x) and thereafter obtain their average count from 5 scopes of every tissue slice 270

[15]. 271

272

Statistical Analysis. Data are expressed as mean ± standard error of the mean (SEM).

273

Statistical evaluation was carried out by one-way analysis of variance (ANOVA followed 274

(13)

by Scheffe's multiple range test). Statistical significance is expressed as *p < 0.05, **p < 275 0.01, ***p < 0.001. 276 277 Results 278

Cell Viability. The effect of C. cassia constituents (cinnamic aldehyde, cinnamic alcohol,

279

cinnamic acid, and coumarin) on RAW264.7 cell viability was determined by a MTT 280

assay. Cells cultured with samples at the concentrations (0, 6.25, 12.5, 25, and 50 M) 281

used in the presence of 100 ng/mL LPS for 24 h did not change cell viability (Fig. 1B). 282

283

Effect of Cinnamic aldehyde, Cinnamic alcohol, Cinnamic acid, and Coumarin on

284

LPS-induced NO Production in Macrophages. In the present study, effects of cinnamic

285

aldehyde, cinnamic alcohol, cinnamic acid, and coumarin on LPS-induced NO production 286

in RAW264.7 macrophages were investigated. Nitrite accumulated in the culture medium 287

was estimated by the Griess reaction as an index for NO release from the cells. After 288

treatment with LPS (100 ng/mL) for 24 h, the nitrite concentration increased in the 289

medium. When RAW264.7 macrophages were treated with different concentrations of 290

cinnamic aldehyde together with LPS for 24 h, the cinnamic aldehyde inhibited nitrite 291

production significantly (Fig. 2). Cinnamic aldehyde did not interfere with the reaction 292

between nitrite and Griess reagents at 50 M (data not shown). Unstimulated 293

macrophages, after 24 h of incubation in culture medium produced background levels of 294

nitrite. When RAW264.7 macrophages were treated with different concentrations of 295

cinnamic aldehyde (0, 6.25, 12.5, 25, and 50 M) together with LPS (100 ng/mL) for 24 296

(14)

h, a significant concentration-dependent inhibition of nitrite production was detected. 297

There was either a significant decrease in the nitrite production of group treated with 12.5 298

M cinnamic aldehyde (p < 0.05), or very or highly significant decrease of groups treated 299

respectively with 25 or 50 M of cinnamic aldehyde when compared with the LPS-alone 300

group (p < 0.01 or p < 0.001). The IC50 value for inhibition of nitrite production of 301

cinnamic aldehyde was about 45.56 ± 1.36 M 302

303

Inhibition of LPS-induced iNOS, COX-2, IB, and NF-B Protein by Cinnamic

304

aldehyde, Cinnamic alcohol, Cinnamic acid, and Coumarin. In order to investigate

305

whether the inhibition of NO production was due to a decreased iNOS, COX-2, IB,

306

and NF-B protein level, the effect of cinnamic aldehyde, cinnamic alcohol, cinnamic 307

acid, and coumarin was studied by immunoblot. The results showed the incubation with

308

cinnamic aldehyde (50 M) in the presence of LPS (100 ng/mL) for 24 h or 1h inhibited

309

iNOS, COX-2, IB, and NF-B proteins expression in mouse macrophage RAW264.7

310

cells in the cytosol (Fig. 3A and Fig. 4A). The detection of β-actin was also performed in

311

the same blot as an internal control. The intensity of protein bands was analyzed by using

312

Kodak Quantity software in three independent experiments and it showed an average of

313

77.4% and 84.8% down-regulation of iNOS and COX-2proteins, respectively, after

314

treatment with cinnamic aldehyde at 50 M compared with the LPS-alone (Fig. 3B). And

315

the intensity of protein bands showed an average of 82.6% and 86.2% up-regulation of

316

NF-BandIBprotein (p<0.001) (Fig.4B).

(15)

318

Inhibition of LPS-induced the level of TNF-and PGE2 by Cinnamic aldehyde.

319

TNF- mediates the production of many other cytokines during inflammation, in

320

particular, the production of interleukin-1 beta (IL-1 and interleukin-6 (IL-6) [16]. We

321

examined the effect of cinnamic aldehyde on LPS induced up-regulation of TNF-. A

322

very low amount of TNF- protein was detected by a specific ELISA for TNF- in

323

controls (Fig. 5A). When RAW264.7 macrophages were treated with different

324

concentrations of cinnamic aldehyde (12.5, 25, and 50 ) together with LPS (100

325

ng/mL) for 24 h, a significant concentration-dependent inhibition of TNF-production

326

was detected. There was either a significant decrease in the TNF-production of group

327

treated with 12.5  cinnamic aldehyde (p < 0.05), or highly significant decrease of

328

groups treated respectively with 25 and 50  of cinnamic aldehyde when compared

329

with the LPS-alone group (p < 0.01 or p < 0.001). The IC50 value for inhibition of TNF- 330

production of cinnamic aldehyde was about 29.58 ± 0.34 M.

331

PGE2 represents the most important inflammatory product of COX-2 activity and it 332

was quantified in cell-free culture supernatant [16]. As shown in Fig. 5B, cells were

333

stimulated with LPS alone raised significant amount of PGE2 in RAW264.7 macrophages.

334

When RAW264.7 macrophages were treated with different concentrations of cinnamic

335

aldehyde (12.5, 25, and 50 ) together with LPS (100 ng/mL) for 24 h, a significant

336

concentration-dependent inhibition of PGE2 production was detected. The IC50 value for 337

inhibition of PGE2 production of cinnamic aldehyde was about 37.67 ± 0.58 M. 338

(16)

339

Effects of Cinnamic aldehyde on Carr-induced Mice Paw Edema. In this study, we

340

used Carr-induced edema because this model is widely employed for screening the effects 341

of anti-inflammatory drugs. Carr-induced paw edema is shown in Fig. 6A. Cinnamic 342

aldehyde (5 mg/kg) inhibited (p < 0.001) the development of paw edema induced by Carr 343

after the 4th and the 5th h of treatment, significantly. Indo (10 mg/kg) significantly 344

decreased the Carr induced paw edema after the 4th and the 5th h of treatment (p < 0.001). 345

346

Effects of Cinnamic aldehyde on the MDA level. The MDA level increased

347

significantly in the edema paw at the 5th h after Carr injection (p < 0.001). However, the 348

MDA level was decreased significantly by treatment with cinnamic aldehyde (5 mg/kg) 349

( p < 0.001), as well as 10 mg/kg Indo (Fig. 6B). 350

351

Effects of Cinnamic aldehyde on the MPO activity. The MPO activity increased

352

significantly in the edema paw at the 5th h after Carr injection (p < 0.001). However, the

353

MPO activity was decreased significantly by the treatment with cinnamic aldehyde (5

354

mg/kg) (p < 0.001), as well as 10 mg/kg Indo (Fig. 6C).

355 356

Effects of Cinnamic aldehyde on the NO Level. In Fig. 6D, the NO level increased

357

significantly in the edema serum at the 5th h after Carr injection (p < 0.001). Cinnamic 358

aldehyde (5 mg/kg) significantly decreased the serum NO level (p < 0.001). The 359

inhibitory potency was similar to that of Indo (10 mg/kg) at the 5th h after induction. 360

(17)

361

Effects of Cinnamic aldehyde on the TNF-α and PGE2 Level. The TNF-α and PGE2 362

level increased significantly in serum at the 5th h after Carr injection (p < 0.001).

363

However, cinnamic aldehyde (1.25 or 2.5 mg/kg) decreased the TNF-α and PGE2 level in

364

serum at the 5th h after Carr injection (p < 0.05 or p < 0.01), as well as 10 mg/kg Indo (Fig.

365

6E and 6F).

366 367

Effects of Cinnamic aldehyde on activities of Antioxidant Enzymes. At the 5th h after 368

the intrapaw injection of Carr, paw tissues were also analyzed for the biochemical 369

parameters such as CAT, SOD, and GPx activities. CAT, SOD, and GPx activities in paw 370

tissue were decreased significantly by Carr administration. CAT, SOD, and GPx activity 371

were increased significantly after treated with 5 mg/kg cinnamic aldehyde and 10 mg/kg 372

Indo (P<0.01 or P<0.001) (Table 1). 373

374

Effects of Cinnamic aldehyde on Carr-induced iNOS, COX-2, and NF-B protein

375

expressions in Mice Paw Edema. To investigate whether the inhibition of NO

376

production was due to a decreased iNOS, COX-2, and NF-B protein level, the effect of 377

cinnamic aldehyde on iNOS, COX-2, and NF-B proteins expression were studied by 378

western blot. The results showed that injection of cinnamic aldehyde (5 mg/kg) on 379

Carr-induced for 5 h inhibited iNOS, COX-2, and NF-B proteins expression in mouse 380

paw edema (Fig. 7A). The detection of β-actin was also performed in the same blot as an 381

internal control. The intensity of protein bands was analyzed by using Kodak Quantity 382

software in three independent experiments and showed an average of 76.1% and 63.3% 383

(18)

down-regulation of iNOS and COX-2 protein respectively after treatment with cinnamic 384

aldehyde at 5 mg/kg compared with the Carr-induced alone (Fig. 7B). In addition, the 385

protein expression showed an average of 57.1% and 45.1% down-regulation of iNOS, 386

and COX-2 protein after treatment with Indo at 10 mg/kg compared with the 387

Carr-induced alone (Fig. 7B). And the intensity of protein bands showed an average of

388

87.6% up-regulation of NF-Bprotein (p<0.001) (Fig.7B). The down-regulation of 389

iNOS, COX-2, and NF-B activity of the cinnamic aldehyde (5 mg/kg) was better than 390

Indo (10 mg/kg). 391

392

Histological Examination. Paw biopsies of Carr model animals showed marked cellular

393

infiltration in the connective tissue. The infiltrates accumulated between collagen fibers 394

and into intercellular spaces. Paw biopsies of animals treated with cinnamic aldehyde (5 395

mg/kg) showed a reduction in Carr-induced inflammatory response. Actually 396

inflammatory cells were reduced in number and confined to near the vascular areas. 397

Intercellular spaces did not show any cellular infiltrations. Collagen fibers were regular in 398

shape and showed a reduction of intercellular spaces. Moreover, the hypoderm 399

connective tissue was not damaged (Fig. 8A). Neutrophils increased with Carr treatment 400

(P < 0.01). As Indo and cinnamic aldehyde (5 mg/kg) could significantly decrease the 401

neutrophils numbers as compared to the Carr-treated group (P < 0.001) (Fig. 8B). 402

403

Discussion

404

In the present study, we demonstrated anti-inflammatory activities of C. cassia 405

constituents (cinnamic aldehyde, cinnamic alcohol, cinnamic acid, and coumarin) in both 406

(19)

in in vitro and in vivo experimental systems, using LPS-stimulated RAW264.7 407

macrophages and a mouse model of topical inflammation respectively. Dual inhibitory 408

activities against iNOS, COX-2, and NF-B as shown in in vitro assays appear to confer 409

on cinnamic aldehyde a potent in vivo efficacy in mouse, Carr-induced, paw edema, 410

comparable with a potent COX inhibitor, indomethacin, suggesting its potential 411

therapeutic usage as a novel topical anti-inflammatory source of health food. 412

The pathology of inflammation is initiated by complex processes triggered by 413

microbial pathogens such as LPS, which is a prototypical endotoxin. LPS can directly 414

activate macrophages, which trigger the production of inflammatory mediators, such as 415

NO and TNF-α [17]. The pharmacological reduction of LPS-inducible inflammatory 416

mediators is regarded as one of the essential conditions to alleviate a variety of disorders 417

caused by activation of macrophages. Thus, RAW264.7 macrophages provide us with an 418

good model for anti-inflammatory drug screening and for subsequently evaluating the 419

inhibitors of the signal pathways that lead to the induction of pro-inflammatory enzymes 420

and to the production of pro-inflammatory cytokines. 421

Cinnamic aldehyde, the major constituent of leaf essential oil from C. cassia. 422

Cinnamic aldehyde has been demonstrated to exhibit anti-tumor activities, anti-bacteria 423

activities, anti LPS-induced NF-B transcriptional activities [18, 19]. Cinnamic aldehyde 424

which has  unsaturated carbonyl moiety exerted suppressive effect on toll-like 425

receptor 4 (TLR4)-mediated signaling [20]. And in this paper, we first evaluated that 426

cinnamic alcohol, cinnamic acid, and coumarin only little or less anti-inflammatory 427

activities in LPS-inducible inflammatory model in vitro. Our current results provided a 428

potential medical application in modulating inflammatory diseases. 429

(20)

As many of these conditions exhibit rapid onset and development, often resulting in 430

the failure of conventional anti-inflammatory therapies and extremely high mortality rates, 431

a simultaneous suppression of NO production pathways, as shown by cinnamic aldehyde, 432

may satisfy the so far unmet need for control of the rapid progression of the inflammatory 433

process. In vitro models such as macrophage cells or other cell lines are useful materials 434

with a steady high-level production of NO. The mechanisms by which cinnamic aldehyde 435

inhibits macrophage functions have not been elucidated. Results in vitro showed that 436

cinnamic aldehyde suppressed LPS-induced production of NO, the expression of 437

inflammatory protein products such as iNOS, COX-2, IB, and NF-B. Examination of 438

the cytotoxicity of cinnamic aldehyde in RAW264.7 macrophages using MTT assay has 439

indicated that cinnamic aldehyde even at 50 M did not affect the viability of RAW264.7 440

cells. Therefore, inhibition of LPS-induced nitrite production by cinnamic aldehyde was 441

not the result of a possible cytotoxic effect on these cells. 442

Excess amounts of NO and PGE2 play a critical role in the aggravation of chronic 443

inflammatory diseases, such as hepatic dysfunction and pulmonary disease. Recently, in

444

vitro and in vivo have indicated an existing cross talk between the release of NO and 445

prostaglandins (PGs) in the modulation of molecular mechanisms that regulate PGs

446

generating pathway [21]. Scientific papers were observed that while the production of

447

both NO and PGE2 was blocked by the NOS inhibitors in mouse macrophages

448

RAW264.7 cells, these inhibitory effects were reversed by co-incubation with the

449

precursor of NO synthesis, L-Arginine. Furthermore, inhibition of iNOS activity by

450

nonselective NOS inhibitors attenuated the release of NO and PGs simultaneously in

451

LPS-activated macrophages, which suggested that endogenously released NO from

(21)

macrophages exerted a stimulatory action on enhancing the PGs production. Conversely,

453

it has been shown that COX activation in turn modulates L-arginine-NO pathway,

454

whereas COX inhibition decreases NOS activity in human platelets [22]. These results

455

are indicative of the cross-talk between NO and PGs pathways.

456

The Carr-induced mice paw edema is a suitable test for evaluating anti-inflammatory 457

drugs and has frequently been used to assess the anti-edematous effect of natural products 458

[23]. The degree of swelling of the Carr-injected paws was maximal 3th h after injection. 459

Cinnamic aldehyde and Indo significantly inhibited the development of edema the 4th and 460

the 5th h after treatment (p<0.001). They both showed anti-inflammatory effects in 461

Carr-induced mice edema paw. It is well known that the third phase of the edema-induced 462

by Carr, in which the edema reaches its highest volume, is characterized by the presence 463

of prostaglandins and other compounds of slow reaction found that the injection of Carr 464

into the rat paw induces the liberation of bradykinin, which later induces the biosynthesis 465

of prostaglandin and other autacoids, which are responsible for the formation of the 466

inflammatory exudates [24]. 467

In the studies of the mechanism on the inflammation, NO plays an important role in 468

the Carr-induced inflammatory response [25]. Our present results confirm that 469

Carr-induced paw edema model results in the production of NO. The expression of the 470

inducible isoform of NO synthase has been proposed as an important mediator of 471

inflammation. In our study, the level of NO was decreased significantly by treatment with 472

1.25, 2.5, and 5 mg/kg cinnamic aldehyde. We suggest the anti-inflammatory mechanism 473

of cinnamic aldehyde may be through the L-arginine–NO pathway because cinnamic 474

aldehyde significantly inhibits the NO production. 475

(22)

The proinflammatory cytokines such as TNF-α and IL-1 are small secreted proteins, 476

which mediate and regulate immunity and inflammation. The production of TNF- is 477

crucial for the synergistic induction of NO synthesis in LPS-stimulated macrophages. 478

TNF-α induces a number of physiological effects including septic shock, inflammation, 479

and cytotoxicity [26]. Also, TNF- is a mediator of Carr-induced inflammatory 480

incapacitation, and is able to induce the further release of kinins and leukotrienes, which 481

is suggested to have an important role in the maintenance of long-lasting nociceptive 482

response [27]. In this study, we found that cinnamic aldehyde decreased the TNF-α level 483

in serum after Carr injection by treatment with 1.25, 2.5, and 5 mg/kg, significantly. 484

Neutrophils and macrophages are critical to the pathogenesis of acute injury, 485

rheumatoid arthritis and other inflammatory diseases [15]. The Carr-induced 486

inflammatory response has been linked to neutrophils infiltration and the production of 487

neutrophils-derived free radicals, such as hydrogen peroxide as well as the release of 488

other neutrophils-derived mediators [1]. Some researches demonstrate that inflammatory 489

effect induced by Carr is associated with free radical. Free radical, prostaglandin and NO 490

will be released when administrating with Carr for 1 ~ 6 h. ROS play an important role in 491

modulating the extent of inflammatory response and consequent tissue and cell injury. 492

MDA is a metabolic product of lipid peroxidation, the level of which is raised in 493

oxidative stress. MDA production is due to free radical attack plasma membrane. 494

Increasing evidence regarding free radical-generating agents and inflammatory processes 495

suggests that accumulation of reactive oxygen species can cause tissue injury [28]. Thus, 496

inflammatory effect would result in the accumulation of MDA. In this study, there is 497

significantly increased in CAT, SOD, and GPx activities with cinnamic aldehyde 498

(23)

treatment. Furthermore, there are significantly decreases in MDA level with cinnamic 499

aldehyde treatment. We assume the suppression of MDA production is probably due to 500

the increases of CAT, SOD, and GPx activities. 501

Activation of polymorphonuclear neutrophils (PMNs) reflects a primary

502

immunological response to invading pathogens [29]. In Carr-induced inflammation,

503

cinnamic aldehyde significantly inhibited cellular infiltration

504

(neutrophils and granulocytes) into the air-pouch fluid. Also, MPO from the neutrophil's

505

azurophilic granules is responsible for invoking tissue injury [30]. Results indicate that

506

cinnamic aldehyde has considerable potential as a therapeutic inhibitor of MPO-mediated

507

tissue damage.

508

NF-B is known to be a major transcription factor to regulate the expressions of

509

pro-inflammatory enzymes and cytokines, such as iNOS, COX-2, and TNF- [31].

510

NF-B subunits (p65 and/or p50) are normally sequestered in the cytosol as an inactive

511

complex by binding to inhibitory factor IB- in un-stimulated cells. Upon stimulation of

512

pro-inflammatory signals including LPS, IB- is phosphorylated by IB kinase (IKK)

513

and inactivated through ubiquitin-mediated degradation [32]. The resulting free NF-B is

514

translocated into the nucleus and it acts as a transcription factor. As shown in Fig. 4A, the

515

treatment with cinnamic aldehyde blocks the degradation of NF-B in LPS-induced

516

macrophage and Carr-induced paw edema. Therefore, these results suggest that cinnamic

517

aldehyde inhibits the expression of iNOS and COX-2, and thus NO production through 518

inactivation of NF-B activation. 519

The anti-inflammatory properties of cinnamic aldehyde would appear to be similar 520

to the anti-inflammatory properties of certain other essential oils deriving from certain 521

(24)

other plants. Hyptis pectinata essential oil exhibits antinociceptive 522

and anti-inflammatory activity through the inhibition of NO and PGE2 production 523

after Carr injection [33]. And, the essential oil of Cordia verbenacea significantly 524

decreased TNF-a production in Carr-injected rat paws [34]. 525

In conclusion, these results suggested that cinnamic aldehyde possessed 526

anti-inflammatory effects. The anti-inflammatory mechanism of cinnamic aldehyde may 527

be related to iNOS and it is associated with the increase in the activities of antioxidant 528

enzymes (CAT, SOD, and GPx). Cinnamic aldehyde may be used as a pharmacological 529

agent in the prevention or treatment of disease in which free radical formation in a 530 pathogenic factor. 531 532 Acknowledgement 533

The authors want to thank the financial supports from the National Science Council 534

(NSC100-2313-B-039-004- and NSC 100-2320-B-039-033-), China Medical University 535

(CMU) (CMU99-TC-35, CMU99-COL-10, and CMU100-TC-11) and Taiwan 536

Department of Heath Clinical Trial and Research Center of Excellence 537 (DOH101-TD-B-111-004). 538 539 Reference 540

(1) S. S. Huang, C. S. Chiu, H. J. Chen, et al. ―Antinociceptive activities and the 541

mechanisms of anti-inflammation of asiatic acid in mice,‖ Evidence-Based 542

Complementary and Alternative Medicine, vol. 2011, pp. 895857, 2011. 543

(2) H. Y. Chang, M. J. Sheu, C. H. et al. ―Analgesic effects and the mechanisms of 544

(25)

anti-inflammation of hispolon in mice,‖ Evidence-Based Complementary and 545

Alternative Medicine, vol. 2011, pp. 478246, 2011. 546

(3) S. C. Fang, C. L. Hsu, and G. C. Yen. ―Anti-inflammatory effects of phenolic 547

compounds isolated from the fruits of Artocarpus heterophyllus,― Journal of 548

Agricultural and Food Chemistry, vol. 56, no. 12, pp. 4463-4468, 2008. 549

(4) M. H. Huang, B. S. Wang, C. S. Chiu, et al. ―Antioxidant, antinociceptive, and 550

anti-inflammatory activities of Xanthii Fructus extract,― Journal of 551

Ethnopharmacology, vol. 135, no. 2, pp. 545-552, 2011. 552

(5) C. T. Chang, S. S. Huang, S. S. Lin, et al., ―Anti-inflammatory activities of tormentic 553

acid from suspension cells of Eriobotrya Japonica ex vivo and in vivo,― Food 554

Chemistry, vol. 127, no. 3, pp. 1131-1137, 2011. 555

(6) G. J. Huang, S. S. Huang, S. S. Lin, et al. ―Analgesic effects and the mechanisms of 556

anti-inflammation of ergostatrien-3-ol from Antrodia camphorata submerged whole 557

broth in mice,― Journal of Agricultural and Food Chemistry, vol. 58, no. 12, pp. 558

7445-7452, 2010. 559

(7) C. Tohda, N. Nakayama, F. Hatanaka, and K. Komatsu. ―Comparison of 560

anti-inflammatory activities of six Curcuma rhizomes: a possible 561

curcuminoid-independent pathway mediated by Curcuma phaeocaulis 562

extract,― Evidence-Based Complementary and Alternative Medicine, vol. 3, no. 2, pp. 563

255-260, 2006. 564

(8) Z. D. He, C. F. Qiao, Q. B. Han, et al.

565

―Authentication and quantitative analysis onthe chemical profile of cassia bark (corte 566

xcinnamomi) by high-pressure liquid chromatography,― Journal of Agricultural and 567

(26)

Food Chemistry, vol. 53, no. 7, pp. 2424-2428, 2005. 568

(9) Y. Y. Sung, T. Yoon, J. Y. Jang, S. J. Park, G. H. Jeong, and H. K. Kim. ―Inhibitory 569

effects of Cinnamomum cassia extract on atopic dermatitis-like skin lesions induced 570

by mite antigen in NC/Nga mice, ― Journal of Ethnopharmacolog, vol. 133, no. 2, pp. 571

621-628, 2011. 572

(10) H. K. Kwon, J. S. Hwang, J. S. So, et al. ―Cinnamon extract induces tumor cell 573

death through inhibition of NFkappaB and AP1,― BMC Cancer, vol. 10, 392-401, 574

2010. 575

(11) D. Bani, E. Masini, M. G. Bello, M. Bigazzi, and T. B. Sacchi. ―Relaxin protects

576

against myocardial injury caused by ischaemia and reperfusion in rat

577

heart,― American Journal of Pathology, vol. 152, no.5, pp. 1367-1376, 1998.

578

(12) L. Flohe, and F. Otting. Superoxide dismutase assays. ―Methods of Enzymology,― vol, 579

105, pp. 93–104, 1984. 580

(13) H. Aebi. Catalase in vitro. ―Methods of Enzymology,― vol, 105. pp.121–126, 1984. 581

(14) E. D. Paglia, and W. N. Valentine. ―Studies on the quantitative and qualitative 582

characterization of erythrocytes glutathione peroxidase,― Journal of Laboratory and 583

clnical medicine, vol, 70, no, 1, pp. 158–169, 1967. 584

(15) M. J. Sheu, P. Y. Chou, H. C. Cheng, et al. ―Analgesic 585

and anti-inflammatory activities of a water extract of Trachelospermum jasminoides 586

(Apocynaceae),― Journal of Ethnopharmacology, vol, 126, no. 2; pp. 332-338, 587

2009. 588

(27)

(16) C. S. Lai, J. H. Lee, C. T. Ho, et al. ―Rosmanol potently inhibits

589

lipopolysaccharide-Induced iNOS and COX-2 expression through downregulating

590

MAPK, NF-κB, STAT3 and C/EBP signaling pathways,― Journal of Agricultural

591

and Food Chemistry, vol. 57, no. 22, pp. 10990–10998, 2009. 592

(17) B. Saad, B. S. Abouatta, W. Basha, et al. ―Hypericum triquetrifolium—Derived 593

factors downregulate the production levels of LPS-Induced nitric oxide and tumor 594

necrosis factor-a in THP-1 Cells,― Evidence-Based Complementary and Alternative 595

Medicine, vol. 4, no, .pp. 425–430, 2007. 596

(18) B. Huang, H. D. Yuan, do Y. Kim, H. Y. Quan, and S. H. Chung. 597

―Cinnamaldehyde prevents adipocyte differentiation and adipogenesis via regulation 598

ofperoxisome proliferator-activated receptor-γ (PPARγ) and AMP- 599

activated protein kinase (AMPK) pathways,― Journal of Agricultural and Food 600

Chemistry, vol. 59, pp. 3666-3673, 2011. 601

(19) A. H. Reddy, J. H. Seo, S. Y. Ryu, et al. ―Cinnamaldehyde and 602

2-methoxycinnamaldehyde as NF-B inhibitors from Cinnamomum 603

cassia,― Planta Medica, vol.70, no. 9, pp. 823–827, 2004. 604

(20) H. S. Youn, J. K. Lee, Y. J. Choi, et al.

605

―Cinnamaldehyde suppresses toll-like receptor 4 activation mediated through 606

the inhibition of receptor oligomerization,― Biochemical Pharmacology, vol. 75, no. 607

2, pp. 494-502, 2008. 608

(21) I. N. Hsieh, A. S. Chang, C. M. Teng, C. C. Chen, and C. R. Yang, ―Aciculatin

609

inhibits lipopolysaccharide-mediated inducible nitric oxide synthase and

(28)

cyclooxygenase-2 expression via suppressing NF-κB and JNK/p38 MAPK activation

611

pathways,―Journal of Biomedical Science, vol. 18, pp. 28, 2011. 612

(22) R. L. Handy, and P. K. Moore, ―A comparison of the effects of L-NAME, 7-NI and

613

L-NIL on carrageenan-induced hindpaw oedema and NOS activity,― British Journal

614

of Pharmacology, vol. 123, no. 6, 1119-1126, 1998. 615

(23) A. F. Viana, I. S. Maciel, E. M. Motta, et al. ―Antinociceptive Activity of Trichilia 616

catigua Hydroalcoholic extract: new evidence on its Dopaminergic 617

Effects,― Evidence-Based Complementary and Alternative Medicine, vol. 2011, pp. 618

120820,2011. 619

(24) C. Tohda, N. Nakayama, F. Hatanaka, and K. Komatsu. ―Comparison of 620

anti-inflammatory activities of six Curcuma rhizomes: a possible 621

curcuminoid-independent pathway mediated by Curcuma phaeocaulis 622

extract,― Evidence-Based Complementary and Alternative Medicine, vol. 3, pp. 623

255–260, 2006. 624

(25) D. Salvemini, Z. Wang, D. M. Bourdon, M. K. Stern, M. G. Curne, P. T. Manning. 625

―Evidence of peroxynitrite involvement in the carrageenan induced rat paw 626

edema,― European Journal of Clinical Pharmacology, vol. 303, no. 3, pp. 217–220, 627

1996. 628

(26) K. J. Yun, D. J. Koh, S. H. Kim, et al. ―Anti-inflammatory effects of sinapic 629

acid through the suppression of inducible nitric oxide synthase, cyclooxygase-2, and 630

proinflammatory cytokines expressions via nuclear factor-kappaB 631

(29)

inactivation,― Journal of Agricultural and Food Chemistry, vol. 56, no. 21, pp. 632

10265-10272, 2008. 633

(27) C. R. Tonussi, and S. H. Ferreira. ―Tumour necrosis factor-alpha mediates carrageen 634

in-induced knee-joint incapacitation and also triggers overt nociception in previously 635

inflamed rat knee-joints,― Pain, vol. 82, no. 1, 81-87, 1999, 636

(28) W. L. Lin, C. J. Wang, Y. Y. Tsai, C. L. Liu, J. M. Hwang, T. H. Tseng. 637

―Inhibitory effect of esculetin on oxidative damage induced by t-butyl hydroperoxide 638

in rat liver,― Archives of Toxicology, vol. 74, no. 8, pp. 467-472, 2000. 639

(29) V. Wittamer, B. Bondue, A. Guillabert, G. Vassart, M. Parmentier, and D. Communi.

640

―Neutrophil-mediated maturation of chemerin: a link between innate and adaptive

641

immunity,― Journal of Immunoogyl. Vol. 175, no. 1, pp. 487-493, 2005.

642

(30) T. C. Busnardo, C. Padoani, T. C. Mora, et al., ―Anti-inflammatory evaluation of

643

Coronopus didymus in the pleurisy and paw oedema models in mice,― Journal of

644

Ethnopharmacology. Vol.128, no. 2, pp. 519-525, 2010. 645

(31) J. S. Deng, C. S. Chiu, S. S. Huang, P. H. Shie, T. H. Lin, and G. J. Huang.

646

―Antioxidant, Analgesic, and Anti-inflammatory activities of the ethanolic extracts of

647

Taxillus liquidambaricola,― Journal of Ethnopharmacology. vol. 137, no. 3, pp. 648

1161-1171, 2011.

649

(32) Karin M, and Ben-Neriah Y. ―Phosphorylation meets ubiquitination: the control of 650

NF-B activity,― Annual Review of Immunology, vol. 18, pp. 621–63, 2000. 651

(33) L. J. Raymundo, C. C. Guilhon, D. S. Alviano, et al. ―Characterisation of 652

the anti-inflammatory and antinociceptive activities of the Hyptis pectinata (L.) 653

Poitessential oil,―Journal of Ethnopharmacology. vol. 34, no.3, pp. 725-32, 2011. 654

(30)

(34) E. S. Fernandes, G. F. Passos, R. Medeiros, et al. ―Anti-inflammatory effects of 655

compounds alpha-humulene and (-)-trans-caryophyllene isolated from the essential 656

oil of Cordia verbenacea,― European Journal of Pharmacology, vol. 569, no.5, pp. 657 228-236, 2007. 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677

(31)

Figure Legends

678

Figure 1. Chemical structure of cinnamomum cassia constituents (cinnamic aldehyde, 679

cinnamic alcohol, cinnamic acid, and coumarin) (A) and cytotoxic effects of 680

cinnamomum cassia constituents in RAW264.7 cells (B). Cells were incubated for 24 h 681

with 100 ng/mL of LPS in the absence or presence of samples (0, 6.25, 12.5, 25, and 50 682

M). Samples were added 1 h before incubation with LPS (lipopolysaccharide). Cell 683

viability assay was performed using MTT assay. The data were presented as mean ± 684

S.D. for three different experiments performed in triplicate. 685

686

Figure 2. Effects of cinnamomum cassia constituents (cinnamic aldehyde, cinnamic 687

alcohol, cinnamic acid, and coumarin) on LPS-induced NO production of RAW264.7 688

macrophages. Cells were incubated for 24 h with 100 ng/mL of LPS in the absence or 689

presence of samples (0, 6.25, 12.5, 25, and 50 M). Samples were added 1 h before 690

incubation with LPS. Nitrite concentration in the medium was determined using Griess 691

reagent. The data were presented as mean ± S.D. for three different experiments 692

performed in triplicate. ###compared with sample of control group. *p < 0.05, **p < 0.01, 693

and ***p < 0.001 were compared with LPS-alone group. 694

695

Figure 3. Inhibition of iNOS and COX-2 protein expression by cinnamomum cassia 696

constituents ((cinnamic aldehyde, cinnamic alcohol, cinnamic acid, and coumarin) in 697

LPS-stimulated RAW264.7 cells. Cells were incubated for 24 h with 100 ng/mL of LPS 698

in the absence or the presence of samples (50 M). Samples were added 1 h before 699

incubation with LPS. Lysed cells were then prepared and subjected to western blotting 700

(32)

using an antibody specific for iNOS and COX-2. β-actin was used as an internal control. 701

(A) A representative western blot from two separate experiments is shown. (B) Relative 702

iNOS and COX-2 protein levels were calculated with reference to a LPS-stimulated 703

culture. ###compared with sample of control group. The data were presented as mean ± 704

S.D. for three different experiments performed in triplicate. *p < 0.05 and ***p < 0.001 705

were compared with LPS-alone group. 706

707

Figure 4. Inhibition of NF-B and IB (A) protein expressions by cinnamomum cassia

708

constituents (cinnamic aldehyde, cinnamic alcohol, cinnamic acid, and coumarin) in

709

LPS-stimulated RAW264.7 cells. Samples (50 M) were added into cells 1 h before LPS

710

(100 ng/mL) stimulation and protein samples were prepared for 1 h after LPS stimulation.

711

Activations of signaling molecules were then evaluated by Western blot analysis. Lysed 712

cells were then prepared and subjected to western blotting using an antibody specific for 713

NF-B (P65) and IB in the cytosol. β-actin was used as an internal control. A 714

representative western blot from two separate experiments is shown. Relative NF-B and

715

IB protein levels were calculated with reference to a LPS-stimulated culture (B). 716

###

compared with sample of control group. The data were presented as mean ± S.D. for 717

three different experiments performed in triplicate. *p < 0.05 and ***p < 0.001 were 718

compared with LPS-alone group. 719

720

Figure 5. The effects of cinnamic aldehyde on lipopolysaccharide (LPS)-induced TNF-

721

(A) and PGE2 (B) in LPS-stimulated RAW264.7 cells. Cells were incubated for 24 h with 722

100 ng/mL of LPS in the absence or in the presence of cinnamic aldehyde (0, 12.5, 25,

(33)

and 50 M). Cinnamic aldehyde was added 1 h before the incubation with LPS.

724

TNF-and PGE2concentrations in the medium were determined using ELISA kit. The 725

data were presented as mean ± S.D. for three different experiments performed in

726

triplicate. ###p < 0.001 compared with sample of control group.*p < 0.05, **p < 0.01, and 727

***

p < 0.001 were compared with LPS-alone group. 728

729

Figure 6. Effects of cinnamic aldehyde and Indo on hind paw edema induced by Carr in

730

mice (A), the tissue MDA (B) and MPO (C) concentrations of foot in mice, Carr-induced

731

NO (D), TNF- (E) and PGE2 (F) concentrations of serum at the 5th hr in mice. The 732

values are averaged, obtained in individual animals (n=6). Each value represents as mean 733

± S.E.M. *p < 0.05, **p < 0.01, and ***p < 0.001 as compared with the Carr group. 734

735

Figure 7. Inhibition of iNOS, COX-2, and NF-B protein expressions by cinnamic 736

aldehyde induced by Carr of foot at the 5th h in mice. Tissue suspended were then 737

prepared and subjected to western blotting using an antibody specific for iNOS, COX-2, 738

and NF-B. β-actin was used as an internal control. (A) A representative western blot 739

from two separate experiments is shown. (B) Relative iNOS, COX-2, and NF-B protein 740

levels were calculated with reference to a Carr-injected mouse. ###compared with sample 741

of control group. The data were presented as mean ± S.D. for three different 742

experiments performed in triplicate. **p < 0.01 and ***p < 0.001 were compared with 743

Carr-alone group. 744

745

Figure 8. Representative light micrographs of mouse hind footpad H&E stained to reveal 746

(34)

hemorrhage, edema and inflammatory cell infiltration in control mice (A), Carr-treated 747

mice demonstrates hemorrhage with moderately extravascular red blood cell and large 748

amount of inflammatory leukocyte mainly neutrophils infiltration in the subdermis 749

interstitial tissue of mice (B), and mice given indomethacin (Indo) (10 mg/kg) before 750

Carr. (C). Cinnamic aldehyde (5 mg/kg) significantly show morphological alterations 751

(100×) (D) and the numbers of neutrophils in each scope (400x) (E) compared to 752

subcutaneous injection of Carr only. ###p < 0.001 as compared with the control group. 753

***p < 0.001 compared with Carr group. Scale bar = 100 μm. 754 755 756 757 758 759 760 761 762 763 764 765 766 767

(35)

Figure 1.

768

A.

769 770

B.

771 Cell Vi abil ity (% ) 0 20 40 60 80 100 120 Cinnamic aldehyde Cinnamic acid Cinnamic alcohol coumarin Control - 6.25 12.5 25 50 LPS (100 ng/mL) (M) 772

(36)

773

Figure 2.

774

Nitri

te (

M)

0 2 4 6 8 10 12 14 16 18 Cinnamic aldehyde Cinnamic acid Cinnamic alcohol Courmarin Control - 6.25 12.5 25 50 LPS (100 ng/mL) (M) ### ###### ### *** ** * 775 776 777 778 779 780 781 782 783

(37)

Figure 3.

784

A.

785 786

B.

787 *** Control - 50 50 50 50 (M) ** **

inos and COX-2

(% control) 0.0 0.2 0.4 0.6 0.8 1.0 1.2 iNOS COX-2 LPS ###### *** * *** * * * Cinnamic acid Cinnamic alcohol Coumarin Cinnamic aldehyde 788

(38)

Figure 4.

789

A.

790 791

B.

792 *** Control - 50 50 50 50 (M) ** ** LPS ### *** Cinnamic acid Cinnamic alcohol Coumarin Cinnamic aldehyde *** ** NF- B and IB (% contro l) 0.0 0.2 0.4 0.6 0.8 1.0 1.2 Cytosol NF-B IB ** *** ###### *** * * 793

(39)

Figure 5.

794

A.

795 TNF- (ng/ mL) 0 20 40 60 80 100 120 Control - 12.5 25 50 LPS (100 ng/mL) *** * ### ** Cinnamic aldehyde (M) 796

B.

797 PGE 2 (p g/m L) 0 200 400 600 800 ### ** * Control - 12.5 25 50 LPS (100 ng/mL) Cinnamic aldehyde (M) *** 798

(40)

Figure 6.

799

A.

800 Time (Hr) 0 1 2 3 4 5 Changes of edema vol ume (mL) 0.00 0.02 0.04 0.06 0.08 Carr Carr and Indo

Carr and Cinnamic aldehyde 1.25 mg/kg Carr and Cinnamic aldehyde 2.5 mg/kg Carr and Cinnamic aldehyde 5.0 mg/kg

*** *** ** * *** *** ** ** 801

B.

802 Ti ssue MDA con cent ratio n (n mol /mg p rotei n) 0.0 0.3 0.6 0.9 1.2 1.5 Control - Indo 1.25 2.5 5 (mg/kg) Carr

**

***

### Cinnamic aldehyde

***

*

803

(41)

C.

804 My el opero xidase (MP O) acti vity (%) 0 20 40 60 80 100 ###

**

***

**

*

Control - Indo 1.25 2.5 5 (mg/kg) Carr Cinnamic aldehyde 805

D.

806 *** Control - Indo 1.25 2.5 5 (mg/kg) ** ** *** *** ** ** Carr ### Nit ri te (M) 0 2 4 6 8 10 12 14 16 ### ** *** Cinnamic aldehyde * ** 807

(42)

E.

808 T NF - ( p g /m L ) 0 100 200 300 400 500 600 Carr ### Control - Indo 1.25 2.5 5 (mg/kg)

***

***

**

Cinnamic aldehyde

*

809

F.

810 PGE 2 ( pg/mL) 0 20 40 60 80 100 120 ###

***

***

**

*

Control - Indo 1.25 2.5 5 (mg/kg) Carr Cinnamic aldehyde 811

(43)

812

Figure 7.

813

A.

814 815

B.

816 *** ** ** *** Control - 10 5 (mg/kg) Indo Cinnamic aldehyde

Carr ** ** *** iNO S , CO X -2, an d NF - B (% of co ntr ol ) 0.0 0.2 0.4 0.6 0.8 1.0 1.2 iNOS COX-2 Cytosol NF-kB ### ### ### *** *** *** ** ** ** 817 818

(44)

Figure 8.

819

A.

820 821

B.

822 *** Control - Indo 5 (mg/kg) Cinnamic aldehyde ** ** Neut ro p h il / sco p e (cell ) 0 20 40 60 80 100 *** *** Carr ### 823 824

數據

Figure 1. 768  A. 769  770  B.   771  Cell Viability (%) 020406080100120 Cinnamic aldehyde Cinnamic acid Cinnamic alcohol coumarin  Control          -           6.25       12.5          25           50  LPS (100 ng/mL) (M) 772
Figure 3.   784  A. 785     786  B. 787  ***     Control          -              50            50           50            50    (M)****
Figure 4.   789  A. 790  791  B. 792  ***     Control          -              50            50           50            50    (M)**** LPS### ***Cinnamic    acid  Cinnamic    alcohol Coumarin Cinnamic  aldehyde*****
Figure 5. 794  A. 795  TNF- (ng/mL) 020406080100120 Control            -               12.5            25               50 LPS (100 ng/mL) ****###** Cinnamic aldehyde (M) 796  B
+3

參考文獻

相關文件

- Multi-layer perceptron with linear, logistic and softmax outputs and appropriate error functions. - Radial basis function (RBF) networks with both Gaussian and non-local

of stupa inscriptions in his time.[31] Here I will examine a few examples of existing stupa inscriptions composed by Po Chü-yi paying special attention.. to the relationship

experiences in choral speaking, and to see a short segment of their performance at the School Speech Day... Drama Festival and In-school Drama Shows HPCCSS has a tradition

Curriculum planning - conduct holistic curriculum review and planning across year levels to ensure progressive development of students’ speaking skills in content, organisation

Therefore, it is our policy that no Managers/staff shall solicit or accept gifts, money or any other form of advantages in their course of duty respectively without the

Macro Evolution of core-collapse supernovae (giant P violation) Chiral kinetic theory. Son, Yamamoto (2012); Stephanov, Yin

Stress and energy distribution in quark-anti-quark systems using gradient flow.. Ryosuke Yanagihara

Teacher / HR Data Payroll School email system Exam papers Exam Grades /.