Capsaicin Inhibits Dimethylnitrosamine-Induced Hepatic Fibrosis by

Dec 19, 2016 - College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea. § Jeollanamdo Institute of Natural Resources Research, ...
0 downloads 0 Views 2MB Size
Subscriber access provided by University of Newcastle, Australia

Article

Capsaicin Inhibits Dimethylnitrosamine-Induced Hepatic Fibrosis by Inhibiting the TGF-#1/Smad Pathway via Peroxisome Proliferator-Activated Receptor Gamma Activation Jae Ho Choi, Sun Woo Jin, Chul Yung Choi, Hyung Gyun Kim, Gi Ho Lee, Yong An Kim, Young Chul Chung, and Hye Gwang Jeong J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.6b04805 • Publication Date (Web): 19 Dec 2016 Downloaded from http://pubs.acs.org on December 20, 2016

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 31

Journal of Agricultural and Food Chemistry 1

1

Capsaicin Inhibits Dimethylnitrosamine-Induced Hepatic Fibrosis by Inhibiting the

2

TGF-β1/Smad

3

Activation

Pathway

via

Peroxisome Proliferator-Activated Receptor

Gamma

4 5

Short title: Capsaicin inhibits DMN-induced hepatic fibrosis

6 †, ¶

†, ¶

‡, ¶

, Hyung Gyun Kim †, Gi Ho Lee †,

7

Jae Ho Choi

8

Yong An Kim †, Young Chul Chung §, Hye Gwang Jeong †,*

, Sun Woo Jin

, Chul Yung Choi

9 10



College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea

11



Jeollanamdo Institute of Natural Resources Research, Jeollanamdo, Republic of Korea

12

§

Department of Food Science, International University of Korea, Jinju, Republic of Korea



These authors contributed equally to this work.

13 14 15 16 17 18

* To whom correspondence should be addressed:

19

Hye Gwang Jeong; College of Pharmacy, Chungnam National University, Daejeon 34134,

20

Republic

21

[email protected]

of

Korea,

Tel:

+82-42-821-5936,

Fax:

22 23 24

ACS Paragon Plus Environment

+82-42-825-4936,

E-mail:

Journal of Agricultural and Food Chemistry

Page 2 of 31 2

25

ABSTRACT

26

Capsaicin (CPS) exerts many pharmacological effects, but any possible influence on liver

27

fibrosis remains unclear. Therefore, we evaluated the inhibitory effects of CPS on

28

dimethylnitrosamine (DMN) and TGF-β1-induced liver fibrosis in rats and hepatic stellate

29

cells (HSCs). CPS inhibited DMN-induced hepatotoxicity, NF-κB activation, and collagen

30

accumulation. CPS also suppressed the DMN-induced increases in α-SMA, collagen type I,

31

MMP-2, and TNF-α. In addition, CPS inhibited DMN-induced TGF-β1 expression (from 2.3

32

± 0.1 to 1.0 ± 0.1) and Smad2/3 phosphorylation (from 1.5 ± 0.1 to 1.1 ± 0.1; and from 1.6 ±

33

0.1 to 1.1 ± 0.1, respectively), by activating Smad7 expression (from 0.1 ± 0.0 to 0.9 ± 0.1)

34

via PPARγ induction (from 0.2 ± 0.0 to 0.8 ± 0.0 ) (p < 0.05). Furthermore, in HSCs, CPS

35

inhibited the TGF-β1-induced increases in α-SMA and collagen type I expression, via PPARγ

36

activation. These results indicate that CPS can ameliorate hepatic fibrosis by inhibiting the

37

TGF-β1/Smad pathway via PPARγ activation.

38 39 40

Keywords: Capsaicin; liver fibrosis; dimethylnitrosamine; PPARγ; hepatic stellate cells

41 42

ACS Paragon Plus Environment

Page 3 of 31

Journal of Agricultural and Food Chemistry 3

43

Introduction

44

The liver is responsible for the biotransformation and detoxification of exogenous and

45

endogenous metabolites and toxicants.

46

environmental toxins is associated with liver damage, ranging from a transient elevation of

47

liver enzymes to life-threatening hepatic inflammation, fibrosis, cirrhosis, and cancer. Liver

48

fibrosis is a dynamic process commonly preceded by chronic inflammation and excessive

49

secretion of matrix proteins by hepatic stellate cells. It is a pathophysiological process

50

involved in excessive extracellular matrix (ECM) deposition in response to chronic hepatic

51

damage and recovery therefrom, such as that occurring in response to the reactive and

52

reparative processes associated with chronic viral hepatitis, alcohol consumption, fat

53

accumulation, and drug abuse.

54

Hepatic stellate cells (HSCs), the main fibrogenic cell type, play an essential role in ECM

55

remodeling. Activated HSCs increase the proliferation of phenotypically transformed

56

fibroblasts, the migration of which is triggered by the binding of fibrogenic transforming

57

growth factor-beta 1 (TGF-β1) to its receptors.

58

subsequently forms a complex with Smad4 and migrates to the nucleus, where it regulates the

59

expression of genes involved in ECM synthesis and deposition.

60

activation is both an important marker of the development of liver fibrosis and a critical

61

therapeutic target.

62

Peroxisome

63

transcription factor that plays pivotal roles in various pathological processes associated with

64

inflammation, obesity, and tumorigenesis, and ECM remodeling. PPAR-γ is highly expressed

65

in quiescent HSCs of the normal liver. PPAR-γ activation inhibits the expression of α-SMA

66

and collagen by reducing HSC proliferation during hepatic fibrogenesis.

1,2

However, exposure of the liver to high levels of

3-5

proliferator-activated

6

Constitutively phosphorylated Smad2/3

receptor-gamma

(PPAR-γ)

ACS Paragon Plus Environment

is

5,7,8

a

Therefore, HSC

ligand-activated

9,10

Therefore,

Journal of Agricultural and Food Chemistry

Page 4 of 31 4

67

PPAR-γ status is a pivotal marker of the success of anti-fibrotic therapy.

68

Natural agents exhibiting anti-fibrotic activity provide an effective alternative therapy for

69

treating chronic liver diseases.

70

and less toxic hepatoprotective agents from natural sources for use in treating a variety of

71

pathologic conditions. Polyphenols are major plant metabolites of foods or nutraceuticals.

72

They are diverse in structure, but possess at least one aromatic ring bearing one or more

73

hydroxyl groups. Polyphenols reportedly protect against inflammation, obesity, and cancer by

74

exerting anti-oxidative effects and scavenging free radicals. Capsaicin (CPS) is a phenolic

75

compound of hot red peppers and chili peppers that is consumed worldwide, and has been

76

used as a spice, food additive, and drug. It has chronic anti-inflammatory and

77

chemopreventative activities in vitro. CPS counters the activities of certain mutagens and

78

exerts anticancer effects on breast, prostate, colon, and liver cancer cells. The proposed

79

molecular mechanism includes induction of apoptosis via inhibition of translocation by the

80

NF-κB and AP-1 signaling pathways. However, despite these known pharmacological

81

activities, the effects of CPS on liver fibrosis have received little attention. There is a need to

82

develop new and therapeutic agents for liver fibrosis.

83

Therefore, we investigated the inhibitory effects of CPS using an animal model of hepatic

84

fibrosis induced by dimethylnitrosamine (DMN). Our findings show that CPS inhibits the

85

development of hepatic inflammation and fibrosis via PPARγ induction and should thus be

86

explored as an alternative therapeutic agent in patients with chronic liver fibrosis.

11,12

There has also been increasing interest in developing new

87 88

Materials and methods

89

Chemicals

90

Capsaicin and DMN were obtained from Sigma Chemical (St. Louis, MO, USA). The

ACS Paragon Plus Environment

Page 5 of 31

Journal of Agricultural and Food Chemistry 5

91

following antibodies were obtained for Western blotting; anti-IκBα, anti-NF-κB p65, anti-

92

Lamin B1, anti-β-actin (Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-collagen

93

type I, anti-PPARγ (Millipore, Merck KGaA, Darmstadt, Germany), anti-Smad7, anti-TGF-

94

β1 (Abcam, Cambridge, MA, USA), anti-α-SMA (Dako, Glostrup, Denmark), anti phospho-

95

Smad2, anti-phospho-Smad3 and secondary antibodies coupled with HRP (anti-rabbit or anti-

96

mouse IgGs) (Cell Signal Technology Inc., Beverly, MA, USA). Polyvinylidene fluoride

97

membranes were purchased from Amersham Pharmacia Biotech (Piscataway, NJ, USA). The

98

Western blot detection kit was purchased from iNtRON Biotechnology Co., Ltd. (Seoul,

99

Korea).

100 101

Animals and DMN-induced liver injury

102

Male Sprague-Dawley rats (130–140 g) were obtained from Samtako (Osan, Korea). Rats

103

were acclimatized for at least 2 weeks prior to the experiments and were allowed free access

104

to a certified rodent diet (LabDiet 5002, Orient Bio Inc., Gyeonggi-Do, Korea) and tap water.

105

All experimental protocols for animal care were performed according to standard guidelines

106

and complied with the rules and regulations of the Animal Ethics Committee, Chungnam

107

National University. The rats were divided into four groups (n = 5 in each group). To induce

108

hepatic fibrosis, rats were injected intraperitoneally (i.p.) with DMN dissolved in sterile

109

saline (10 mg/kg body weight) three times per week for 4 weeks. 13 CPS dissolved in absolute

110

ethanol and then diluted with saline to yield final doses of 0.5 and 1.0 mg/kg was

111

administered to the rats intragastrically (i.g.) six times per week for 4 weeks, with treatment

112

occurring 1 h before DMN administration. Rats in the control and DMN-treated groups were

113

administered saline instead of CPS. At the end of the study, all of the animals were fasted

114

overnight and euthanized via CO2 anesthesia (Figure 1). Blood was collected and sera

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 6 of 31 6

115

separated by centrifugation. The livers were collected, blotted, and weighed. Each right lobe

116

(1 cm × 1 cm) was fixed in 10% (v/v) buffered formalin.

117 118

Biochemical analysis and ELISA

119

Hepatotoxicity was determined by measuring serum ALT and AST activities using diagnostic

120

kits (Asan Pharmaceutical Co., Seoul, Korea). Lipid peroxidation was analyzed by measuring

121

thiobarbituric acid reactive substances (TBARS). Phosphorylation of NF-κB p65 (Ser536)

122

and IκBα (Ser32) was measured using PathScan Sandwich ELISA kits (Cell Signaling

123

Technology, Beverly, MA, USA). Each absorbance was corrected by reference to that of the

124

negative control and normalized to those of NF-κB p65 and IκBα.

125 126

Histopathological examination and immunohistochemical staining

127

Histopathological examinations were carried out as described previously. 14 Histopathological

128

changes were examined by light microscopy. Microscopic fields for examination were chosen

129

randomly and viewed at a magnification of ×100. A minimum of 10 fields were scored per

130

liver slice. The extent of fibrosis was graded according to Knodell’s scoring method. 15

131 132

Hepatic collagen content

133

Hepatic collagen content was determined using the Sircol collagen assay kit. 14

134 135

HSC cell culture

136

HSC-T6, an immortalized rat HSC line, was cultured at 37°C in a 5% (v/v) carbon dioxide

137

humidified atmosphere in DMEM media (Gibco BRL, Grand Island, NY) containing 10%

138

(v/v) fetal bovine serum (FBS), streptomycin (100 µg/mL), and penicillin (100 U/mL)

ACS Paragon Plus Environment

Page 7 of 31

Journal of Agricultural and Food Chemistry 7

139 140

Semi-quantitative RT-PCR

141

Total RNA was isolated from liver tissue samples using RNAiso reagent (Takara, Kyoto,

142

Japan). After reverse transcription of 0.5 µg RNA, semi-quantitative RT-PCR was performed

143

as described previously.

144

program. The PCR primer sequences are listed in Table S1.

14

RT-PCR bands densities were obtained using NIH Image J

145 146

Western blotting

147

Hepatic protein expression levels were determined by Western blotting.

148

were obtained using NIH Image J software.

14

Band densities

149 150

Statistical analysis

151

The results are reported as the means ± SEMs. All of the data were compared by one-way

152

analysis of variance, followed by Tukey-Kramer multiple comparisons testing. A p-value less

153

than 0.05 indicates statistical significance.

154 155

RESULTS

156

CPS inhibited hepatotoxicity in DMN-treated rats.

157

Chemically induced hepatic damage is related to increased oxidative stress and lipid

158

peroxidation, which can lead to liver dysfunction. 16 Repeated injection of DMN causes liver

159

fibrosis

160

manifestations of liver injury. In rats treated for 4 weeks with DMN, the elevated serum ALT,

161

AST and TBARS levels together with reduction of body and liver weights evidenced

162

hepatotoxicity and hepatic lipid peroxidation. All of these effects were dramatically inhibited

17

and has been traditionally used to study the biochemical and pathological

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 8 of 31 8

163

by CPS treatment (Table 1).

164 165

CPS inhibited DMN-induced α-SMA and collagen I expression in rats.

166

The transactivation of quiescent HSCs is one of the hallmarks of hepatic fibrosis in the

167

fibrotic liver. Activated HSCs induce the accumulation of collagen and ECM deposition. 18

168

Repeated DMN injection results in severe liver injury with increased collagen synthesis, thick

169

fibrous bands, an inflammatory infiltrate, and hepatocytes injury in the liver, as shown by

170

staining of hematoxylin and eosin (H&E) as well as Masson’s trichrome. Chronic DMN

171

exposure also leads to the increased hepatic deposition of α-SMA, a marker of the

172

transactivation of HSCs to myofibroblasts. CPS treatment dramatically improved the

173

histopathological changes, reduced ECM deposition (Figure 2), and significantly suppressed

174

the expression of α-SMA and collagen I induced by DMN (Figure 3A, B). These results

175

indicate that CPS has the anti-fibrotic effect in rats with DMN-induced liver fibrosis.

176 177

CPS inhibited MMP-2 and TIMP-1 expression in rats with DMN-induced liver fibrosis

178

An imbalance between enhanced ECM synthesis and reduced ECM decomposition is a main

179

cause of liver fibrosis. Matrix metalloproteinase-2 (MMP-2) increases ECM synthesis during

180

hepatic fibrogenesis, but tissue inhibitor of metalloproteinase-1 (TIMP-1) controls the rate of

181

ECM degradation, and is thus an important regulator of liver fibrosis. We evaluated the

182

inhibitory effects of CPS on DMN-induced ECM-associated markers in liver tissue. Repeated

183

DMN injections induced MMP-2 expression and reduced TIMP-1 expression. However, CPS

184

dose-dependently inhibited the DMN-induced MMP-2 expression and the DMN-reduced

185

TIMP-1 expression (Figure 3C, D).

186

ACS Paragon Plus Environment

Page 9 of 31

Journal of Agricultural and Food Chemistry 9

187

CPS inhibited DMN-induced TNF-α and NF-κB activation in rats.

188

Damaged liver cells secrete cytokines that stimulate inflammation and contribute to the

189

pathogenesis of chronic liver diseases.

190

levels of DMN-induced inflammatory markers in liver tissue. The hepatic level of TNF-α, a

191

cytokine released from Kupffer cells after liver injury, increased in response to repeated

192

DMN injections. CPS treatment dose-dependently inhibited the DMN-induced increase in

193

TNF-α expression (Figure 4A). NF-κB, an important transcription factor, regulates MMP-2

194

and TNF-α expression during liver fibrogenesis. The activation of NF-κB by a variety of

195

extracellular stimuli leads to the phosphorylation, ubiquitination, and finally, proteolytic

196

degradation of the NF-κB inhibitor IκB. The subsequent release of NF-κB from IκB allows it

197

to translocate to the nucleus.

198

phosphorylation, IκBα degradation, and nuclear translocation of NF-κB p65, in a dose-

199

dependent manner (Figure 4B–D). Thus, the inactivation of NF-κB p65 is an additional

200

mechanism by which CPS inhibits liver fibrosis.

20

19

We evaluated the inhibitory effects of CPS on the

CPS inhibited DMN-induced NF-κB p65 and IκBα

201 202

CPS inhibited DMN-induced TGF-β1-dependent Smad regulation in rats.

203

TGF-β1, a potent fibrogenic cytokine, activates HSCs in the liver as part of a spectrum of

204

pathological responses, including the enhancement of α-SMA expression, ECM formation,

205

Smad activation, and collagen expression.

206

DMN-induced increase in TGF-β1 expression as well as Smad2 and Smad3 phosphorylation

207

in liver tissue, in addition to restoring the DMN-induced reduction in hepatic Smad7

208

expression (Figure 5). These results demonstrate the inhibitory effect of CPS on an

209

underlying mechanism of liver fibrosis: TGF-β1-dependent Smad2/3 activation through

210

Smad7 inactivation.

21

CPS treatment dose-dependently inhibited the

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 31 10

211 212

Effects of CPS on PPARγγ expression

213

PPARγ, a group of nuclear receptor isoforms, blocks HSC activation and thereby inhibits

214

hepatic fibrogenesis. Several studies have reported that up-regulation of PPARγ induces an

215

anti-fibrotic effect in mice and rats with CCl4- or DMN-induced liver fibrosis. 13,22 We found

216

that PPARγ expression was reduced by repeated DMN injections but was restored by CPS in

217

a dose-dependent manner (Figure 6). We also confirmed the effect of CPS on TGF-β1-

218

induced reduction in PPARγ expression. We observed no significant differences in HSC-T6

219

cell viability after CPS treatment at any concentration (data not shown). Figure 7A shows that

220

TGF-β1 reduced PPARγ expression in a concentration- and time-dependent manner. CPS

221

inhibited the TGF-β1-induced reduction in PPARγ expression in a dose-dependent manner.

222

Furthermore, a PPARγ antagonist, GW9662, partially reversed this process (Figure 7B). CPS

223

also inhibited the TGF-β1-induced increases in α-SMA and collagen type I expression; both

224

of these effects were partially reversed by GW9662 (Figure 7C, D). Thus, by inhibiting liver

225

fibrosis through PPARγ activation, CPS offers a promising therapeutic strategy for patients

226

with liver fibrosis.

227 228

DISCUSSION

229

Liver injury is induced by a wide range of factors, including alcohol abuse, obesity, chemical

230

injection, and hepatitis virus infection. Hepatic fibrosis reflects the cellular and molecular

231

wound-healing responses to liver damage. The latter is characterized by the persistent

232

production of inflammatory cytokines, proteolytic enzymes, angiogenic factors and growth

233

factors, together with enhanced ECM deposition.

234

fibrosis can progress to irreversible hepatic cirrhosis and in the end lead to organ failure

23,24

Without proper treatment, hepatic

ACS Paragon Plus Environment

Page 11 of 31

Journal of Agricultural and Food Chemistry 11

235

and/or death. Although several studies have reported considerable progress in targeting the

236

pathogenesis of hepatic fibrosis, effective therapeutic agents have yet to be developed.

237

Well-characterized inducers of hepatotoxicity include CCl4, DMN, thioacetamide, ethanol,

238

and bile duct ligation. Chemically induced liver injuries lead to liver dysfunction via the

239

production of ROS. DMN-induced hepatic fibrosis is a useful model of human liver fibrosis

240

because it mimics most of its clinical symptoms, including hepatotoxicity, inflammation, the

241

overproduction of ECM, and the characteristic histopathological changes. 25 Therefore, in this

242

study, we examined the potential therapeutic effects of CPS using DMN-induced hepatic

243

fibrosis as the experimental animal model.

244

Natural agents with good efficacy and a low risk of adverse effects have been used in the

245

prevention and treatment of hepatic inflammation, fibrosis, and cancer. CPS, one of the active,

246

pungent compounds in chili pepper, has and anti-oxidant, anti-inflammatory, and anti-obesity

247

activities.

248

deposition via the inhibition of HSC activation-on hepatic fibrosis induced by bile duct

249

ligation and CCl4 in mice was recently demonstrated.

250

TGF-β effects was not investigated. Therefore, in this study, we evaluated the inhibitory

251

effects of CPS on DMN-induced hepatic fibrosis in rats, in which TGF-β is known to play a

252

role.

253

In the chemically damaged liver, levels of ALT and AST are elevated in serum,

254

generation of ROS causes the oxidation of hepatic lipids. In this study, all three indicators

255

were increased in response to DMN-induced liver fibrosis. Our results showed that CPS

256

administration dose-dependently inhibited serum hepatotoxicity and

257

peroxidation and increased the weights of body and liver in DMN-treated rats. These results

258

are accordance with those in previously reported studies, 28,29 and demonstrate the protective

26,27

Its inhibitory effects-a reduction in α-SMA-positive cells and collagen

27

However, liver fibrosis related to

ACS Paragon Plus Environment

9

and the

hepatic lipid

Journal of Agricultural and Food Chemistry

Page 12 of 31 12

259

effect of CPS in the setting of chronic liver diseases.

260

HSC activation is a pivotal step in the development of hepatic fibrosis. Liver injury increases

261

the phenotypic transformation of HSCs to α-SMA-positive myofibroblasts and stimulates

262

cellular proliferation, the production of inflammatory cytokines, and ECM deposition.

263

Histopathological examination using H&E and Masson’s trichrome staining showed that CPS

264

prevented the extensive changes and collagen deposition in the liver that were induced by

265

DMN. Immunohistochemical staining showed that CPS attenuated the DMN-induced

266

expression of α-SMA-positive cells. Western blotting confirmed the CPS-mediated inhibition

267

of DMN-induced increases in α-SMA and collagen type I. At the mRNA level, CPS also

268

inhibited the DMN-induced MMP-2 expression and DMN reduced TIMP-1 expression.

269

Together, these results demonstrate the inhibitory effect of CPS on HSC activation by DMN.

270

In previous work, we showed that DMN-induced hepatic fibrosis is a chronic inflammatory

271

response to liver injury and that Platycodi Radix, an extract of the flowering plant Platycodon

272

grandiflorum, inhibited hepatic inflammation by reducing NF-κB activation via the induction

273

of antioxidant enzymes. 14 The present study showed that pretreatment with CPS inhibits the

274

DMN-induced increase in NF-κB activation and IκBα degradation, similar to the ability of

275

CPS to suppress PMA- or Helicobacter-pylori-induced NF-κB activation by a similar

276

mechanism. 31,32 We also demonstrated the ability of CPS to inhibit the ROS production, as

277

previously reported in HepG2 cells; in the latter study, the effect was attributed to the

278

increased nuclear translocation of Nrf2 and the expression of HO-1.

279

findings highlight the role of antioxidant enzymes and their inducers, including CPS, in

280

reducing hepatic inflammation and therefore liver fibrosis.

281

TGF-β1, a major fibrogenic cytokine in the liver, stimulates the activation of HSCs by

282

increasing the synthesis and secretion of type-1 collagen to promote ECM formation. The

ACS Paragon Plus Environment

33

30

Together, these

Page 13 of 31

Journal of Agricultural and Food Chemistry 13

283

binding of TGF-β1 to its type II receptor leads to the recruitment and phosphorylation of the

284

type I receptor and its incorporation into the complex. Phosphorylated Smad2/3 binds to

285

Smad4 to form a trimer, which then translocates to the nucleus and binds to the promoter of

286

the TGF-β-responsive element.

287

fibrotic changes in the liver by inhibiting TGF-β-induced Smad2 and Smad3 activation, as

288

previously reported in an experimental animal model of liver fibrosis.

289

administration inhibited DMN-induced increases in TGF-β1 expression and Smad2/3

290

phosphorylation, and maintained Smad7 expression. Our results imply that liver fibrosis can

291

be inhibited, at least partially, by blocking the TGF-β1/Smad signaling pathway, as achieved

292

with CPS (Figure 8).

293

PPARγ plays an important role in the transcriptional regulation of genes involved in

294

adipogenesis, fat deposition, insulin sensitivity, the inflammatory response, and hepatic

295

fibrosis.

296

quiescent, 37 and it has been shown that PPARγ agonists/ligands inhibit both liver fibrosis and

297

HSC activation by activating the PPARγ pathway. 13,38 Previous reports described the anti-

298

fibrogenic effect of curcumin in CCl4-induced liver fibrosis. Curcumin’s mechanism of action

299

includes the attenuation of oxidative stress, suppression of inflammation, inhibition of HSC

300

proliferation, induction of apoptosis, and suppression of ECM production via PPARγ

301

activation.

302

expression, in agreement with the reported ability of CPS to inhibit TGF-β1 secretion, as well

303

as collagen secretion and expression in HSCs by activating PPARγ expression.

304

findings thus demonstrate that CPS is able to prevent DMN-induced liver fibrosis by acting

305

on several of the responsible pathways (Figure 8).

306

A clinical study reported that CPS-induced satiety may be involved in pain from

36

8,34

Smad7 inhibits the trans-differentiation of HSCs and

35

In our study, CPS

In the liver, PPARγ induces a phenotypic switch in HSCs, from activated to

39,40

Our results showed that CPS administration restored DMN-reduced PPARγ

ACS Paragon Plus Environment

41

Our

Journal of Agricultural and Food Chemistry

Page 14 of 31 14

42

307

gastrointestinal stress,

308

appropriate dosing of CPS for treating of patients with liver fibrosis. Therefore, CPS should

309

be further studied on the proper management and use as a candidate anti-fibrotic agent before

310

the clinical application for patients with liver fibrosis.

and it indicates that there is still an important task to determine

311 312 313

ABBREVIATIONS USED

314

ALT, alanine aminotransferase; α-SMA, alpha-smooth muscle actin; AST, aspartate

315

aminotransferase; CPS, Capsaicin; DMN , dimethylnitrosamine; ECM, extracellular matrix;

316

HSCs, hepatic stellate cells; PPARγ, Peroxisome proliferator-activated receptor-γ; TBARS,

317

thiobarbituric acid reactive substances; TGF-β1, transforming growth factor-beta 1; TIMP-1,

318

tissue inhibitor of metalloproteinase-1; ROS, reactive oxygen species.

319 320 321

ACKNOWLEDGEMENTS

322

This research was supported by Basic Science Research Program through the National

323

Research Foundation of Korea (NRF) funded by the Ministry of Education (2009-0093815),

324

Republic of Korea.

325 326 327

Notes

328

The authors declare no competing financial interest.

329

ACS Paragon Plus Environment

Page 15 of 31

Journal of Agricultural and Food Chemistry 15

330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377

References (1) Ingawale, D. K.; Mandlik, S. K.; Naik, S. R., Models of hepatotoxicity and the underlying cellular, biochemical and immunological mechanism(s): a critical discussion. Environmental toxicology and pharmacology 2014, 37, 118-33. (2) Shin, S. M.; Yang, J. H.; Ki, S. H., Role of the Nrf2-ARE pathway in liver diseases. Oxidative medicine and cellular longevity 2013, 2013, 763257. (3) Duarte, S.; Baber, J.; Fujii, T.; Coito, A. J., Matrix metalloproteinases in liver injury, repair and fibrosis. Matrix biology : journal of the International Society for Matrix Biology 2015, 44-46, 147-56. (4) Karsdal, M. A.; Manon-Jensen, T.; Genovese, F.; Kristensen, J. H.; Nielsen, M. J.; Sand, J. M.; Hansen, N. U.; Bay-Jensen, A. C.; Bager, C. L.; Krag, A.; Blanchard, A.; Krarup, H.; Leeming, D. J.; Schuppan, D., Novel insights into the function and dynamics of extracellular matrix in liver fibrosis. American journal of physiology. Gastrointestinal and liver physiology 2015, 308, G807-30. (5) Pinzani, M., Pathophysiology of Liver Fibrosis. Digestive diseases 2015, 33, 492-7. (6) Ding, N.; Yu, R. T.; Subramaniam, N.; Sherman, M. H.; Wilson, C.; Rao, R.; Leblanc, M.; Coulter, S.; He, M.; Scott, C.; Lau, S. L.; Atkins, A. R.; Barish, G. D.; Gunton, J. E.; Liddle, C.; Downes, M.; Evans, R. M., A vitamin D receptor/SMAD genomic circuit gates hepatic fibrotic response. Cell 2013, 153, 601-13. (7) Hong, S. W.; Jung, K. H.; Lee, H. S.; Zheng, H. M.; Choi, M. J.; Lee, C.; Hong, S. S., Suppression by fucoidan of liver fibrogenesis via the TGF-beta/Smad pathway in protecting against oxidative stress. Bioscience, biotechnology, and biochemistry 2011, 75, 833-40. (8) Lin, X.; Chen, Y.; Lv, S.; Tan, S.; Zhang, S.; Huang, R.; Zhuo, L.; Liang, S.; Lu, Z.; Huang, Q., Gypsophila elegans isoorientin attenuates CCl(4)-induced hepatic fibrosis in rats via modulation of NF-kappaB and TGF-beta1/Smad signaling pathways. International immunopharmacology 2015, 28, 305-12. (9) Wang, C. Y.; Liu, Q.; Huang, Q. X.; Liu, J. T.; He, Y. H.; Lu, J. J.; Bai, X. Y., Activation of PPARgamma is required for hydroxysafflor yellow A of Carthamus tinctorius to attenuate hepatic fibrosis induced by oxidative stress. Phytomedicine : international journal of phytotherapy and phytopharmacology 2013, 20, 592-9. (10)Zhang, X.; Han, X.; Yin, L.; Xu, L.; Qi, Y.; Xu, Y.; Sun, H.; Lin, Y.; Liu, K.; Peng, J., Potent effects of dioscin against liver fibrosis. Scientific reports 2015, 5, 9713. (11)Greiner, A. K.; Papineni, R. V.; Umar, S., Chemoprevention in gastrointestinal physiology and disease. Natural products and microbiome. American journal of physiology. Gastrointestinal and liver physiology 2014, 307, G1-15. (12)Zhang, A.; Sun, H.; Wang, X., Recent advances in natural products from plants for treatment of liver diseases. European journal of medicinal chemistry 2013, 63, 570-7. (13)Lee, M. F.; Liu, M. L.; Cheng, A. C.; Tsai, M. L.; Ho, C. T.; Liou, W. S.; Pan, M. H., Pterostilbene inhibits dimethylnitrosamine-induced liver fibrosis in rats. Food chemistry 2013, 138, 802-7. (14)Choi, J. H.; Jin, S. W.; Kim, H. G.; Khanal, T.; Hwang, Y. P.; Lee, K. J.; Choi, C. Y.; Chung, Y. C.; Lee, Y. C.; Jeong, H. G., Platycodi Radix attenuates dimethylnitrosamine-induced liver fibrosis in rats by inducing Nrf2-mediated antioxidant enzymes. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 2013, 56, 231-9. (15)Moragas, A.; Garcia-Bonafe, M.; Sans, M.; Toran, N.; Huguet, P.; Martin-Plata, C.,

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 16 of 31 16

378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425

Image analysis of dermal collagen changes during skin aging. Analytical and quantitative cytology and histology 1998, 20, 493-9. (16)Zheng, X. Y.; Yang, Y. F.; Li, W.; Zhao, X.; Sun, Y.; Sun, H.; Wang, Y. H.; Pu, X. P., Two xanthones from Swertia punicea with hepatoprotective activities in vitro and in vivo. Journal of ethnopharmacology 2014, 153, 854-63. (17)Guengerich, F. P.; Kim, D. H.; Iwasaki, M., Role of human cytochrome P-450 IIE1 in the oxidation of many low molecular weight cancer suspects. Chemical research in toxicology 1991, 4, 168-79. (18)Suarez-Cuenca, J. A.; Chagoya de Sanchez, V.; Aranda-Fraustro, A.; Sanchez-Sevilla, L.; Martinez-Perez, L.; Hernandez-Munoz, R., Partial hepatectomy-induced regeneration accelerates reversion of liver fibrosis involving participation of hepatic stellate cells. Experimental biology and medicine 2008, 233, 827-39. (19)Tipoe, G. L.; Leung, T. M.; Liong, E. C.; Lau, T. Y.; Fung, M. L.; Nanji, A. A., Epigallocatechin-3-gallate (EGCG) reduces liver inflammation, oxidative stress and fibrosis in carbon tetrachloride (CCl4)-induced liver injury in mice. Toxicology 2010, 273, 45-52. (20)Chen, H. J.; Kang, S. P.; Lee, I. J.; Lin, Y. L., Glycyrrhetinic acid suppressed NFkappaB activation in TNF-alpha-induced hepatocytes. Journal of agricultural and food chemistry 2014, 62, 618-25. (21)Fang, L.; Huang, C.; Meng, X.; Wu, B.; Ma, T.; Liu, X.; Zhu, Q.; Zhan, S.; Li, J., TGF-beta1-elevated TRPM7 channel regulates collagen expression in hepatic stellate cells via TGF-beta1/Smad pathway. Toxicology and applied pharmacology 2014, 280, 335-44. (22)Moran-Salvador, E.; Titos, E.; Rius, B.; Gonzalez-Periz, A.; Garcia-Alonso, V.; Lopez-Vicario, C.; Miquel, R.; Barak, Y.; Arroyo, V.; Claria, J., Cell-specific PPARgamma deficiency establishes anti-inflammatory and anti-fibrogenic properties for this nuclear receptor in non-parenchymal liver cells. Journal of hepatology 2013, 59, 1045-53. (23)Lee, U. E.; Friedman, S. L., Mechanisms of hepatic fibrogenesis. Best practice & research. Clinical gastroenterology 2011, 25, 195-206. (24)Lee, Y.; Friedman, S. L., Fibrosis in the liver: acute protection and chronic disease. Progress in molecular biology and translational science 2010, 97, 151-200. (25)Kao, H. W.; Chen, C. L.; Chang, W. Y.; Chen, J. T.; Lin, W. J.; Liu, R. S.; Wang, H. E., (18)F-FBHGal for asialoglycoprotein receptor imaging in a hepatic fibrosis mouse model. Bioorganic & medicinal chemistry 2013, 21, 912-21. (26)Cheng, Q.; Li, N.; Chen, M.; Zheng, J.; Qian, Z.; Wang, X.; Huang, C.; Xu, S.; Shi, G., Cyclooxygenase-2 promotes hepatocellular apoptosis by interacting with TNFalpha and IL-6 in the pathogenesis of nonalcoholic steatohepatitis in rats. Dig Dis Sci 2013, 58, 2895-902. (27)Tang, J.; Luo, K.; Li, Y.; Chen, Q.; Tang, D.; Wang, D.; Xiao, J., Capsaicin attenuates LPS-induced inflammatory cytokine production by upregulation of LXRalpha. International immunopharmacology 2015, 28, 264-9. (28)Bitencourt, S.; Stradiot, L.; Verhulst, S.; Thoen, L.; Mannaerts, I.; van Grunsven, L. A., Inhibitory effect of dietary capsaicin on liver fibrosis in mice. Molecular nutrition & food research 2015, 59, 1107-16. (29)Shubha, M. C.; Reddy, R. R.; Srinivasan, K., Antilithogenic influence of dietary capsaicin and curcumin during experimental induction of cholesterol gallstone in mice. Applied physiology, nutrition, and metabolism = Physiologie appliquee,

ACS Paragon Plus Environment

Page 17 of 31

Journal of Agricultural and Food Chemistry 17

426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472

nutrition et metabolisme 2011, 36, 201-9. (30)Zhao, Q.; Qin, C. Y.; Zhao, Z. H.; Fan, Y. C.; Wang, K., Epigenetic modifications in hepatic stellate cells contribute to liver fibrosis. The Tohoku journal of experimental medicine 2013, 229, 35-43. (31)Han, S. S.; Keum, Y. S.; Chun, K. S.; Surh, Y. J., Suppression of phorbol esterinduced NF-kappaB activation by capsaicin in cultured human promyelocytic leukemia cells. Archives of pharmacal research 2002, 25, 475-9. (32)Lee, I. O.; Lee, K. H.; Pyo, J. H.; Kim, J. H.; Choi, Y. J.; Lee, Y. C., Antiinflammatory effect of capsaicin in Helicobacter pylori-infected gastric epithelial cells. Helicobacter 2007, 12, 510-7. (33)Joung, E. J.; Li, M. H.; Lee, H. G.; Somparn, N.; Jung, Y. S.; Na, H. K.; Kim, S. H.; Cha, Y. N.; Surh, Y. J., Capsaicin induces heme oxygenase-1 expression in HepG2 cells via activation of PI3K-Nrf2 signaling: NAD(P)H:quinone oxidoreductase as a potential target. Antioxidants & redox signaling 2007, 9, 2087-98. (34)Hung, W. L.; Tsai, M. L.; Sun, P. P.; Tsai, C. Y.; Yang, C. C.; Ho, C. T.; Cheng, A. C.; Pan, M. H., Protective effects of garcinol on dimethylnitrosamine-induced liver fibrosis in rats. Food & function 2014, 5, 2883-91. (35)Yao, Q. Y.; Xu, B. L.; Wang, J. Y.; Liu, H. C.; Zhang, S. C.; Tu, C. T., Inhibition by curcumin of multiple sites of the transforming growth factor-beta1 signalling pathway ameliorates the progression of liver fibrosis induced by carbon tetrachloride in rats. BMC complementary and alternative medicine 2012, 12, 156. (36)Sahebkar, A.; Chew, G. T.; Watts, G. F., New peroxisome proliferator-activated receptor agonists: potential treatments for atherogenic dyslipidemia and nonalcoholic fatty liver disease. Expert opinion on pharmacotherapy 2014, 15, 493-503. (37)Hazra, S.; Xiong, S.; Wang, J.; Rippe, R. A.; Krishna, V.; Chatterjee, K.; Tsukamoto, H., Peroxisome proliferator-activated receptor gamma induces a phenotypic switch from activated to quiescent hepatic stellate cells. The Journal of biological chemistry 2004, 279, 11392-401. (38)Miyahara, T.; Schrum, L.; Rippe, R.; Xiong, S.; Yee, H. F., Jr.; Motomura, K.; Anania, F. A.; Willson, T. M.; Tsukamoto, H., Peroxisome proliferator-activated receptors and hepatic stellate cell activation. The Journal of biological chemistry 2000, 275, 3571522. (39)Fu, Y.; Zheng, S.; Lin, J.; Ryerse, J.; Chen, A., Curcumin protects the rat liver from CCl4-caused injury and fibrogenesis by attenuating oxidative stress and suppressing inflammation. Molecular pharmacology 2008, 73, 399-409. (40)Zheng, S.; Chen, A., Curcumin suppresses the expression of extracellular matrix genes in activated hepatic stellate cells by inhibiting gene expression of connective tissue growth factor. American journal of physiology. Gastrointestinal and liver physiology 2006, 290, G883-93. (41)Bitencourt, S.; de Mesquita, F. C.; Caberlon, E.; da Silva, G. V.; Basso, B. S.; Ferreira, G. A.; de Oliveira, J. R., Capsaicin induces de-differentiation of activated hepatic stellate cell. Biochemistry and cell biology = Biochimie et biologie cellulaire 2012, 90, 683-90. (42)van Avesaat, M.; Troost, F. J.; Westerterp-Plantenga, M. S.; Helyes, Z.; Le Roux, C. W.; Dekker, J.; Masclee, A. A.; Keszthelyi, D. Capsaicin-induced satiety is associated with gastrointestinal distress but not with the release of satiety hormones. The American journal of clinical nutrition 2016, 103, 305-13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 18 of 31 18

473 474

Table 1. Effects of CPS on DMN-induced hepatotoxicity in rats. ALT (IU/L)

AST (IU/L)

TBARS (nmole/g liver)

Control

42.0 ± 1.5

70.7 ± 1.4

3.6 ± 0.4

DMN 10mg/kg

119 ± 4.5

DMN + CPS 0.5 mg/kg

107 ± 9.5

149 ± 16.3

9.0 ± 0.2

DMN + CPS 1.0 mg/kg

85.4 ± 9.7 *

100 ± 9.9 *

5.7 ± 0.9 *

#

166 ± 17.8

#

9.5 ± 0.7

#

475

476

Results are expressed as the means ± SEM. # P < 0.05, significantly different from the control

477

group. * P < 0.05, significantly different from the DMN-treated group.

478

479

ACS Paragon Plus Environment

Page 19 of 31

Journal of Agricultural and Food Chemistry 19

480 481

Table 2. Effects of CPS on DMN-induced body and liver weights in rats. Liver/Body weight (g/body weight) 4.21 ± 0.05

Body weight (g)

Liver weight (g)

Control

305 ± 28.4

12.6 ± 1.4

DMN 10mg/kg

198 ± 18.9

DMN + CPS 0.5 mg/kg

212 ± 20.6

6.6 ± 0.6

3.09 ± 0.45

DMN + CPS 1.0 mg/kg

251 ± 24.7 *

7.9 ± 0.7 *

3.52 ± 0.43 *

#

6.0 ± 0.5

#

2.93 ± 0.01

#

482 483 484

Results are expressed as the means ± SEM. # P < 0.05, significantly different from the control

485

group. * P < 0.05, significantly different from the DMN-treated group.

486 487 488

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 20 of 31 20

489

Figure Legends

490

Figure 1. Schematic diagram of the experimental protocol. Rats were divided into four

491

groups of five rats each. Hepatic fibrosis was induced by dissolving DMN in sterile saline

492

(10 mg/kg body weight) and then injecting the rats intraperitoneally (i.p.) three times per

493

week for 4 weeks. Capsaicin (CPS) was dissolved in absolute ethanol, diluted in saline and

494

then administered to the rats intragastrically (i.g.) at doses of 0.5 and 1.0 mg/kg/day six times

495

per week for 4 weeks. Control and DMN-treated groups were administered saline alone (i.g.)

496

without CPS. The animals were euthanized on day 29.

497 498

Figure 2. Effects of CPS on DMN-induced histopathological changes and hepatic

499

collagen content. (A) Liver tissue was collected and fixed in 10% formaldehyde. Thin

500

sections (5 µm) were cut and then stained with hematoxylin and eosin (H&E) and Masson’s

501

trichrome (MT). α-SMA expression was detected immunohistochemically. (B) Fibrosis

502

scores were determined by morphometric analysis of the computerized images of MT-stained

503

liver sections. (C) Liver collagen content was determined by assaying total soluble collagen

504

using the Sircol collagen assay kit according to the manufacturer’s directions.

505

significantly different from the control group. * P < 0.05, significantly different from the

506

DMN-treated group.

#

P < 0.05,

507 508

Figure 3. Effects of CPS on DMN-induced α-SMA, collagen type I, MMP-2, and TIMP-

509

1 expression. (A) Total protein extracted from liver tissue was subjected to Western blotting

510

to determine α-SMA, collagen type I, and β-actin levels. (B) The intensities of the α-SMA

511

and collagen type I bands were measured using the NIH Image J program. (C) Total RNA

512

extracted from liver tissue was used in a RT-PCR to determine the levels MMP-2, TIMP-1,

ACS Paragon Plus Environment

Page 21 of 31

Journal of Agricultural and Food Chemistry 21

513

and β-actin mRNA. (D) The intensities of the MMP-2 and TIMP-1 RT-PCR bands were

514

measured using the NIH Image J program. # P < 0.05, significantly different from the control

515

group. * P < 0.05, significantly different from the DMN-treated group

516 517

Figure 4. Effects of CPS on DMN-induced TNF-α and NF-κB activation. (A) Total RNA

518

from liver tissue was subjected to RT-PCR to measure the levels of mRNAs encoding TNF-α

519

and β-actin. (B) Nuclear protein extracted from liver tissue was analyzed by Western blotting

520

to determine NF-κB and laminin B1 levels. Total protein extracted from liver tissue was also

521

subjected to Western blotting to determine IκBα and β-actin levels. (C) The intensities of the

522

bands were measured using the NIH Image J program. (D) Liver tissues were lysed and the

523

levels of p-NF-κB P65 (Ser536) and p-IκBα (Ser32) detected using PathScan Sandwich

524

ELISA kits.

525

significantly different from the DMN-treated group.

#

P < 0.05, significantly different from the control group. * P < 0.05,

526 527

Figure 5. Effects of CPS on the DMN-induced Smad2/3 phosphorylation and DMN-

528

induced reduction in Smad7 expression. (A) Total protein extracted from liver tissue was

529

subjected to Western blotting to determine Smad2 and Smad3 phosphorylation and Smad7

530

and β-actin levels. (B) The intensities of the bands were measured using the NIH Image J

531

program. # P < 0.05, significantly different from the control group. * P < 0.05, significantly

532

different from the DMN-treated group.

533 534

Figure 6. Effects of CPS on the DMN-induced increase in TGF-β1 expression and

535

reduction in PPARγ expression. (A) Total protein extracted from liver tissue was subjected

536

to Western blotting to determine TGF-β1, PPARγ, and β-actin levels. (B) The intensities of

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 22 of 31 22

537

the bands were measured using the NIH Image J program. # P < 0.05, significantly different

538

from the control group. * P < 0.05, significantly different from the DMN-treated group.

539 540

Figure 7. Effects of CPS on TGF-β β 1-induced changes in α-SMA and collagen type I

541

expression via PPARγγ activation in HSC-T6 cells. (A) Cells were treated with various

542

concentrations of TGF-β1 for 24 h or with TGF-β1 (5 ng/mL) for 6, 12, and 24 h. (B–D)

543

Cells were pretreated with various levels of CPS for 1 h and then stimulated with TGF-β1 (5

544

ng/mL) for 24 h. Cells were pretreated with GW9662 (10 µM) for 1h and then with CPS (10

545

µM) for 1 h. Finally, cells were stimulated with TGF-β1 (5 ng/mL) for 24 h. Total protein

546

extracts were subjected to Western blotting to determine PPARγ, α-SMA, and β-actin levels.

547

Total RNAs from liver tissue were subjected to RT-PCR to measure the levels of mRNAs

548

encoding collagen type I and β-actin.

549 550

Figure 8. Effect of CPS on DMN-induced liver fibrosis. CPS reduced hepatotoxicity,

551

inflammation, and hepatic stellate cell (HSC) activation during hepatic fibrogenesis.

552

Especially, CPS ultimately inhibited HSC activation via a mechanism involving the TGF-

553

β/Smad2/3/7 and PPARγ signaling pathways. This is strong evidence that CPS may be useful

554

in the treatment of liver fibrosis. The arrows indicate stimulatory effects and the lines indicate

555

suppressive effects.

556 557 558 559

ACS Paragon Plus Environment

Page 23 of 31

Journal of Agricultural and Food Chemistry 23

560 561

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 24 of 31 24

562

ACS Paragon Plus Environment

Page 25 of 31

Journal of Agricultural and Food Chemistry 25

563

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 26 of 31 26

564

ACS Paragon Plus Environment

Page 27 of 31

Journal of Agricultural and Food Chemistry 27

565

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 28 of 31 28

566 567

ACS Paragon Plus Environment

Page 29 of 31

Journal of Agricultural and Food Chemistry 29

568 569

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 30 of 31 30

570 571

ACS Paragon Plus Environment

Page 31 of 31

Journal of Agricultural and Food Chemistry

DMN Capsaicin Oxidative stress 

PPAR Liver fibrosis  -SMA ↑ Col1 ↑ ECM ↑

Hepatotoxicity  ALT/AST ↑ Lipid peroxidation ↑

HSC activation Inflammation  NF-B ↑ TNF- ↑

ACS Paragon Plus Environment