Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7 macrophages by mediating NF-κB and Nrf2 pathways

Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7 macrophages by mediating NF-κB and Nrf2 pathways

Journal Pre-proof Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7 macrophages by mediating NF-κB and Nrf2 pathways Ke-Gang Linghu, Qi...

2MB Sizes 0 Downloads 4 Views

Journal Pre-proof Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7 macrophages by mediating NF-κB and Nrf2 pathways Ke-Gang Linghu, Qiu Shuo Ma, Guan Ding Zhao, Wei Xiong, Ligen Lin, Qing-Wen Zhang, Zhaoxiang Bian, Yitao Wang, Hua Yu PII:

S0014-2999(19)30806-4

DOI:

https://doi.org/10.1016/j.ejphar.2019.172854

Reference:

EJP 172854

To appear in:

European Journal of Pharmacology

Received Date: 19 May 2019 Revised Date:

4 December 2019

Accepted Date: 9 December 2019

Please cite this article as: Linghu, K.-G., Ma, Q.S., Zhao, G.D., Xiong, W., Lin, L., Zhang, Q.-W., Bian, Z., Wang, Y., Yu, H., Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7 macrophages by mediating NF-κB and Nrf2 pathways, European Journal of Pharmacology (2020), doi: https://doi.org/10.1016/j.ejphar.2019.172854. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier B.V.

1

Leocarpinolide B attenuates LPS-induced inflammation on RAW264.7

2

macrophages by mediating NF-κB and Nrf2 pathways

3 4

Ke-Gang Linghua, Qiu Shuo Maa, Guan Ding Zhaoa, Wei Xionga, Ligen Lina,

5

Qing-Wen Zhanga, Zhaoxiang Bianc, Yitao Wanga, Hua Yua,b,c*

6 7

a

8

Chinese Medicine, University of Macau 519000, Macao, China

9

b

HKBU Shenzhen Research Center, Shenzhen 518000, Guangdong, China

10

c

School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong 999077,

11

Hong Kong, China

Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in

12 13

*

14

Hua YU

15

Room 8008, Building N22,

16

Institute of Chinese Medical Sciences, University of Macau,

17

Avenida da Universidade, Taipa, Macao SAR, China

18

Tel: +853-8822 8540

19

E-mail: [email protected]

Correspondence to:

20 21 22

1

23 24

ABSTRACT Macrophages-mediated inflammation is involved in the regulation of rheumatoid

25

arthritis (RA). Sigesbeckiae Herba (SH) has been traditionally used for rheumatism.

26

However, the bioactive ingredients of SH are still unclear. Recently, we isolated a

27

compound (Leocarpinolide B, LB) from SH and identified its potent

28

anti-inflammatory and antioxidant effects on RAW264.7 macrophages for the first

29

time. LB effectively inhibited excessive production of nitric oxide (NO),

30

prostaglandin E2 (PGE2), cytokines (IL-6, TNF-α and MCP-1), and the expression of

31

cyclooxygenase-2 (COX-2) and inducible nitric oxide synthases (iNOS) in

32

lipopolysaccharide (LPS)-induced RAW264.7 cells. LB blocked the degradation of

33

inhibitor of kappa B ( IκBα) and translocation of nuclear factor kappa B (NF-κB) p65.

34

Additionally, LB reduced the intracellular reactive oxygen species, and increased the

35

expression of heme oxygenase-1 (HO-1) and the translocation of nuclear factor

36

erythroid 2-related factor 2 (Nrf2) in the presence or absence of LPS. The results

37

suggested that LB might be one of the bioactive components of SH, and be potential

38

for the treatment of RA and valuable to be further investigated.

39

Keywords: Rheumatoid arthritis (RA); Sigesbeckiae Herba (SH); Leocarpinolide B;

40

Nuclear factor erythroid 2-related factor 2 (Nrf2) ; Nuclear transcription factor-kappa

41

B (NF-κB).

42 43

2

44 45

1. Introduction Rheumatoid arthritis (RA) is a kind of autoimmune disease mediated by multiple

46

immune cells and their respective inflammatory mediators (Smolen et al., 2016).

47

Macrophage is one of the most critical cells involved in the initiation and propagation

48

of RA, activated macrophages could secret large amount of inflammatory mediators

49

to drive persistent inflammation and joint destruction (Scott DL, Wolfe F, 2010).

50

Therefore, a proper approach to reduce or reverse the excessive inflammatory

51

mediators to maintain the normal physiological function of macrophage is especially

52

significant.

53

RAW264.7 cell is a kind of macrophage widely used for the research of

54

inflammation (Li et al., 2018a; Ralph and Nakoinz, 1977). There are several signaling

55

pathways closely related to the inflammation in RAW264.7 macrophages. Nuclear

56

factor-kappa B (NF-κB) is a typical inflammation-related signaling pathway that

57

regulates the expression of pro-inflammatory cytokines (Cao et al., 2019). Pathogenic

58

microorganisms and pro-inflammatory mediators (such as interleukin (IL)-6, IL1β,

59

tumor necrosis factor alpha) can trigger the NF-κB pathway (Lee et al., 2017). NF-κB

60

is activated by phosphorylation of IκB via activating IKK in response to

61

pro-inflammatory stimuli (Li et al., 2018b). Inhibition of the activation of NF-κB has

62

been confirmed to ameliorate inflammation (Cao et al., 2019; Li et al., 2017).

63

It is well believed that inflammation is linked with oxidative stress. Oxidative

64

stress refers to elevated intracellular level of reactive oxygen species that is

65

considered as the most potent inflammatory mediator (Shin et al., 2008). Antioxidants

66

play a critical role in reducing inflammation (Arulselvan et al., 2016). The activation

67

of nuclear factor erythroid 2-related factor 2 (Nrf2) pathway could block the

68

progression of inflammation (Ahmed et al., 2017). Nrf2-mediated antioxidant gene

69

expression could reduce the macrophage M1 phenotype and reactive oxygen species

70

production (Jaiswal, 2004; Kobayashi et al., 2016). Nrf2 regulates the expression of

71

second-stage detoxification enzymes including glutathione peroxidase, heme

72

oxygenase-1 (HO-1) gene and antioxidants, which protect cells from various damages

73

through their anti-inflammatory effects, thereby affecting the progression of the 3

74

disease (Lee et al., 2017; Li et al., 2019).

75

Sigesbeckiae Herba (SH) is a traditional anti-inflammatory herbal medicine,

76

which has been used for rheumatism since Tang dynasty in China (Zhang et al., 2019).

77

We have focused on the modern pharmacological research of SH in the past years, and

78

identified its anti-inflammatory effects and anti-arthritis properties (Chu et al., 2018;

79

Sang et al., 2018; Zhang et al., 2019; Zhong et al., 2019). However, the bioactive

80

ingredients of SH are still unclear. Recently, we isolated several compounds from SH

81

and identified that Leocarpinolide B (LB, C22H26O8) exhibited potent

82

anti-inflammatory effects on LPS-induced RAW264.7 cells, which suggested the

83

potential contribution of LB for HS on RA treatment. LB is a sesquiterpene lactones

84

firstly isolated and reported in 1992 by Macías & H. Fischer from Lecocarpus

85

pinnatifidus (Macías and H. Fischer, 1992). Cartagena E et al reported that LB from

86

Acanthospermum hispidum exerted antibacterial action by inhibiting the production of

87

biofilm (Cartagena et al., 2007). In this study, we isolated LB from Sigesbeckia and

88

evaluated the anti-inflammatory effects and potential molecular mechanisms of this

89

compound on RAW264.7 cells for the first time. The present study provides research

90

basis and experimental data for developing new treatments on RA with LB.

91 92

2. Materials and Methods

93

2.1 Chemicals and reagents

94

Acetonitrile (ACN, HPLC grade) and methanol (HPLC grade) were purchased

95

from RCI Labscan Limited (Thailand). Phosphoric acid (analytical grade) was

96

purchased from Sigma Chemicals Ltd. (St. Louis, MO, USA). Milli-Q water was

97

prepared using a Milli-Q system (Millipore, MA, USA).

98

3-[4, 5-Dimethyl-2-thiazolyl]-2, 5-diphenyltetrazolium bromide (MTT),

99

dimethyl sulfoxide (DMSO) and lipopolysaccharides (LPS) from Escherichia coli

100

O111:B4 were purchased from Sigma-Aldrich (St. Louis, MO, United States).

101

Dulbecco's modified eagle's medium (DMEM), fetal bovine serum (FBS),

102

phosphate-buffered saline (PBS), penicillin-streptomycin (10,000 U/ml, P/S), 0.25%

103

Trypsin-EDTA (w/v), Nuclear and Cytoplasmic Protein Extraction Kit and Pierce 4

104

LDH Cytotoxicity Assay Kit were obtained from Thermo Fisher Scientific (Waltham,

105

MA, USA). Enzyme-linked immunosorbent assay (ELISA) kits were purchased from

106

Neobioscience Technology Co., Ltd. (Shenzhen, China). ELISA kit for PGE2 were

107

from Cayman Chemical (Cayman Chemical, Ann Arbor, MI, United States).

108

Radioimmunoprecipitation assay (RIPA) lysis buffer was obtained from Beyotime

109

Biotechnology (Jiangsu, China). Chemiluminescent kit was supplied by GE

110

Healthcare UK Limited (Buckinghamshire, HP7 9NA, UK). Primary antibodies

111

against phosphorylation (p)-IKKα/β, IKKα, IKKβ, p-IκBα, IκBα, p-NFκB p65,

112

COX-2, iNOS, HO-1, Nrf2, Lamin B2 and GAPDH (glyceraldehyde-3-phosphate

113

dehydrogenase), and the secondary antibody were purchased from Cell Signaling

114

Technology (Danvers, MA, United States).

115

2.2 Preparation of LB

116

The SH samples were authenticated by the corresponding author of Dr. Hua YU.

117

The voucher specimens (SG-003) were deposited at the Institute of Chinese Medical

118

Sciences, University of Macau, Macao, China.

119

The powdered SH herb (700g) was extracted thrice with 50% ethanol (1:10, w/v)

120

for 1 h each under reflux. The combined extracts were filtered with filter paper after

121

cooling and then concentrated under reduced pressure to a appreciate volume (700 ml).

122

The concentrated extract was loaded onto a macroporous resin (D101) column (~700

123

ml) and allowed for statically-adsorbing overnight. Subsequently, the sample was

124

washed with distilled water (10-fold column volume) and then eluted with 8-fold

125

column volume of 95% ethanol. The collected eluent was concentrated under reduced

126

pressure to 200 ml and extracted 3 times with 200 ml of ethyl acetate. The combined

127

ethyl acetate extract was dried under nitrogen at room temperature. The dried extract

128

was dissolved in methanol and separated using a preparative liquid chromatographic

129

system. The corresponding chromatographic peaks were collected, concentrated and

130

lyophilized to obtain LB. The purity of LB was >98% (detected by HPLC analysis).

131

Spectroscopy data of the prepared LB were in agreement with those reported in the

132

literature (Macías and H. Fischer, 1992).

133 5

134

2.3 Cell culture

135

RAW264.7 cells were purchased from the American Type Culture Collection

136

(ATCC; Manassas, VA, USA). The cells were cultured with 10% heat-inactivated FBS

137

and 1% P/S in the atmosphere of 95% humidity and 5% CO2 at 37 °C. When the cells

138

reached 80% confluence, the cells were sub-cultured after scraping from a flask (25

139

cm 2 ; Thermo Fisher Scientific, MA, USA).

140

2.4 Cytotoxicity assay

141

RAW264.7 cells were seeded in 96-well plates (1 x 104 cells/well) and allowed

142

to adhere overnight. Cells were co-treated with the indicated concentrations of LB for

143

24 h in the absence or presence of LPS (1 µg/ml). Thereafter, the supernatant was

144

collected to determine the release of lactate dehydrogenase (LDH) according to the

145

manufacture’s protocol. The cells in the plate were incubated for an additional 3 h

146

with fresh medium containing 0.5 mg/ml MTT. The supernatant was removed, and the

147

absorbance of the dissolved precipitate (in 150 µl of DMSO) was measured at 490 nm

148

using a microplate reader (FlexStation 3; Molecular Devices, USA).

149

2.5 Measurement of NO

150

RAW264.7 cells were seeded in 96-well plates (2 x 104 cells/well) and allowed

151

to adhere overnight. Cells were pretreated with indicated concentrations of LB for 1 h,

152

then stimulated with LPS (1µg/ml) for 12 h. Subsequently, NO production was

153

determined by measuring the nitrite accumulated in the medium with Griess reagent.

154

The absorbance of the final reaction solution was measured at 540 nm using a

155

microplate reader.

156

2.6 ELISA assay

157

RAW264.7 cells were seeded in 24-well plates (1 x 105 cells/well) and allowed

158

to adhere overnight. Cells were pretreated with LB (5, 10, 20 µM) for 1 h, then

159

stimulated with LPS (1µg/ml) for 12 h. The cytokines (TNF-α, IL-6 and MCP-1) and

160

prostaglandin E2 (PGE2) in the supernatant were detected using an ELISA kit

161

according to the manufacturer's instructions.

162

2.7 Determination of intracellular reactive oxygen species levels

163

RAW264.7 cells were seeded in 12-well plates (2 x 105 cells/well) and allowed 6

164

to adhere overnight. Cells were pretreatment with LB (5, 10, 20 µM) for 1 h, then

165

stimulated with LPS (1µg/ml) for 12 h. The cells were scraped from the plates after

166

washing once with PBS, then centrifugated 5 min at 200×g.

167

Dichloro-dihydro-fluorescein diacetate (DCFH-DA, 10 µM in DMEM) was added to

168

the cells and incubated the cells for 30 min at room temperature. After that, the cells

169

were washed twice with PBS. Finally, the cells were resuspended into PBS and then

170

taken photos by immunofluorescence microscope and count by flow cytometer.

171

2.8 Western blot analysis

172

Total protein was extracted from the harvested cells with RIPA buffer containing

173

a mixture of 1 mM phenylmethylsulfonyl fluoride and protease inhibitor. Nuclear

174

Protein was extracted with Nuclear and Cytoplasmic Protein Extraction Kit according

175

to the manufacture’s protocol. Proteins (15-40 µg per sample) were isolated by

176

SDS-PAGE (8%-12%) and then transferred to a polyvinylidene fluoride (PVDF)

177

membrane (Bio-Rad, Hercules, CA, USA). The membrane was blocked with 5%

178

defatted dry milk for 2 h and then incubated overnight at 4 °C with antibodies against

179

p-IKKα/β, IKKα, IKKβ, p-IκBα, IκBα, p-NFκB p65, COX-2, iNOS, HO-1, Nrf2,

180

Lamin B2 and GAPDH. After washing, the membrane was incubated with the

181

secondary antibody for 1.5 h at room temperature. The blot was visualized using an

182

enhanced chemiluminescence kit. Digital images of the blots were generated by a

183

Syngene gel imaging system (Bio-Rad) and quantified using Syngene software.

184

2.9 Immunofluorescence staining

185

RAW264.7 cells were seeded at 5 x 105 cells per dish into a confocal culture dish

186

(NEST Biotechnology Co., Ltd; Wuxi, Jiangsu, China). Overnight, the cells were

187

pre-treated with LB for 1 h then stimulated with LPS (1 µg/ml) for 1h. After treatment,

188

cells were washed once with PBS, fixed with 4% paraformaldehyde for 10 min,

189

washed three times with washing solution (PBS containing 0.1% Triton X-100 ), and

190

blocked with 3% BSA (containing 0.1% Triton X-100 ) for 1 h at room temperature.

191

Then, the cells were incubated with primary antibody against NF-κB p65 (Cell

192

Signaling Technology, #8242) for 1 h at room temperature, washed three times with

193

washing solution, and then incubated with Aelxa 594 Fluor (red) secondary antibody 7

194

for 1 h at room temperature. Finally, cells were stained with

195

4',6-diamidino-2-phenylindole (DAPI) for 4 min, then observed with a confocal laser

196

microscope (Leica, Buffalo Grove, USA).

197 198 199

2.10 Statistical analysis Data were analyzed using GraphPad Prism 6.0 software. All experimental data

200

are presented as mean ± S.D., and each experiment was performed at least three times.

201

Significant differences between groups were determined using a one-way ANOVA

202

with Dunnet’s multiple comparisons test; P < 0.05 was considered difference

203

significantly.

204

3. Results

205

3.1. Toxicity of LB on RAW264.7 cells

206

The MTT results showed that LB at concentrations of 0.0625–20 µM for 24 h

207

was non-toxic to RAW264.7 cells (Fig. 1B), and the LB (0.0625–20 µM) attenuated

208

the LPS-induced cell injury when co-incubated with LPS (1µg/ml) for 24 h (Fig. 1C).

209

Lactate dehydrogenase (LDH) in the culture medium is an index to evaluate the

210

integrity of cell membrane, the Fig. 1D showed that LB reduced the LDH release

211

induced by LPS (1µg/ml) dose-dependently and LB (20 µM) alone did not lead to the

212

release of LDH. These results show that LB (≦20 µM) was non-toxic to RAW264.7

213

cells and rescued the cells from LPS-induced injury.

214

3.2. LB reduced the production of NO and pro-inflammatory proteins in

215

LPS-stimulated RAW264.7 cells

216

Excessive NO is a classical marker for inflammation in activated macrophages

217

(Wu et al., 2018). As Fig. 2A shows, LPS induced the dramatic increase of NO, which

218

could be significantly reduced by LB (1.25–20 µM) in a concentration dependent

219

manner. When inflammation occurs, activated macrophages would secrete large

220

amounts of pro-inflammatory proteins to aggravate inflammation (Su et al., 2017). As

221

shown in Fig. 2B-E, LPS increased the expression of IL-6, TNF-a, MCP-1 and PGE2,

222

which could be reduced by the pretreatment of LB.

223

3.3. LB inhibited the reactive oxygen species production in LPS-stimulated RAW264.7 8

224

cells It is well accepted that inflammation is linked with oxidative stress. Oxidative

225 226

stress refers to elevated intracellular levels of reactive oxygen species that is

227

considered as the most potent inflammatory mediators (Ji et al., 2018). As shown in

228

Fig. 3A&B, LPS (1 µg/ml, 12 h) induced the excessive reactive oxygen species

229

production, LB inhibited the LPS-induced reactive oxygen species dose-dependently.

230

The Fig. 3C&D showed that LB significantly decreased the fluorescence intensity

231

compared to LPS group, these data reconfirmed that LB could reverse LPS-induced

232

reactive oxygen species level.

233

3.4. LB suppressed the expression of iNOS and COX-2 in LPS-stimulated RAW264.7

234

cells

235

It has been reported that iNOS and COX-2 play an important role in the

236

inflammatory process (Fu et al., 2014). Based on the significant inhibition of NO and

237

PGE2 by LB, we examined the expression of iNOS and COX-2. As shown in Fig. 4,

238

LPS induced the expression of iNOS and COX-2, which could be reduced by LB

239

dose-dependently.

240

3.5. LB blocked the activation of NF-κB in LPS-stimulated RAW264.7 cells

241

NF-κB is a typical signaling pathway involved in LPS-mediated inflammation

242

(Mingfeng et al., 2014). As shown in Fig. 5A&B, the expression of p-IKKα/β and

243

p-IκBα in the LB group was lower than LPS group, which indicated the LB could

244

reduce the phosphorylation of IKKα/β (Fig. 5A) and IκBα. Fig. 5C&D showed that

245

LB decreased the expression of NF-κB p65 in nucleus and reduced the nuclear

246

translocation of NF-κB p65. These data indicated that LB blocked LPS-induced

247

NF-κB signaling pathway.

248

3.6. LB increased the translocation of Nrf2 and the expression of HO-1 in

249

LPS-stimulated or unstimulated RAW264.7 cells

250

The activation of Nrf2 pathway could inhibit the progression of inflammation

251

(Abd El-Twab et al., 2019). As shown in Fig. 6A, LB increased the expression of Nrf2

252

in nucleus in the presence or absence of LPS, which suggested LB could activate the

253

Nrf2. Nrf2 increased the transcription of its target genes, including HO-1 and NQO1 9

254

(Abd El-Twab et al., 2019). Antioxidant protein HO-1 has anti-oxidative and

255

anti-inflammatory effects (Pooladanda et al., 2019). In order to understand whether

256

LB could exert its anti-inflammatory effects through activating Nrf2 pathway, the

257

downstream protein HO-1 of Nrf2 pathway was investigated. As shown in Fig. 6B,

258

the expression of HO-1 was increased significantly in the presence or absence of LPS.

259

These data suggested that LB could increase the transcription of Nrf2, and further

260

up-regulate the protein expression of HO-1.

261 262 263

4. Discussion Sigesbeckiae Herba (SH) is a traditional Chinese medicine used for chronic

264

inflammatory diseases, especially for rheumatoid arthritis (RA). Based on our

265

previous research on SH since 2015, we isolated and identified an active

266

sesquiterpene lactone (Lecocarpinolide B, LB) which shows stronger NO inhibitory

267

effect and better solubility than kirenol, one of the main compounds widely reported

268

in the literatures (Kim et al., 2014; Lu et al., 2012; Wang et al., 2011; Xiao et al.,

269

2015). Further investigations showed that LB inhibited the production of TNF-α, IL-6,

270

MCP-1 and PGE2 in LPS-induced RAW264.7 macrophages (Fig. 2), suggesting the

271

potentiality of LB for developing an anti-inflammatory drug against RA and/or other

272

chronic inflammatory diseases.

273

COX-2 and iNOS are two critical proteins in the process of initiation and

274

progression of inflammation, both of them are reported to be the trigger of

275

pro-inflammatory mediators (NO, PGE2, TNF-α, IL-6 and MCP-1), inhibition of these

276

two proteins ameliorated the inflammation (Kwon et al., 2013). COX-2 is also a key

277

target for non-steroidal anti-inflammatory drugs (NSAIDs) in anti-inflammation. As

278

shown in Fig. 4, LB reduced the expression of these two proteins, which was well

279

consistent with the strong inhibition on the production of NO and PGE2 (Fig. 2).

280

NF-κB has been confirmed to be one of the up-stream signals of inflammation

281

cascade reaction, inhibition of NF-κB reduced the expression of COX-2 and iNOS

282

and decreased the production of inflammatory mediators (Park et al., 2007). NF-κB

283

has been widely reported to be involved in the modulation of RA (Makarov, 2001). 10

284

Therefore, we examined the effects of LB on NF-κB, the results in Fig. 5 showed that

285

LB blocked the NF-κB p65 activation and translocation from the cytoplasm to nucleus

286

by inhibition the phosphorylation of IKKα/β and IκBα. Similarly, Helenalin, an

287

anti-inflammatory sesquiterpene lactone from Arnica, selectively inhibits transcription

288

factor NF-κB (G et al., 1997). Parthenolide, a sesquiterpene lactone, has been well

289

known to be an anti-inflammatory agent that inhibits NF-κB activation (Kwok et al.,

290

2001; Mathema et al., 2012).

291

Reactive oxygen species is also a critical mediator of oxidative stress and

292

inflammation, and involved in the development and deleterious stage of several

293

inflammatory diseases (Mittal et al., 2014). Dichloro-dihydro-fluorescein diacetate

294

(DCFH-DA) is a cell-permeable fluorescent probe and has been widely used to detect

295

intracellular production of reactive oxygen species (Nitrogen, 2017), it is commonly

296

used for measuring reactive oxygen species or oxidant stress in cells or tissues in

297

combination with confocal microscopy or flow cytometry (Shen et al., 2018). As

298

shown in Fig. 3, the positive cell counting and fluorescence images all revealed that

299

LB reduced the intracellular reactive oxygen species in LPS-induced RAW264.7 cells,

300

and the LB alone did not alter the level of reactive oxygen species in normal cell.

301

Therefore, we further detected the effects of LB on Nrf2 which was a domain

302

mediator of oxidative stress. As shown in Fig. 6A, LB increased the expression of

303

Nrf2 in nucleus in the presence or absence of LPS, LB also increased the expression

304

of HO-1 (Fig. 6B) which was a critical anti-oxidative and anti-inflammatory protein

305

in the down-stream of Nrf2 signal pathway (Pooladanda et al., 2019). These data

306

suggested that the attenuation of LB on oxidative stress might be involved in the

307

activation of Nrf2/HO-1 signal pathway.

308

Sesquiterpenoids are groups of important bioactive ingredients in SH herbs.

309

Based on the chemical structures, most of them are germacrane-type sesquiterpenoids

310

(including LB) (Liu et al., 2019), while only a few were cadinane-type and

311

eudesmane-type squiterpenoids (Sakuda, 1987). The reported pharmacological

312

activity of SH sesquiterpenoids including anti-cancer, anti-microbial and

313

anti-inflammation. In one recent study, Liu et al isolated 27 germacrane-type 11

314

sesquiterpenoids from SH and some of them presented anti-cancer activities against

315

human A549 and MDA-MB-231 cells in vitro (Liu et al., 2019). In addition, Jang et al

316

compared the anti-inflammatory activities of 21 compounds isolated from SH, and

317

found that highly oxygenated germacrane-type sesquiterpenoids could significantly

318

inhibit LPS-induced NO production in RAW 264.7 macrophages (Jang et al., 2018).

319

However, due to the limited amount of the obtained compounds, the potential

320

pharmacological mechanisms of these sesquiterpenoids were not further investigated

321

and reported. In this study, a germacrane-type sesquiterpenoid of LB was purified

322

from the SH extract and presented potent anti-inflammatory effect in LPS-induced

323

RAW 264.7 macrophages through regulating NF-κB and Nrf2 signaling pathways. In

324

addition, the structure-activity relationship among the SH sesquiterpenoids to their

325

anti-inflammatory activities should be further investigated and elucidated.

326

In summary, the present study isolated and identified the anti-inflammatory and

327

anti-oxidative activities of natural compound (LB) from SH for the first time, and the

328

mechanisms of these actions could at least be related to the inhibition of

329

NF-κB-mediated pro-inflammatory mediators and the activation of Nrf2-mediated

330

anti-oxidant proteins.

331 332 333

Conflict of interest The authors declare that there is no conflict of interest.

334 335 336

Acknowledgments This work was supported by grants from the National Natural Science

337

Foundation of China [NSFC, No. 81470170], the Science and Technology

338

Development Fund, Macau SAR [File No. 0096/2019/A2] and the Research

339

Committee of the University of Macau [MYRG2017-00178-ICMS and

340

MYRG2018-00043-ICMS].

341 342

References

343

Abd El-Twab, S.M., Hussein, O.E., Hozayen, W.G., Bin-Jumah, M., Mahmoud, A.M., 12

344

2019. Chicoric acid prevents methotrexate-induced kidney injury by suppressing

345

NF-κB/NLRP3 inflammasome activation and up-regulating Nrf2/ARE/HO-1

346

signaling. Inflamm. Res. 68, 511–523.

347

https://doi.org/10.1007/s00011-019-01241-z

348

Ahmed, S.M.U., Luo, L., Namani, A., Wang, X.J., Tang, X., 2017. Nrf2 signaling

349

pathway: Pivotal roles in inflammation. Biochim. Biophys. Acta - Mol. Basis Dis.

350

1863, 585–597. https://doi.org/https://doi.org/10.1016/j.bbadis.2016.11.005

351

Arulselvan, P., Fard, M.T., Tan, W.S., Gothai, S., Fakurazi, S., Norhaizan, M.E.,

352

Kumar, S.S., 2016. Role of Antioxidants and Natural Products in Inflammation.

353

Oxid. Med. Cell. Longev. 2016, 5276130. https://doi.org/10.1155/2016/5276130

354

Cao, Y., Li, F., Luo, Y., Zhang, L., Lu, S., Xing, R., Yan, B., Zhang, H., Hu, W.,

355

2019. 20-Hydroxy-3-Oxolupan-28-Oic Acid Attenuates Inflammatory Responses

356

by Regulating PI3K–Akt and MAPKs Signaling Pathways in LPS-Stimulated

357

RAW264.7 Macrophages. Mol. . https://doi.org/10.3390/molecules24030386

358

Cartagena, E., Colom, O. Álvarez, Neske, A., Valdez, J.C., Bardón,

359

A., 2007. Effects of Plant Lactones on the Production of Biofilm of

360

Pseudomonas aeruginosa. Chem. Pharm. Bull. 55, 22–25.

361

https://doi.org/10.1248/cpb.55.22

362

Chu, J.M.T., Xiong, W., Linghu, K.G., Liu, Y., Zhang, Y., Zhao, G.D., Irwin, M.G.,

363

Wong, G.T.C., Yu, H., 2018. Siegesbeckia Orientalis L. Extract Attenuates

364

Postoperative Cognitive Dysfunction, Systemic Inflammation, and

365

Neuroinflammation. Exp. Neurobiol. 27, 564–573.

366

https://doi.org/10.5607/en.2018.27.6.564

367

Fu, S.-P., Li, S.-N., Wang, J.-F., Li, Y., Xie, S.-S., Xue, W.-J., Liu, H.-M., Huang,

368

B.-X., Lv, Q.-K., Lei, L.-C., Liu, G.-W., Wang, W., Liu, J.-X., 2014. BHBA

369

suppresses LPS-induced inflammation in BV-2 cells by inhibiting NF-κB

370

activation. Mediators Inflamm. 2014, 983401.

371

https://doi.org/10.1155/2014/983401

372 373

G, L., TJ, S., I, M., HL, P., 1997. Helenalin, an anti-inflammatory sesquiterpene lactone from Arnica, selectively inhibits transcription factor NF-κB. Format Biol 13

374 375

Chem. 378, 951–61. Jaiswal, A.K., 2004. Nrf2 signaling in coordinated activation of antioxidant gene

376

expression. Free Radic. Biol. Med. 36, 1199–1207.

377

https://doi.org/https://doi.org/10.1016/j.freeradbiomed.2004.02.074

378

Jang, H., Lee, J.W., Kim, J.G., Hong, H.R., Phoung, T., Le, L., Hong, J.T., Kim, Y.,

379

Lee, M.K., Hwang, B.Y., 2018. Bioorganic Chemistry Nitric oxide inhibitory

380

constituents from Siegesbeckia pubescens. Bioorg. Chem. 80, 81–85.

381

https://doi.org/10.1016/j.bioorg.2018.05.022

382

Ji, Y., Dai, Z., Sun, S., Ma, X., Yang, Y., Tso, P., Wu, G., Wu, Z., 2018.

383

Hydroxyproline Attenuates Dextran Sulfate Sodium-Induced Colitis in Mice:

384

Involvment of the NF-κB Signaling and Oxidative Stress. Mol. Nutr. Food Res.

385

62, 1800494. https://doi.org/10.1002/mnfr.201800494

386

Kim, M.-B., Song, Y., Hwang, J.-K., 2014. Kirenol stimulates osteoblast

387

differentiation through activation of the BMP and Wnt/β-catenin signaling

388

pathways in MC3T3-E1 cells. Fitoterapia 98, 59–65.

389

https://doi.org/https://doi.org/10.1016/j.fitote.2014.07.013

390

Kobayashi, E.H., Suzuki, T., Funayama, R., Nagashima, T., Hayashi, M., Sekine, H.,

391

Tanaka, N., Moriguchi, T., Motohashi, H., Nakayama, K., Yamamoto, M., 2016.

392

Nrf2 suppresses macrophage inflammatory response by blocking

393

proinflammatory cytokine transcription. Nat. Commun. 7, 11624.

394

https://doi.org/10.1038/ncomms11624

395

Kwok, B.H.B., Koh, B., Ndubuisi, M.I., Elofsson, M., Crews, C.M., 2001. The

396

anti-inflammatory natural product parthenolide from the medicinal herb

397

Feverfew directly binds to and inhibits IκB kinase. Chem. Biol. 8, 759–766.

398

https://doi.org/https://doi.org/10.1016/S1074-5521(01)00049-7

399

Kwon, D.J., Ju, S.M., Youn, G.S., Choi, S.Y., Park, J., 2013. Suppression of iNOS

400

and COX-2 expression by flavokawain A via blockade of NF-κB and AP-1

401

activation in RAW 264.7 macrophages. Food Chem. Toxicol. 58, 479–486.

402

https://doi.org/10.1016/j.fct.2013.05.031

403

Lee, S.C., Kwon, Y.-W., Park, J.-Y., Park, S.Y., Lee, J.-H., Park, S.-D., 2017. 14

404

Antioxidant and Anti-Inflammatory Effects of Herbal Formula SC-E3 in

405

Lipopolysaccharide-Stimulated RAW 264.7 Macrophages. Evidence-based

406

Complement. Altern. Med. 2017. https://doi.org/10.1155/2017/1725246

407

Li, D., Liu, Q., Sun, W., Chen, X., Wang, Y., Sun, Y., Lin, L., 2018a.

408

1,3,6,7-Tetrahydroxy-8-prenylxanthone ameliorates inflammatory responses

409

resulting from the paracrine interaction of adipocytes and macrophages. Br. J.

410

Pharmacol. 175, 1590–1606. https://doi.org/10.1111/bph.14162

411

Li, D., Liu, Q., Sun, W., Chen, X., Wang, Y., Sun, Y., Lin, L., 2018b.

412

1,3,6,7-Tetrahydroxy-8-prenylxanthone ameliorates inflammatory responses

413

resulting from the paracrine interaction of adipocytes and macrophages. Br. J.

414

Pharmacol. 175, 1590–1606. https://doi.org/10.1111/bph.14162

415

Li, Yan, Wu, Yannan, Yao, X., Hao, F., Yu, C., Bao, Y., Wu, Yin, Song, Z., Sun, Y.,

416

Zheng, L., Wang, G., Huang, Y., Sun, L., Li, Yuxin, 2017. Ginkgolide a

417

ameliorates LPS-induced inflammatory responses in vitro and in vivo. Int. J. Mol.

418

Sci. 18, 1–15. https://doi.org/10.3390/ijms18040794

419

Li, Z., Feng, H., Wang, Y., Shen, B., Tian, Y., Wu, L., Zhang, Q., Jin, M., Liu, G.,

420

2019. Rosmarinic acid protects mice from

421

lipopolysaccharide/d-galactosamine-induced acute liver injury by inhibiting

422

MAPKs/NF-κB and activating Nrf2/HO-1 signaling pathways. Int.

423

Immunopharmacol. 67, 465–472.

424

https://doi.org/https://doi.org/10.1016/j.intimp.2018.12.052

425

Liu, N., Wu, C., Yu, J., Zhu, K., Song, M., Yang, F., Feng, R., 2019. Bioorganic

426

Chemistry Germacrane-type sesquiterpenoids with cytotoxic activity from

427

Sigesbeckia orientalis. Bioorg. Chem. 92, 103196.

428

https://doi.org/10.1016/j.bioorg.2019.103196

429

Lu, Y., Xiao, J., Wu, Z.-W., Wang, Z.-M., Hu, J., Fu, H.-Z., Chen, Y.-Y., Qian, R.-Q.,

430

2012. Kirenol exerts a potent anti-arthritic effect in collagen-induced arthritis by

431

modifying the T cells balance. Phytomedicine 19, 882–889.

432

https://doi.org/https://doi.org/10.1016/j.phymed.2012.04.010

433

Macías, F.A., H. Fischer, N., 1992. Melampolides from Lecocarpus pinnatifidus. 15

434

Phytochemistry 31, 2747–2754.

435

https://doi.org/https://doi.org/10.1016/0031-9422(92)83624-8

436

Makarov, S.S., 2001. NF-kappa B in rheumatoid arthritis: a pivotal regulator of

437

inflammation, hyperplasia, and tissue destruction. Arthritis Res. 3, 200–206.

438

https://doi.org/10.1186/ar300

439

Mathema, V.B., Koh, Y.-S., Thakuri, B.C., Sillanpää, M., 2012. Parthenolide, a

440

Sesquiterpene Lactone, Expresses Multiple Anti-cancer and Anti-inflammatory

441

Activities. Inflammation 35, 560–565.

442

https://doi.org/10.1007/s10753-011-9346-0

443

Mingfeng, D., Xiaodong, M., Yue, L., Taikui, P., Lei, X., Ming, L., 2014. Effects of

444

PPAR-γ Agonist Treatment on LPS-Induced Mastitis in Rats. Inflammation 37,

445

1919–1924. https://doi.org/10.1007/s10753-014-9924-z

446

Mittal, M., Siddiqui, M.R., Tran, K., Reddy, S.P., Malik, A.B., 2014. Reactive oxygen

447

species in inflammation and tissue injury. Antioxid. Redox Signal. 20,

448

1126–1167. https://doi.org/10.1089/ars.2012.5149

449

Park, H.J., Kim, I.T., Won, J.H., Jeong, S.H., Park, E.Y., Nam, J.H., Choi, J., Lee,

450

K.T., 2007. Anti-inflammatory activities of ent-16αH,17-hydroxy-kauran-19-oic

451

acid isolated from the roots of Siegesbeckia pubescens are due to the inhibition

452

of iNOS and COX-2 expression in RAW 264.7 macrophages via NF-κB

453

inactivation. Eur. J. Pharmacol. 558, 185–193.

454

https://doi.org/10.1016/j.ejphar.2006.11.036

455

Pooladanda, V., Thatikonda, S., Bale, S., Pattnaik, B., Sigalapalli, D.K., Bathini, N.B.,

456

Singh, S.B., Godugu, C., 2019. Nimbolide protects against endotoxin-induced

457

acute respiratory distress syndrome by inhibiting TNF-α mediated NF-κB and

458

HDAC-3 nuclear translocation. Cell Death Dis. 10, 81.

459

https://doi.org/10.1038/s41419-018-1247-9

460

Ralph, P., Nakoinz, I., 1977. Antibody-Dependent Killing of Erythrocyte and Tumor

461

Targets by Macrophage-Related Cell Lines: Enhancement by PPD and LPS. J.

462

Immunol. 119, 950 LP – 954.

463

Sakuda, Y., 1987. The Constituents of Essential Oil from Siegesbeckia pubescens 16

464

Makino. J. Japan Oil Chem. Soc. 36, 667–670.

465

https://doi.org/10.5650/jos1956.36.667

466

Sang, W., Zhong, Z., Linghu, K., Xiong, W., Tse, A.K.W., Cheang, W.S., Yu, H.,

467

Wang, Y., 2018. Siegesbeckia pubescens Makino inhibits Pam3CSK4-induced

468

inflammation in RAW 264.7 macrophages through suppressing

469

TLR1/TLR2-mediated NF-ΚB activation. Chinese Med. (United Kingdom) 13,

470

1–10. https://doi.org/10.1186/s13020-018-0193-x

471

Scott DL, Wolfe F, H.T., 2010. Rheumatoid arthritis. Lancet. 376, 1094–108.

472

Shin, E.M., Zhou, H.Y., Guo, L.Y., Kim, J.A., Lee, S.H., Merfort, I., Kang, S.S., Kim,

473

H.S., Kim, S., Kim, Y.S., 2008. Anti-inflammatory effects of glycyrol isolated

474

from Glycyrrhiza uralensis in LPS-stimulated RAW264.7 macrophages. Int.

475

Immunopharmacol. 8, 1524–1532.

476

https://doi.org/https://doi.org/10.1016/j.intimp.2008.06.008

477

Smolen, J.S., Aletaha, D., McInnes, I.B., 2016. Rheumatoid arthritis. Lancet 388,

478

2023–2038. https://doi.org/https://doi.org/10.1016/S0140-6736(16)30173-8

479

Su, M., Cao, J., Huang, J., Liu, S., Im, D.S., Yoo, J.W., Jung, J.H., 2017. The in vitro

480

and in vivo anti-inflammatory effects of a phthalimide PPAR-γ agonist. Mar.

481

Drugs 15. https://doi.org/10.3390/md15010007

482

Wang, J., Zhou, Y., Ye, Y., Shang, X., Cai, Y., Xiong, C., Wu, Y., Xu, H., 2011.

483

Topical anti-inflammatory and analgesic activity of kirenol isolated from

484

Siegesbeckia orientalis. J. Ethnopharmacol. 137, 1089–1094.

485

https://doi.org/https://doi.org/10.1016/j.jep.2011.07.016

486

Wu, X., Gao, H., Hou, Y., Yu, J., Sun, W., Wang, Y., Chen, Xiaoyan, Feng, Y., Xu,

487

Q. ming, Chen, Xiuping, 2018. Dihydronortanshinone, a natural product,

488

alleviates LPS-induced inflammatory response through NF-κB, mitochondrial

489

ROS, and MAPK pathways. Toxicol. Appl. Pharmacol. 355, 1–8.

490

https://doi.org/10.1016/j.taap.2018.06.007

491

Xiao, J., Yang, R., Yang, L., Fan, X., Liu, W., Deng, W., 2015. Kirenol attenuates

492

experimental autoimmune encephalomyelitis by inhibiting differentiation of Th1

493

and th17 cells and inducing apoptosis of effector T cells. Sci. Rep. 5, 9022. 17

494 495

https://doi.org/10.1038/srep09022 Zhang, Q.R., Zhong, Z.F., Sang, W., Xiong, W., Tao, H.X., Zhao, G.D., Li, Z.X., Ma,

496

Q.S., Tse, A.K.W., Hu, Y.J., Yu, H., Wang, Y.T., 2019. Comparative

497

comprehension on the anti-rheumatic Chinese herbal medicine Siegesbeckiae

498

Herba: Combined computational predictions and experimental investigations. J.

499

Ethnopharmacol. 228, 200–209.

500

https://doi.org/https://doi.org/10.1016/j.jep.2018.09.023

501

Zhong, Z., Zhang, Q., Tao, H., Sang, W., Cui, L., Qiang, W., Cheang, W.S., Hu, Y.,

502

Yu, H., Wang, Y., 2019. Anti-inflammatory activities of Sigesbeckia glabrescens

503

Makino: combined in vitro and in silico investigations. Chin. Med. 14, 35.

504

https://doi.org/10.1186/s13020-019-0260-y

505 506 507 508 509 510

18

511 512

Figure legends

513

Fig. 1 Toxicity of Lecocarpinolide B (LB) on RAW264.7 cells. The chemical structure

514

of LB drawn by ChemDraw software (A). RAW264.7 cells were treated with LB

515

(0.625-40 µM) in the absence (B) or presence (C) of LPS (1µg/ml) for 24 h, then the

516

cell viability was detected by MTT assay. RAW264.7 cells were treated with LB (5,

517

10, 20 µM) in the absence or presence of LPS (1µg/ml) for 24 h, then the lactate

518

dehydrogenase (LDH) in the culture medium was determined by LDH assay kit (D).

519

Data are mean ± S.D. of minimum three independent experiments. * P<0.05, **

520

P<0.01 and ***P<0.001 versus Control; ## P<0.01 and ### P<0.001 versus LPS; No

521

significance (NS).

522

Fig. 2 Lecocarpinolide B (LB) reduced the production of NO and pro-inflammatory

523

proteins in LPS-stimulated RAW264.7 cells. Cells were pretreatment indicated

524

concentrations of LB for 1 h, then stimulated with LPS (1µg/ml) for 12 h, the

525

supernatant was collected for the detection of NO with Griess reagent (A), detection

526

of IL-6 (B), MCP-1 (C), TNF-α (D), PGE2 (E) with ELISA kits. Data are mean ± S.D.

527

of minimum three independent experiments. *** P<0.001 versus Control; # P<0.05, ##

528

P<0.01 and ### P<0.001 versus LPS.

529

Fig. 3 Lecocarpinolide B (LB) inhibited the reactive oxygen species production in

530

LPS-stimulated RAW264.7 cells. Cells were pretreatment with LB (5, 10, 20 µM) for

531

1 h, then stimulated with LPS (1µg/ml) for 12 h. After incubation the cells with

532

DCFH-DA (10 µM in DMEM) for 30min at room temperature, the positive cells were

533

used to count by flow cytometer (A&B) and visualize with immunofluorescence

534

microscope (C&D). Data are mean ± S.D. of minimum three independent experiments.

535

***

536

significance (NS).

537

Fig. 4 Lecocarpinolide B (LB) suppressed the expression of iNOS and COX-2 in

538

LPS-stimulated RAW264.7 cells. Cells were pretreatment with LB (5, 10, 20 µM) for

539

1 h, then stimulated with LPS (1µg/ml) for 12 h. The expression of COX-2 and iNOS

540

were analyzed by western blotting. Data are mean ± S.D. of minimum three

P<0.001 versus Control; # P<0.05, ## P<0.01 and ### P<0.001 versus LPS; No

19

541

independent experiments. *** P<0.001 versus Control; # P<0.05 and ### P<0.001

542

versus LPS.

543

Fig. 5 Lecocarpinolide B (LB) blocked the activation of NF-κB in LPS-stimulated

544

RAW264.7 cells. Cells were pretreatment with LB (5, 10, 20 µM) for 1 h, then

545

stimulated with LPS (1µg/ml) for 1 h. The phosphorylation of IKKα/β (A),

546

phosphorylation of IκBα (B), the expression of p65 in nucleus (C), the translocation

547

of p65 to nucleus (D). Data are mean ± S.D. of minimum three independent

548

experiments. *** P<0.001 versus Control; ### P<0.001 versus LPS.

549

Fig. 6 Lecocarpinolide B (LB) increased the translocation of Nrf2 and the expression

550

of HO-1 in LPS-stimulated or unstimulated RAW264.7 cells. Cells were pretreatment

551

with LB (5, 10, 20 µM) for 1 h, then stimulation with LPS (1µg/ml) for 1 h. The

552

expression of HO-1 in the total proteins (A) and the expression of Nrf2 in nucleus (B)

553

were analyzed by western blotting. Data are mean ± S.D. of minimum three

554

independent experiments. ** P<0.01 and *** P<0.001 versus Control.

20

555 556

Fig. 1. Toxicity of Lecocarpinolide B (LB) on RAW264.7 cells

557

21

558

559 560

Fig. 2. Lecocarpinolide B (LB) reduced the production of NO and pro-inflammatory

561

proteins in LPS-stimulated RAW264.7 cells

22

562 563

Fig. 3. Lecocarpinolide B (LB) inhibited the reactive oxygen species (ROS)

564

production in LPS-stimulated RAW264.7 cells

565

23

566

567 568

Fig.4. Lecocarpinolide B (LB) suppressed the expression of iNOS and COX-2 in

569

LPS-stimulated RAW264.7 cells

570

24

571 572

Fig. 5. Lecocarpinolide B (LB) blocked the activation of NF-κB in LPS-stimulated

573

RAW264.7 cells

574

25

575

576 577

Fig. 6. Lecocarpinolide B (LB) increased the translocation of Nrf2 and the expression

578

of HO-1 in LPS-stimulated or unstimulated RAW264.7 cells

579

26