Suppression of Lipopolysaccharide-Induced ... - ACS Publications

Dec 29, 2016 - and Hee-Sun Kim*,†. †. Department of Molecular Medicine and Tissue Injury Defense Research Center and. §. Department of Surgery, E...
0 downloads 0 Views 1MB Size
Subscriber access provided by UNIV OF CALIFORNIA SAN DIEGO LIBRARIES

Article

Suppression of LPS-induced Neuroinflammation by Morin via MAPK, PI3K/Akt, and PKA/HO-1 Signaling Pathway Modulation Ji-Sun Jung, Min-Ji Choi, Yu Young Lee, Byung-In Moon, Jin-Sun Park, and Hee-Sun Kim J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.6b05147 • Publication Date (Web): 29 Dec 2016 Downloaded from http://pubs.acs.org on December 30, 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 36

Journal of Agricultural and Food Chemistry

1

Suppression of LPS-induced Neuroinflammation by Morin via MAPK,

2

PI3K/Akt, and PKA/HO-1 Signaling Pathway Modulation

3 4

Ji-Sun Jung1,2, Min-Ji Choi1, Yu Young Lee1, Byung-In Moon3, Jin-Sun Park1,

5

Hee-Sun Kim1,*

6 7

1

8

Womans University Medical School, Seoul, Republic of Korea

9

2

Department of Molecular Medicine and Tissue Injury Defense Research Center, Ewha

Division of Functional Food Research, Korea Food Research Institute, Gyeonggi-do,

10

Republic of Korea

11

3

12

Korea

Department of Surgery, Ewha Womans University Medical School, Seoul, Republic of

13 14

Running title: Anti-inflammatory Mechanism of Morin in Microglia

15 16 17

*Corresponding Author: Hee-Sun Kim, Department of Molecular Medicine, Ewha

18

Womans University School of Medicine, 1071, Anyangchen-ro, Yangchun-Gu, Seoul

19

07985, South Korea

20

Tel: 82-2-2650-5823

21

Fax: 82-2-2653-8891

22

Email: [email protected]

23

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

24

Abstract

25

Morin is a flavonoid isolated from certain fruits and Chinese herbs and is known to

26

possess various medicinal properties. In this study, we investigated the anti-

27

inflammatory effects of morin on lipopolysaccharide (LPS)-induced microglial

28

activation, both in vitro and in vivo. We found that morin inhibited inducible nitric

29

oxide synthase (iNOS) and proinflammatory cytokines in LPS-stimulated BV2

30

microglial cells. Furthermore, morin suppressed the microglial activation and the

31

cytokine expression in the brains of LPS-stimulated mice. Subsequent mechanistic

32

studies revealed that morin inhibited the action of LPS-activated MAP kinases

33

(MAPK), protein kinase B (Akt) phosphorylation, nuclear factor-κB (NF-κB), and

34

activating protein-1 (AP-1). Further, the phosphorylation and the DNA binding activity

35

of cAMP responsive element binding protein (CREB) was enhanced by morin.

36

Moreover, morin suppressed the LPS-induced expression of nicotinamide adenine

37

dinucleotide phosphate (NADPH) oxidase subunits while it increased heme

38

oxygenase-1 (HO-1) expression and nuclear factor-E2-related factor-2 (Nrf-2)

39

activation. Therefore, our data suggest that morin exerts anti-inflammatory effects in

40

LPS-stimulated microglia by downregulating MAPK and PI3K/Akt signaling pathways

41

while upregulating PKA/CREB and Nrf2/HO-1 signaling pathways.

42 43

Keywords: Morin, Microglia, Neuroinflammation, Anti-inflammation, Molecular

44

mechanisms

45

2

ACS Paragon Plus Environment

Page 2 of 36

Page 3 of 36

Journal of Agricultural and Food Chemistry

46

Introduction

47

Microglia are immune cells found in the central nervous system (CNS) that

48

perform homeostatic functions such as phagocytosis of apoptotic cells and debris

49

throughout the CNS and support neuronal survival during brain development.1,2 There

50

are two types of microglial cells, M1 and M2 cells. Activated M1 microglia initiate

51

inflammation processes in the body through the production of pro-inflammatory

52

molecules such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), nitric oxide

53

(NO), and reactive oxygen species (ROS), which can exacerbate brain injury.3,4

54

Activated M2 microglia produce anti-inflammatory cytokines that reduce inflammation

55

and repair tissue.5-7 Therefore, maintenance of the balance between the inflammatory

56

M1 cells and anti-inflammatory M2 cells is crucial for homeostasis in the brain.

57

However, prolonged or excessive microglial activation induces neuroinflammation,

58

resulting in a homeostatic imbalance, that can cause neurodegenerative disorders.5

59

Therefore, the inhibition of excessive microglial activation has been suggested as

60

potential therapy for brain injury and various neurodegenerative diseases such as

61

Parkinson’s disease (PD) and Alzheimer’s disease (AD).

62

Morin (2′,3,4′,5,7-pentahydroxyflavone) is a well-known flavonoid that is

63

naturally found in various fruits (for e.g., osage orange), almonds, red wines, and many

64

Chinese medicinal herbs.8,9 Numerous studies have recently reported antioxidant, anti-

65

apoptotic, and anti-inflammatory properties of morin. It has been found to be effective

66

against lung injury by the suppression of the lung NLRP3 inflammasome.9 A different

67

study reported the anti-inflammatory effect of morin on gastric mucosal damage via

68

the modulation of the nuclear factor NF-κB signal transduction pathway in mammary

69

epithelial cells.8,10 Further, recent studies have reported the protective and antioxidative

3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 36

70

effects of morin in mice models of cisplatin-induced kidney and acrylamide-induced

71

hepatic injuries. 11,12 Morin has been shown to target multiple pathogenic mechanisms

72

in AD model mice. It inhibits tau hyperphosphorylation and glial activation, promotes

73

amyloid-β (Aβ) degradation, and enhances synaptic protein expression.13,14 In addition,

74

morin suppresses autophagic signaling by inhibiting the production of cytokines and

75

nitric oxide.15

76

Although numerous studies have described the pharmacological effects of morin

77

in various disease models and cell types, details of the underlying molecular

78

mechanisms of its anti-inflammatory effects in activated microglia have not been

79

reported so far. Therefore, we examine the underlying molecular mechanisms of the

80

anti-inflammatory and antioxidant effects of morin in LPS-induced BV2 microglial

81

cells in the current study. We investigate the anti-inflammatory effects of morin in vivo

82

by using a sepsis model induced by the peripheral administration of LPS, based on

83

previous

84

neurodegeneration caused by systemic LPS.16,17

reports

that

demonstrate

neuroinflammation

85

4

ACS Paragon Plus Environment

and

progressive

Page 5 of 36

Journal of Agricultural and Food Chemistry

86

Materials and Methods

87

Chemicals

88

Morin hydrate was purchased from Sigma-Aldrich Biotechnology (St. Louis, MO,

89

USA). Reagents used for cell cultures were obtained from Gibco BRL (Grand Island,

90

NY, USA). LPS (Escherichia coli serotype 055:B5) was purchased from Sigma-

91

Aldrich Biotechnology (St. Louis, MO, USA). Antibodies against heme oxygenase-1

92

(HO-1), TNF-α, cyclooxygenase 2 (COX-2), nuclear factor erythroid 2-related factor 2

93

(Nrf2), and interleukin-6 (IL-6) were purchased from Santa Cruz Biotechnology (Santa

94

Cruz, CA, USA). Antibodies against phospho-MAPK, total-MAPK, Akt, and CREB

95

were purchased from Cell Signaling Technology (Beverley, MA, USA). Antibodies for

96

p47phox (Ser345) were purchased from Assay Biotechnology Company, Inc. (Sunnyvale,

97

CA, USA).

98 99

Microglial cell culture and treatment

100

Immortalized murine BV2 microglial cells18 were cultivated and maintained at

101

37°C in Dulbecco’s modified Eagles medium supplemented with 10% heat-inactivated

102

fetal bovine serum, penicillin (10 U/ml), and streptomycin (10 µg/ml). BV2 cells were

103

grown to confluence and seeded into plates and incubated for 16-24 h. To examine the

104

anti-inflammatory effects of morin, BV2 cells were treated with morin (100-300 µM)

105

with or without LPS (0.1 µg/ml). MTT assay was performed to examine the effect of

106

morin on BV2 cell viability.19 The cell viability remained unaffected by morin

107

concentrations of up to 300 µM (data not shown).

108 109

Measurement of cytokines and nitrite levels

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

110

BV2 cells (1 × 105 cells per well on a 24-well plate) were pre-treated with morin

111

(100-300 µM) for 1 h and stimulated with LPS (0.1 µg/ml). The supernatants of the

112

cultured microglia were collected after 16 h of LPS stimulation and the concentrations

113

of TNF-α, IL-6, and IL-1β were measured using an enzyme-linked immunosorbent

114

assay (ELISA) kit (R&D Systems, Minneapolis, MN, USA) as per the manufacturer’s

115

instructions. Accumulated nitrite was also measured from the supernatants using the

116

Griess Reagent System (Promega, Madison, WI, USA).

117 118

Intracellular reactive oxygen species measurement

119

Intracellular accumulation of ROS was measured using 2',7'-dichlorofluorescein

120

(H2DCF-DA) and a modified version of a previously reported method.20 BV2

121

microglial cells (1 × 105 cells per well in a 24-well plate) were pretreated with morin

122

(100-300 µM) for 1 h, stimulated with LPS for 16 h, and stained with 20 µM H2DCF-

123

DA in sodium phosphate buffer (PBS) for 1 h at 37°C. DCF fluorescence intensity was

124

measured on a fluorescence plate reader (Molecular Devices, Sunnyvale, CA, USA) at

125

excitation and emission wavelengths of 485 nm and 535 nm, respectively. For the

126

image analysis of ROS production, BV2 cells were placed on coverslips and treated for

127

1 h with morin 1 h prior to LPS stimulation. Cells were stained with H2DCF-DA and

128

allowed to rest for 1 h, mounted on a clean, glass slide, and analyzed with the help of a

129

confocal laser scanning microscope.

130 131

Lipopolysaccharide-induced inflammation and morin administration

132

The study involved the use of ICR mice (aged 8-9 weeks) (Orient Co., Ltd., Seoul,

133

Korea). All animal experiments were approved by the Institutional Animal Care and

6

ACS Paragon Plus Environment

Page 6 of 36

Page 7 of 36

Journal of Agricultural and Food Chemistry

134

Use Committee at the Ewha Womans University School of Medicine, Seoul, Republic

135

of Korea. Efforts were made to minimize animal suffering, reduce the number of

136

animals used in the study, and utilize alternatives to in vivo techniques when possible.

137

Systemic

138

mg/kg, intraperitoneal (ip) injection) to male ICR mice as described in previous

139

studies.21 Morin (200 mg/kg) was dissolved in a vehicle solution (1% dimethyl

140

sulfoxide (DMSO) and 0.9% sodium chloride) and administered via an intraperitoneal

141

injection daily for 4 days before LPS stimulation. Samples were obtained 3 h after LPS

142

stimulation.

inflammation

was

induced

by

LPS

administration

(5

143 144

Immunohistochemistry

145

Mice were anesthetized with sodium pentobarbital (120 mg/kg ip) and perfused

146

transcardially with normal saline containing heparin (5 U/ml) after a 3-hour period

147

involving LPS stimulation, followed by the addition of 4% paraformaldehyde (PFA) in

148

0.1 M PBS (pH 7.2). The brains were removed, incubated overnight in fixatives, and

149

stored in a 30% sucrose solution. Serial coronal brain sections of the cortex and the

150

hippocampus (40 µm thickness, at 600 µm intervals) were collected through a freezing

151

sliding microtome (Leica Biosystems Nussloch GmBH, Nussloch, Germany).

152

Endogenous peroxidase was quenched with the help of 3% H2O2. Further, any

153

nonspecific binding was blocked by incubating the sections in PBS containing 4%

154

bovine serum albumin for 60 min at 37°C. Following an overnight incubation at 4°C

155

with either the primary antibody for IBA-1 at 1:1000 dilution (Wako, Osaka, Japan) or

156

CD11b at 1:500 dilution (Bio-Rad, Hercules, CA, USA), the sections were incubated

157

with a biotinylated secondary antibody for 1 h at 37°C and washed with 0.1% Triton

7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

158

X-100 in PBS. The sections were incubated with avidin-biotin-HRP complex reagent

159

(Vector Laboratories, Burlingame, CA, USA) for 1.5 h followed by washing with PBS,

160

followed by the peroxidase reaction using diaminobenzidine tetrahydrochloride (Vector

161

Laboratories, Burlingame, CA, USA). The treated sections were mounted with the help

162

of a mounting medium (Thermo Fisher Scientific, Pittsburgh, PA, USA) and examined

163

under a light microscope (Leica Biosystems Nussloch GmBH, Nussloch, Germany).

164

Quantification of IBA-1-positive cells was conducted with the help of the ImageJ

165

software (NIH, Bethesda, MD, USA). Three serial coronal brain sections were

166

obtained from each mice and 1 mm2 area was quantified for each brain section for the

167

lateral cortex, the hippocampus, and the dentate gyrus areas, respectively. Activated

168

and resting microglia were counted according to intensity and morphological

169

characteristics, as indicated in previous studies. 22,23

170 171

Traditional and real-time reverse transcription polymerase chain reaction

172

BV2 cells (7.5 × 105 cells per well on a 6-well plate) were stimulated with LPS in

173

with or without morin. For the isolation of total RNA from the brain cortex, brain

174

tissue was homogenized using a homogenizer (Thermo Fisher Scientific, Pittsburgh,

175

PA, USA) and total RNA was extracted using the TRI reagent (Thermo Fisher

176

Scientific, Pittsburgh, PA, USA). For reverse transcription polymerase chain reaction

177

(RT-PCR), total RNA (1 µg) was reverse-transcribed in a reaction mixture containing 1

178

U RNase inhibitor, 500 ng random primers, 3 mM magnesium chloride (MgCl2), 0.5

179

mM deoxyribonucleoside triphosphate (dNTP), 1X reverse transcription (RT) buffer,

180

and 10 U reverse transcriptase (Promega, Madison, WI). The synthesized cDNA was

181

used as a template for the polymerase chain reaction (PCR) reaction using GoTaq

8

ACS Paragon Plus Environment

Page 8 of 36

Page 9 of 36

Journal of Agricultural and Food Chemistry

182

polymerase (Promega, Madison, WI, USA) and primers for COX-2, iNOS, IL-1β, IL-6,

183

TNF-α, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), as described

184

previously.21 GAPDH was used as an internal control for normalizing the target gene

185

expression. Amplification of NADPH oxidase subunit genes was achieved through RT-

186

PCR. The synthesized cDNA was amplified with SYBR Green PCR Master Mix

187

(Applied Biosystems, Foster City, CA, USA), and the RT-PCR was performed on an

188

ABI Prism 7000 Sequence Detection System (Applied Biosystems, Foster City, CA,

189

USA). Expression levels of the target genes were normalized against that of GADPH

190

using the formula, 2(Ct test

191

shown in Table 1.

gene – Ct GAPDH)

. The primers used in the PCR reaction are

192 193

Electrophoretic mobility shift assay

194

BV2 cells (7.5 × 105 cells per well on a 6 cm dish) were pretreated for 1 h with

195

morin before LPS addition. Cells were harvested after LPS stimulation of the cells for

196

1 h. Nuclear extracts from the stimulated microglia were prepared by a method

197

described in a previous study.24 The double-stranded DNA oligonucleotides containing

198

NF-κB, AP-1, antioxidant response element (ARE), and cAMP response element (CRE)

199

consensus sequences (Promega, Madison, WI, USA) were end-labeled using T4

200

polynucleotide kinase (New England Biolabs, Beverly, MA, USA) in the presence of

201

[γ-32P] labeled ATP. Nuclear proteins (10 µg) were incubated with a 32P-labeled probe

202

on ice for 0.5 h and separated on a 5% acrylamide gel before visualization by

203

autoradiography.

204 205

Western blot analysis

9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

206

Whole cell protein lysates were prepared in lysis buffer (10 mM Tris, pH 7.4, 30

207

mM NaCl, 1% Triton, 0.1% SDS, 0.1% sodium deoxycholate, and 1 mM EDTA)

208

containing the protease inhibitor cocktail. The lysates were centrifuged at 13,200 rpm

209

for 15 min at 4oC, following which the supernatant was collected. The protein

210

concentration was determined using the Bradford protein assay. Protein samples

211

ranging from 80 to 200 µg in weight were separated by SDS-PAGE, transferred to

212

nitrocellulose membranes, and incubated with antibodies against the following markers:

213

(a) iNOS, TNF-α, IL-1β, IL-6, COX-2 (1:1000); (b) phospho-MAPK, total-MAPK,

214

Akt, and CREB (1:1000); and (c) HO-1 and Nrf2; or p-p47phox (1:1000). Following

215

thorough washing with Tris-buffered saline and Tween 20 (TBST) (Bio-Rad, Hercules,

216

CA, USA), horseradish peroxidase-conjugated secondary antibody (1:2000 dilution in

217

TBST) was applied. The blots developed using an enhanced chemiluminescence

218

detection kit (Pierce Biotechnology, Rockford, IL, USA).

219 220

Transient transfection and luciferase assay

221

BV2 cells (2 × 105 cells per well on a 12-well plate) were transfected with 1 µg of

222

plasmid DNA ([κB]3-luc, AP-1-luc, ARE-luc, and CRE-luc) using the Convoy

223

Platinum transfection reagent (ACTGene, Inc., Piscataway, NJ, USA). The effect of

224

morin on reporter gene activity was determined by pre-treating with morin prior to

225

stimulation with LPS (100 ng/ml) followed by incubation for 6 h prior to cell harvest.

226 227

Statistical analysis

228

Unless otherwise stated, all experiments were performed using triplicated samples

229

and the experiments were repeated at least thrice. The data are presented as mean and

10

ACS Paragon Plus Environment

Page 10 of 36

Page 11 of 36

Journal of Agricultural and Food Chemistry

230

the standard error of the mean (SEM) and statistical comparisons between groups were

231

performed using one-way analysis of variance (ANOVA), followed by the Newman-

232

Keuls multiple comparison test. A p value of less than 0.05 was considered significant.

233

11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

234

Results

235

Morin inhibits inducible nitric oxide synthase and pro-inflammatory molecules in

236

LPS-stimulated BV2 microglial cells

237

The anti-inflammatory effect of morin in microglia was investigated by treating

238

BV2 cells with morin for 1 h prior to LPS stimulation, and examining the effects of

239

morin on LPS-induced production of NO and cytokines. Pre-treated morin

240

significantly suppressed the production of the proinflammatory molecules, NO, TNF-α,

241

IL-6, and IL-1β (Figure 1A). Additionally, morin increased the production of the anti-

242

inflammatory molecule, IL-10. We further examined the effects of morin on mRNA

243

and protein expression of various pro- or anti-inflammatory molecules. RT-PCR

244

(Figure 1B and Figure 1C) and Western blot analyses (Figure 1D and Figure 1E)

245

showed that morin suppressed the expression of iNOS and the proinflammatory

246

molecules, TNF-α, IL-1β, IL-6, and COX-2, while it upregulated the expression of IL-

247

10.

248 249

Morin suppresses lipopolysaccharide-induced microglial activation and pro-

250

inflammatory cytokine expression in brain

251

The preventive and accumulative therapeutic effects of morin during

252

neuroinflammation were examined by injecting morin daily into mice for 4 days before

253

LPS stimulation. This was followed by determining the immunoreactivity of IBA1 (a

254

marker of microglia activation) in the cortex and hippocampus, 3 h after systemic

255

injection of LPS. The number of IBA1-positive cells with thick and densely stained

256

processes, an indicator of activated microglia, increased in the cortex and hippocampus

257

of LPS-stimulated mice as compared with that observed in control mice (Figure 2A).

12

ACS Paragon Plus Environment

Page 12 of 36

Page 13 of 36

Journal of Agricultural and Food Chemistry

258

However, pre-treatment with morin (200 mg/kg) significantly reduced the number of

259

IBA1-positive microglia in the LPS-inflamed areas of the brain. We quantified the

260

reduction of IBA1-positive activated microglia by morin in the cortex and the

261

hippocampus (Figure 2B). Staining with the microglial activation marker, CD11b, also

262

showed inhibition of microglial activation by morin in septic mice brains

263

(supplementary Figure 1). Furthermore, pre-treatment with morin significantly reduced

264

the LPS-induced expression of iNOS, TNF-α, IL-1β, IL-6, and COX-2 in the cortex

265

(Figure 2C and Figure 2D).

266 267

Morin inhibits NF-κB- and AP-1-mediated transcription and phosphorylation of

268

MAPKs and Akt

269

Further investigation of the anti-inflammatory mechanisms of morin involved

270

examining the effect of morin on NF-κB and AP-1, known to be key modulators of

271

cytokine and iNOS expression during an inflammatory response in the body.25 The

272

stimulation of BV2 cells with LPS showed a strong increase in the DNA binding

273

activity of NF-κB and AP-1 (Figure 3A and Figure 3B). While morin did not affect the

274

DNA binding activity of NF-κB, it inhibited the DNA binding activity of AP-1.

275

However, morin suppressed the transcriptional activity of NF-κB and AP-1 (Figure 3C

276

and Figure 3D). Furthermore, as shown in Figure 3E, morin inhibited the LPS-induced

277

phosphorylation of three types of MAPKs and Akt, which are important upstream

278

signaling molecules in inflammatory reactions mediated by activated microglia.

279 280 281

Morin increases the CREB signaling pathway activity The PKA/CREB signaling pathway in microglia contributes to the reduction of

13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

282

inflammation through anti-inflammatory gene expression regulation. In particular,

283

PKA activation and the subsequent CREB phosphorylation are upstream modulators of

284

HO-1 expression.26,27 Our group has previously reported the involvement of the

285

PKA/CREB signaling pathway in reducing inflammation.21,28 The inhibitory effect of

286

morin on NF-κB-mediated transcriptional activity, despite not affecting DNA binding

287

activity, might be due to the competitive binding of pCREB with CBP29, which

288

suppresses the interaction between the NF-κB subunit, p65, and the CREB-binding

289

protein (CBP). Therefore, we determined whether morin plays a role in CREB

290

phosphorylation. Our results confirmed that morin did increase the CREB

291

phosphorylation (Figure 4) and increased other CREB activities such as DNA binding,

292

transcriptional action, and nuclear translocation. Overall, the data suggests that the

293

anti-inflammatory or antioxidant mechanisms of morin involve modulation of CREB

294

signaling processes.

295 296

Morin suppresses reactive oxygen species production by NADPH oxidase complex

297

modulation and heme oxygenase-1 expression upregulation

298

We found that morin significantly suppressed LPS-induced ROS production in

299

BV2 cells (Figure 5A and Figure 5B). To further elucidate the mechanisms underlying

300

ROS inhibition by morin, we examined the expression of NADPH oxidase subunits

301

responsible for microglial ROS production.30 RT-PCR analysis revealed that morin

302

significantly inhibited the LPS-induced expression of p47phox and gp91phox but did not

303

affect the expression of p67phox and gp22phox (Figure 5C). Morin also inhibited the

304

phosphorylation of p47phox (Figure 5D), responsible for the activation of the NADPH

305

oxidase complex.

14

ACS Paragon Plus Environment

Page 14 of 36

Page 15 of 36

Journal of Agricultural and Food Chemistry

306

Heme oxygenase-1 plays the role of an anti-inflammatory and antioxidant

307

modulator in the microglia.31,32 Western blot and RT-PCR analyses showed that morin

308

upregulated HO-1 expression at the protein and mRNA levels (Figure 6A and Figure

309

6B). To investigate the underlying mechanism of HO-1 upregulation by morin, we

310

examined its effect on Nrf2, a key transcription factor in the expression of HO-1 and

311

other phase 2 antioxidant enzymes. Morin significantly increased Nrf2 binding to the

312

antioxidant response element (ARE) (Figure 6C). Moreover, morin increased nuclear

313

translocation of Nrf2 and ARE-driven luciferase activity (Figure 6D and Figure 6E).

314

These data suggest that the upregulation of HO-1 may be partly involved in the anti-

315

inflammatory and the antioxidant mechanisms of morin in activated microglia.

316

15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

317

Discussion

318

The current study demonstrated the anti-inflammatory effects of morin in vitro

319

and in vivo. Morin significantly suppressed the LPS-induced production of NO and

320

proinflammatory cytokines in BV2 cells and mice brains. Further mechanistic studies

321

showed that morin inhibited the LPS-induced phosphorylation of MAPKs or Akt, and

322

downregulated the NF-κB-mediated transcriptional activity via PKA signaling. Morin

323

also suppressed intracellular ROS levels by inhibiting the NADPH oxidase subunits

324

and by upregulating the Nrf2/HO-1 axis. Our results suggest a therapeutic potential for

325

morin in the treatment of neuroinflammatory disorders.

326

Previous studies by other research groups and us have found that the PKA/CREB

327

pathway contributes to the resolution of inflammation and ROS detoxification and that

328

PKA is an upstream modulator of HO-1 expression in the microglia28,33,34 In the

329

current study, morin increased the CREB phosphorylation, which is a downstream

330

regulatory target of PKA. Furthermore, morin increased the DNA binding and

331

transcriptional activities and the nuclear translocation of CREB. Several papers have

332

demonstrated that phosphorylated CREB competes for binding to CBP with NF-

333

κB.25,29 Based on this hypothesis, morin-mediated activation of CREB may augment

334

the inhibition of NF-κB-mediated transcriptional activity. Therefore, the anti-

335

inflammatory and antioxidant activity by morin seem to be a result of the enhancement

336

of the CREB signal.

337

Oxidative stress and neuroinflammation results from an overexpression of ROS

338

through modulation of the NADPH oxidase family of enzymes.30,35 Therefore, we

339

investigated whether morin inhibits ROS production by suppressing the subunits of

340

NADPH oxidase, including p47phox, p67phox, gp91phox, and p22phox. We observed that

16

ACS Paragon Plus Environment

Page 16 of 36

Page 17 of 36

Journal of Agricultural and Food Chemistry

341

morin suppressed mRNA expression of p47phox and gp91phox and inhibited the

342

phosphorylation of p47phox in LPS-stimulated microglia. Phosphorylation of p47phox

343

leads to the translocation of the p47phox-p67phox complex to the plasma membrane

344

where p47phox interacts with p22phox, and p67phox acts as the NADPH oxidase activator

345

through a direct protein-protein interaction.30 The data suggest that morin inhibits ROS

346

production by suppressing the expression and the phosphorylation of NADPH oxidase

347

subunits in microglia.

348

According to previous studies, morin is a known natural polyphenolic antioxidant.

349

In LPS-stimulated microglia, morin activates HO-1 induction to play an antioxidant

350

role,36,37 and this has been shown in an ischemia model as well.38 It is also known to

351

demonstrate neuroprotective effects in a PD model39 and contributes to the resolution

352

of inflammation in human chondrocytes40 and RAW 264.7 macrophages.41 In this study,

353

morin exerted its antioxidant activity in LPS-stimulated microglia by suppressing ROS

354

production and enhancing the HO-1 expression via Nrf2-ARE signaling. Heme

355

oxygenase-1 plays an antioxidant, anti-apoptotic, and anti-inflammatory role through

356

the modulation of the Nrf2-ARE signaling pathway.42-44 We observed that morin

357

increases the expression of HO-1 by enhancing the DNA binding activity of Nrf2 to the

358

ARE site. In addition, previous studies reported on the activation of HO-1 through

359

PKA signaling and the subsequent phosphorylation of CREB.28,45 Given this

360

perspective, morin exhibits its effects by suppressing proinflammatory and neurotoxic

361

molecules with a concomitant enhancement of anti-inflammatory and antioxidant

362

molecules such as PKA and HO-1.

363

Sepsis, which results from a severe inflammatory response to an infection, is a

364

leading cause of death and involves the dysfunction and failure of organ systems. A

17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

365

previous study has reported that pro-inflammatory cytokines and other mediators are

366

important in the pathogenesis of sepsis.46 Furthermore, sepsis has been associated with

367

microglial activation.47 This study used LPS-injected mice because peripheral injection

368

with LPS is a well-known sepsis model and systemic LPS induces microglial

369

activation and progressive neurodegeneration.16,17,48 Morin was found to inhibit the

370

microglial activation and inflammatory mediator expression in LPS-injected mice

371

brains.

372

In conclusion, our study provides evidence that morin has significant potential as

373

a protective agent for LPS-induced microglial activation. This is the first study to

374

report the pharmacological effects of morin in brain microglia and the first to analyze

375

its underlying molecular mechanisms in detail. Therefore, the anti-inflammatory and

376

the antioxidant effects of morin in brain microglia may be useful as a potential

377

therapeutic agent in the treatment of various neuroinflammatory disorders related to

378

inflammation and oxidative stress.

18

ACS Paragon Plus Environment

Page 18 of 36

Page 19 of 36

Journal of Agricultural and Food Chemistry

379

Abbreviations:

380

AP-1, activator protein-1; ARE, antioxidant response element; CBP, CREB binding

381

protein; CRE, cAMP response element; COX, cyclooxygenase; CREB, cAMP

382

response element binding protein; EMSA, electrophoretic mobility shift assay; HO-1,

383

heme oxygenase-1; IL, interleukin; iNOS, inducible nitric oxide synthase; LPS,

384

lipopolysaccharide; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor-

385

κB; Nrf, nuclear factor-E2-related factor; PI3K, phosphatidylinositol 3-kinase; PKA,

386

protein kinase A; ROS, reactive oxygen species; TNF, tumor necrosis factor

387 388

Funding sources

389

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

390

National Research Foundation of Korea (NRF) and funded by the Ministry of Science,

391

ICT & Future Planning (Grant No. NRF-2010-0027945 & NRF-2015R1A2A2A0100

392

5226).

19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

393

References

394 395

(1) Ulland, T. K.; Wang, Y.; Colonna, M. Regulation of microglial survival and

396

proliferation in health and diseases. Semin. Immunol. 2015, 27, 410-415.

397 398

(2) Perdiguero, E. G.; Geissmann, F. The development and maintenance of resident

399

macrophages. Nat. Immunol. 2016, 17, 2-8.

400 401 402

(3) Bordt, E. A.; Polster, B. M. NADPH oxidase- and mitochondria-derived reactive oxygen species in proinflammatory microglial activation: a bipartisan affair? Free

403

Radic. Biol. Med. 2014, 76, 34-46.

404 405

407

(4) Sato, A.; Ohtaki, H.; Tsumuraya, T.; Song, D.; Ohara, K.; Asano, M.; Iwakura, Y.; Atsumi, T.; Shioda, S. Interleukin-1 participates in the classical and alternative activation of microglia/macrophages after spinal cord injury. J. Neuroinflammation

408

2012, 9, 65.

406

409 410

(5) Tang, Y.; Le, W. Differential roles of M1 and M2 microglia in neurodegenerative

411

diseases. Mol. Neurobiol. 2016, 53(2), 1181-1194.

412 413 414

(6) Rojo, A. I.; McBean, G.; Cindric, M.; Egea, J.; Lopez, M.; Rada, P.; Zarkovic, N.; Cuadrado, A. Redox control of microglial function: Molecular mechanisms and

415

functional significance. Antioxid. Redox. Signal. 2014, 21(12), 1766-1801.

416 417

(7) Jin, X.;Yamashita, T. Microglia in central nervous system repair after injury. J.

418

Biochem. 2016, 159, 491-496.

419 420

(8) Sinha, K.; Sadhukhan, P.; Saha, S.; Pal, P. B.; Sil, P. C. Morin protects gastric

421 422

mucosa from nonsteroidal anti-inflammatory drug, indomethacin induced inflammatory damage and apoptosis by modulating NF-κB pathway. Biochim. Biophys.

423

Acta. 2015, 1850, 769-783.

424 425

(9) Tianzhu, Z.; Shihai, Y.; Juan, D. The effects of morin on lipopolysaccharide-

426

induced acute lung injury by suppressing the lung NLRP3 inflammasome.

427

Inflammation 2014, 37, 1976-1983.

428

20

ACS Paragon Plus Environment

Page 20 of 36

Page 21 of 36

Journal of Agricultural and Food Chemistry

429 430 431

(10) Wang, J.; Guo, C.; Wei, Z.; He, X.; Kou, J.; Zhou, E.; Yang, Z.; Fu, Y. Morin suppresses inflammatory cytokine expression by downregulation of nuclear factor-κB

432

and mitogen-activated protein kinase (MAPK) signaling pathways in lipopolysaccharide-stimulated primary bovine mammary epithelial cells. J. Dairy Sci.

433

2016, 99, 3016-3022.

434 435

(11) Wei, Z.; He, X.; Kou, J.; Wang, J.; Chen, L.; Yao, M.; Zhou, E.; Fu, Y.; Guo, C.;

436

Yang, Z. Renoprotective mechanisms of morin in cisplatin-induced kidney injury. Int.

437

Immunopharmacol. 2015, 28, 500-506.

438 439

(12) Singh, M. P.; Jakhar, R.; Kang, S. C. Morin hydrate attenuates the acrylamide-

440

induced imbalance in antioxidant enzymes in a murine model. Int. J. Mol. Med. 2015, 36, 992-1000.

441 442 443 444

(13) Du, Y.; Qu, J.; Zhang, W.; Bai, M.; Zhou, Q.; Zhang, Z.; Li, Z.; Miao, J. Morin reverses neuropathological and cognitive impairments in APPswe/PS1dE9 mice by

445

targeting multiple pathogenic mechanisms. Neuropharmacology 2016, 108, 1-13.

446 447

(14) Gong, E. J.; Park, H. R.; Kim, M. E.; Piao, S.; Lee, EJ.; Jo, D. G.; Chung, H. Y.;

448

Ha, N. C.; Mattson, M. P.; Lee, J. W. Morin attenuates tau hyperphosphorylation by

449

inhibiting GSK3β. Neurobiol. Dis. 2011, 44(2), 223-230.

450 451 452

(15) Jakhar, R.; Paul, S.; Chauhan, A. K.; Kang, S. C. Morin hydrate augments phagocytosis mechanism and inhibits LPS induced autophagic signaling in murine

453

macrophage. Int. Immunopharmacol. 2014, 22, 356-365.

454 455

(16) Perry, V.H.; Newman, T.A.; Cunningham, C. The impact of systemic infection on

456

the progress of neurodegenerative disease. Nat Rev Neurosci 2003, 4, 103-112.

457 458

(17) Cunningham, C.

459

inflammation. Glia 2013, 61, 71-90.

Microglia and neurodegeneration: the role of systemic

460 461

(18) Bocchini, V.; Mazzolla, R.; Barluzzi, R.; Blasi, E.; Sick, P.; Kettenmann, H. An

462

immortalized cell line expresses properties of activated microglial cells. J. Neurosci.

463

Res. 1992, 31, 616-621.

464

21

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

465

(19) Shim, A. R.; Dong, G. Z.; Lee, H. J.; Ryu, J. H. Aractylchromene is a repressor of

466

wnt/β-catenin signaling in colon cancer cells. Biomol. Ther. 2015, 23, 26-30.

467 468

(20) Kim, B.; Kim, J. E.; Choi, B. K.; Kim, H. S. Anti-inflammatory effects of water

469

chestnut extract on cytokine responses via nuclear factor-κB-signaling pathway. Biomol. Ther. 2015, 23, 90-97.

470 471 472 473 474 475

(21) Jung, J. S.; Shin, K. O.; Lee, Y. M.; Shin, J. A.; Park, E. M.; Jeong, J.; Kim, D. H.; Choi, J. W.; Kim, H. S. Anti-inflammatory mechanism of exogenous C2 ceramide in lipopolysaccharide-stimulated microglia. Biochim. Biophys. Acta. 2013, 1831, 10161026.

476 477 478

(22) Soltys, Z.; Ziaja, M.; Pawlinski, R.; Setkowicz, Z.; Janeczko, K. Morphology of reactive microglia in the imjured cerebral cortex. Fractal analysis and complementary

479

quantitative methods. J. Neurosci. Res. 2001, 63, 90-97.

480 481

(23) Kreutzberg, G.W.; Microglia: a sensor for pathological events in the CNS. Trends

482

Neurosci. 1996, 19, 312-318.

483 484 485

(24) Lee, E. J.; Ko, H. M.; Jeong, Y.H.; Park, E. M.; Kim, H. S. β-Lapachone suppresses neuroinflammation by modulating the expression of cytokines and matrix

486

metalloproteinases in activated microglia. J. Neuroinflammation 2015, 12, 133.

487 488

(25) Smale, S. T. Selective transcription in response to an inflammatory stimulus. Cell

489

2010, 140, 833-844.

490 491 492

(26) Ziady, A. G.; Sokolow, A.; Shank, S.; Corey, D.; Myers, R.; Plafker, S.; Kelley, T. J. Interaction with CREB binding protein modulates the activities of Nrf2 and NF-

493

kappaB in cystic fibrosis airway epithelial cells. Am. J. Physiol. Lung cell Mol. Physiol.

494

2012, 302, L1221-L1231.

495 496 497

(27) Smith, K. T.; Nicholls, R. D.; Rdines, D. The gene encoding the gragile X RNAbinding protein is controlled by nuclear respiratory factor 2 and the CREB family of

498

transctiption factors. Nucleic Acids Res. 2006, 34, 1205-1215.

499 500

(28) Jung, J. S.; Shin, J. A.; Park, E. M.; Lee, J. E.; Kang, Y. S.; Min, S. W.; Kim, D. H.;

22

ACS Paragon Plus Environment

Page 22 of 36

Page 23 of 36

Journal of Agricultural and Food Chemistry

501 502

Hyun, J. W.; Shin, C. Y.; Kim, H. S. Anti-inflammatory mechanism of ginsenoside Rh1 in lipopolysaccharide-stimulated microglia: critical role of the protein kinase A

503

pathway and hemeoxygenase-1 expression. J. Neurochem. 2010, 115, 1668-1680.

504 505

(29) Delgado, M. Vasoactive intestinal peptide and pituitary adenylate cyclase-

506

activating polypeptide inhibit CBP-NF-kappaB interaction in activated microglia.

507

Biochem. Biophys. Res. Commun. 2002, 297, 1181-1185.

508 509

(30) Bedard, K.; Krause, K. H. The NOX family of ROS-generating NADPH oxidases:

510

physiology and pathophysiology. Physiol. Rev. 2007, 87, 245-313.

511 512

(31) Cuadrado, A.; Rojo, A. I. Heme oxygenase-1 as a therapeutic target in

513

neurodegenerative diseases and brain infections. Curr. Pharm. Des. 2008, 14, 429-442.

514 515

(32) Keum, Y. S. Regulation of Nrf2-mediated phase II detoxification and anti-oxidant

516

genes. Biomol. Ther. 2012, 20, 144-151.

517 518

(33) Kast, R. E. Tumor necrosis factor has positive and negative self regulatory feed

519

back cycles centered around cAMP. Int. J. Immunopharmacol. 2000, 22, 1001-1006.

520 521 522

(34) Lee, B.; Cao, R.; Choi, Y. S.; Cho, H. Y.; Rhee, A. D.; Hah, C. K.; Hoyt, K. R.; Obrietan, K. The CREB/CRE transcriptional pathway: protection against oxidative

523

stress-mediated neuronal cell death. J. Neurochem. 2009, 108, 1251-1265.

524 525

(35) Gao, H. M.; Zhou, H.; Hong, J. S. NADPH oxidases: novel therapeutic targets for

526

neurodegenerative diseases. Trends pharmacol. Sci. 2012, 33, 295-303.

527 528

(36) Dilshara, M. G.; Jayasooriya, R. G.; Lee, S. H.; Choi, Y. H.; Kim, G. Y. Morin

529

downregulates nitric oxide and prostaglandin E2 production in LPS-stimulated BV2

530

microglial cells by suppressing NF-κB activity and activating HO-1 induction. Environ. Toxicol. Pharmacol. 2016, 44, 62-68.

531 532 533

(37) Kim, J. M.; Lee, E. K.; Park, G.; Kim M. K.; Yokozawa, T.; Yu, B. P. Morin

534

modulates the oxidative stress-induced NF-kappaB pathway through its anti-oxidant

535

activity. Free Radic. Res. 2010, 44(4), 454–461.

536

23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

537 538

(38) Gottlieb, M.; Leal-Campanario R.; Campos-Esparza M. R.; Sanchez-Gomez, M. V.; Alberdi, E.; Arranz, A.; Delgado-Garcia, J. M.; Gruart, A.; Matute, C.

539

Neuroprotection by two polyphenols following excitotoxicity and experimental

540

ischemia. Neurobiol. Dis. 2006, 23(2), 374-386.

541 542 543

(39) Zhang, Z. T.; Cao, X. B.; Xiong, N.; Wang, H. C.; Huang, J. S.; Sun, S. G.; Wang, T. Morin exerts neuroprotective actions in Parkinson disease models in vitro and in

544

vivo. Acta Pharmacol. Sin. 2010, 31, 900–906

545 546 547

(40) Chen, W. P.; Wang, Y. L.; Tang, J. L.; HU, P. F.; Bao, J. P.; Wu, L. D. (2012) Morin inhibits interleukin-1β-induced nitric oxide and prostaglandin E2 production in

548

human chondrocytes. Int Immunopharmacol. 2012, 12(2), 447–452.

549 550

552

(41) Dhanasekar, C.; Kalaiselvan, S.; Rasool, M. Morin, a Bioflavonoid suppresses monosodium urate crystal-induced inflammatory immune response in RAW 264.7 macrophages through the inhibition of inflammatory mediators, intracellular ROS levels

553

and NF-κB activation. PLOS one, 2015, 10(12), e0145093.

551

554 555

(42) Jamal, U. M.; Joe, Y.; Kim, S. K.; Oh, J. S.; Ryter, S. W.; Pae, H. O.; Chung, H. T.

556

IRG1 induced by heme oxygenase-1/carbon monoxide inhibits LPS-mediated sepsis

557

and pro-inflammatory cytokine production. Cell Mol. Immunol. 2016, 13, 170-179.

558 559 560

(43) Naidu, S.; Vijayan, V.; Santoso, S.; Kietzmann, T.; Immenschuh, S. Inhibition and genetic deficiency of p38 MAPK up-regulates heme oxygenase-1 gene expression via

561

Nrf2. J. Immunol. 2009, 182, 7048-7057.

562 563 564

(44) Chen, X. L.; Kunsch, C. Induction of cytoprotective genes through Nrf2 /antioxidant response element pathway: a new therapeutic approach for the treatment

565

of inflammatory diseases. Curr. Pharm. Des. 2004, 10, 879-891.

566 567

569

(45) Park, S. Y.; Bae, Y. S.; Ko, M. J.; Lee, S. J.; Choi, Y. W. Comparison of antiinflammatory potential of four different dibenzocyclo-octadiene lignans in microglia; action via activation of PKA and Nrf-2 signaling and inhibition of MAPK/STAT/NF-

570

κB pathways. Mol. Nutr. Food. Res. 2014, 58, 738–748.

568

571 572

(46) Netea, M. G.; van der Meer, J. W.; van Deuren, M.; Kullberg, B. J.

24

ACS Paragon Plus Environment

Page 24 of 36

Page 25 of 36

Journal of Agricultural and Food Chemistry

573

Proinflammatory cytokines and sepsis syndrome: not enough, or too much of a good

574

thing? Trends Immunol. 2003, 24, 254–258.

575 576 577

(47) Lemstra, A. W.; Woud, J. C.; Hoozemans, J. J.; van Haastert, E. S.; Rozemuller, A. J.; Eikelenboom, P. Microglia activation in sepsis: a case–control study. J.

578

Neuroinflammation 2007, 4, 4.

579 580 581

(48) Hoogland, I. C. M.; Houbolt, C.; van Westerloo, D. J.; van Gool, W. A.; van de Beek, D. Systemic inflammation and microglial activation: systematic review of

582

animal experiements. J. Neuroinflmmation 2015, 12, 114.

25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

583

Figure Legends

584

Figure 1. Inhibition of the expression of iNOS and proinflammatory molecules by

585

morin in LPS-stimulated microglia. (A) BV2 cells are pre-treated with morin for 1 h

586

followed by LPS stimulation (0.1 µg/ml) for 16 h. The supernatants are obtained and

587

the amount of NO, TNF-α, IL-1β, IL-6, and IL-10 released into the media is

588

determined. (B, D) BV2 cells are pretreated with morin for 1 h followed by LPS

589

stimulation for 6 h. RT-PCR (B) and Western blot (D) are performed to see the effects

590

of morin on mRNA and protein expression of various proinflammatory molecules. (C,

591

E) Quantification analysis of 3 independent experiments by RT-PCR (C) and Western

592

blot (E) are shown. Data are expressed as the means (± SEM). *p < 0.05, significantly

593

different from LPS-stimulated samples.

594 595

Figure 2. Reduction of inflammatory response in the brains of LPS-stimulated mice by

596

morin. (A) Morin (200 mg/kg) pre-treatment inhibits microglial activation in each

597

brain region 3 h after systemic LPS injection (5 mg/kg, ip). The images represent

598

IBA1-positive microglia in the cortex and the hippocampus. (B) Quantified data of

599

IBA1-positive microglia in the cortex and the hippocampus (Scale, 50 µm; n=6 per

600

group). (C) RT-PCR for mRNA expression levels of proinflammatory molecules in the

601

cortex. Representative gels are shown. (D) Quantified data (n = 3 per group). #p < 0.05,

602

significantly different from the control group; *p < 0.05, significantly different from

603

the LPS-stimulated group.

604 605

Figure 3. Effect of morin on the activity of NF-κB, AP-1 and the phosphorylation of

606

MAPKs and Akt in LPS-stimulated BV2 cells. EMSA for NF-κB (A) and AP-1 (B)

26

ACS Paragon Plus Environment

Page 26 of 36

Page 27 of 36

Journal of Agricultural and Food Chemistry

607

was performed using nuclear extracts isolated from the BV2 cells treated with morin in

608

the presence of LPS for 1 h. Transient transfection analysis of (C) (κB)3-luc and (D)

609

AP-1-luc reporter gene activity. (E) Cell extracts were prepared from BV2 cells

610

stimulated with LPS for 0.5 h with or without morin, followed by Western blotting

611

using antibodies against phospho-MAPK, total-MAPK, or Akt. (F) Quantified data.

612

Levels of the phosphorylated forms of MAPKs and Akt were normalized to the level of

613

each total form and expressed as relative fold changes versus the control group. Data

614

are expressed as the means ± SEM for three (A-D) or 4 (F) independent experiments.

615

*p < 0.05, significantly different from the LPS-stimulated samples.

616 617

Figure 4. Morin enhancement of the PKA-CREB signaling pathway. (A) Cell extracts

618

are prepared from BV2 cells stimulated with LPS for 1 h with or without morin and

619

subjected to Western blot analysis using antibodies against phospho- or total- CREB.

620

Levels of the phosphorylated CREB are normalized to the level of each total form.

621

Quantified data are shown in the bottom panel. (B) EMSA for CREB binding activity.

622

The data are representative of 3 independent experiments. (C) Western blot is

623

performed to detect the nuclear translocation of CREB. (D) Effect of morin on CRE-

624

luc reporter gene activity. The data are expressed as the mean ± SEM for 3 independent

625

experiments. #p < 0.05, significantly different from the control samples; *p < 0.05,

626

significantly different from the LPS-stimulated samples.

627 628

Figure 5. Morin reduction of ROS production via suppression of NADPH oxidase

629

subunits. (A) BV2 cells are pre-treated with morin 1 h prior to LPS stimulation.

630

Intracellular ROS levels are measured using the DCF-DA method after 16 h. Data are

27

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

631

expressed as the means ± SEM of 3 independent experiments. (B) DCF-derived

632

fluorescence in BV2 cells. (C) RT-PCR for mRNA expression levels of NADPH

633

oxidase subunits (p47phox, gp91phox, gp22phox, p67phox) in BV2 cells (n = 3). (D) Cell

634

extracts are first prepared from BV2 cells stimulated with LPS for 0.5 h in the absence

635

or presence of morin, and subsequently subjected to Western blot analysis using

636

antibodies against phospho-p47phox. Quantified data are shown in the bottom panel (n =

637

3). Levels of phosphorylated p47phox are normalized with β-actin. *p < 0.05,

638

significantly different from the LPS-stimulated group.

639 640

Figure 6. Enhancement of HO-1 expression by morin via Nrf2/ARE signaling

641

upregulation (A, B) BV2 cells are pretreated with morin for 1 h and stimulated with

642

LPS for 6 h. Western blot (A) and RT-PCR (B) for protein and mRNA expression

643

levels of HO-1 post morin treatment. (C) EMSA for Nrf2 binding activity. (D) Western

644

blot analysis to detect the nuclear translocation of Nrf2. Quantified data are shown in

645

the bottom panel (n = 3). (E) The effect of morin on ARE-luc reporter gene activity. #p

646

< 0.05, significantly different from the control samples; *p < 0.05, significantly

647

different from the LPS-stimulated samples.

28

ACS Paragon Plus Environment

Page 28 of 36

Page 29 of 36

Journal of Agricultural and Food Chemistry

Table 1. Primers used in RT-PCR Gene

Forward Primer (5’→3’)

Reverse Primer (5’→3’)

Size

p47phox

TTCACCACCATGGAGAAGGC

GGCATGGACTGTGGTCATGA

212 bp

p67phox

CTGGCTGAGGCCATCAGACT

AGGCCACTGCAGAGTGCTTG

214 bp

gp91phox

TTGGGTCAGCACTGGCTCTG

TGGCGGTGTGCAGTGCTATC

185 bp

p22phox

GTCCACCATGGAGCGATGTG

CAATGGCCAAGCAGACGGTC

164 bp

GAPDH

TTCACCACCATGGAGAAGGC

GGCATGGACTGTGGTCATGA

236 bp

29

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 30 of 36

A

*

20 *

*

0 100 200 300 (μM) Morin: 0

80

*

600

*

400

*

200

0 0 100 200 300 (μM) Morin: 0

B 0

20

*

* *

10

0 100 200 300 (μM) Morin: 0 + LPS

+ LPS 300

30

0

+ LPS

+ LPS

Morin: 0

*

200 0

0

* *

40 20

0 0 100 200 300 (μM) Morin: 0

0 100 200 300 (μM)

+ LPS

+ LPS

100 200 300 (μM) 8

iNOS

iNOS

TNF-α

COX-2

COX-2 IL-6

Fold induction

6

TNF-α

IL-1β

*

*

4 2

*

* *

* *

0 8

IL-6

IL-1β

IL-10

6 4

*

*

*

*

*

* *

* *

2 0

IL-10

Morin: 0 300 0 100 200 300 + LPS

GAPDH

D Morin: 0

+ LPS 300

0

100 200

0 300 0 100 200 300 + LPS

0 300 0 100 200 300 (μM) + LPS

E 300 (μM) 8

iNOS TNF-α COX-2 IL-6 IL-1β

iNOS

TNF-α

6 4

COX-2

* *

2 0 8

*

* * *

IL-6

IL-1β

*

* * IL-10

* *

6 4 2

*

*

* * *

* *

0

IL-10

Morin: 0 300 0 100 200 300 + LPS

β-actin

Fig. 1 ACS Paragon Plus Environment

0 300 0 100 200 300 + LPS

*

60

C

Fold induction

Morin: 0

*

100

40

IL-10 (pg/ml)

*

400

50

800

IL-1β (pg/ml)

40

1000 IL-6 (pg/ml)

600 TNF-α (pg/ml)

NO (μM)

60

0 300 0 100 200 300 (μM) + LPS

Page 31 of 36

Journal of Agricultural and Food Chemistry

LPS (5 mg/kg)

A

B

cortex

morin

hippocampus

cortex

hippocampus

Iba1+ activated microglia (cells/mm2)

vehicle

Iba1+ activated microglia (cells/mm2)

saline

80

#

60

*

40 20 0

sal

LPS 60

# 40

* 20 0

sal

D

LPS (5 mg/kg) vehicle

morin

#

iNOS

4

* 2

iNOS TNF-α COX-2 IL-6 IL-1β

Fold induction

saline

6

0 6 #

TNF-α

#

IL-6

*

* 2

2 0 6

#

0 COX-2 6

*

4 2

GAPDH

6 4

4

Veh morin LPS

ACS Paragon Plus Environment

IL-1β

*

4

0 Sal

Fig. 2

#

2

0

veh morin LPS

Scale bar : 50 μm

C

veh morin

Sal

Veh morin LPS

Journal of Agricultural and Food Chemistry

A

B

+ LPS

Morin: 0

300

0

100

+ LPS

Morin:

200 300 (μM)

Page 32 of 36

300

0

0

100 200 300 (μM)

NF-κB

AP-1

C

D

► Reporter plasmid: (κB)3-luc

► Reporter plasmid: AP1-luc 4

6

*

4

Fold induction

Fold induction

8

* *

2

*

2

*

1 0

0

Morin:

*

3

0

0

100 200 300 (μM)

Morin:

0

0

+ LPS

100 200 300 (μM) p-ERK ERK p-JNK JNK p-p38

p-JNK/JNK

0

p-p38/p38

300

p-ERK/ERK

F

+ LPS

6 4

*

*

*

*

*

*

*

*

*

*

2 0 6 4 2 0 6 4

*

2 0 6

p38 p-Akt

p-Akt/Akt

Morin: 0

+ LPS

Fold induction

E

100 200 300 (μM)

4 2

*

0 Morin: 0 300 0 100 200 300 (μM)

Akt

+ LPS

Fig. 3

ACS Paragon Plus Environment

Page 33 of 36

Journal of Agricultural and Food Chemistry

A

B

+ LPS

Morin: 0

300

Morin:

100 200 300 (μM)

0

+ LPS 0

300 0 100 200 300 (μM)

pCREB CRE+ protein complex

Fold induction

CREB 8 6

#

4

*

*

*

#

2 0

Morin:

0

300

0

100 200 300 (μM)

◀F

Fold Induction:

+ LPS

C

+ LPS

Morin: 0

300

0

D

100 200 300 (μM)

► Reporter plasmid: CRE-luc

CREB

8

*

6 # 4

#

*

*

Fold induction

Fold induction

Histon H1

4 3 2

#

#

300

0

*

0

100 200 300 (μM)

0

Morin: 0

300

0

*

1

0 Morin:

2

*

+ LPS

100 200 300 (μM) + LPS

Fig. 4

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

B

A

ROS

(Fold induction)

4

con

LPS

Morin 300

LPS + Morin

3

*

2

* 1

*

0 Morin:

0

100 200 300 (μM)

0

+ LPS

*

Fold induction p22phox mRNA

2

*

1

0 4

*

3

Fold induction p67phox mRNA

5

*

2 1

0 Morin:

2

1

0

1.5 1.0 0.5

0 Morin:

0 300 0 100 200 300 (μM)

0 300 0 100 200 300 (μM)

+ LPS

+ LPS

D

+ LPS

Morin: 0

300

0

100 200 300 (μM) p-p47phox β-actin

Fold induction

Fold induction gp91phox mRNA

Fold induction P47phox mRNA

C

4 3

*

2

*

1 0

Morin:

0

300

0

100

*

200 300 (μM)

+ LPS

Fig. 5

ACS Paragon Plus Environment

Page 34 of 36

Journal of Agricultural and Food Chemistry

A Morin: 0

B

+ LPS 300

0

20

100 200 300 (μM) HO-1

Fold induction

β-actin 8 6 4

#

*

*

Fold induction HO-1 mRNA

Page 35 of 36

*

15

* 10

*

5

0 Morin:

*

0

#

#

300

0

#

100 200 300 (μM) + LPS

2

0 Morin: 0

300

0

D

100 200 300 (μM)

+ LPS

Morin: 0

+ LPS

300

0

100 200 300 (μM) Nrf2

C

+ LPS Histon H1 300 0

100 200 300 (μM)

ARE + protein complex

Fold induction

Morin: 0

8 6 4

*

#

#

300

0

*

*

2

0 Morin: 0

100 200 300 (μM) + LPS

E

► Reporter plasmid: ARE-luc 4

Fold induction

Fold Induction:

◀F

*

3

# #

2

*

*

1

0 Morin:

0

300

0

100 200 300 (μM) + LPS

Fig. 6

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

TOC Graphic

LPS

Morin

Morin PI3K/Akt, MAPKs

NF-κ κB, AP-1

iNOS, Cytokines

PKA

CREB, Nrf2

HO-1

Neuroinflammation

ACS Paragon Plus Environment

: activate : inhibit

Page 36 of 36