Immune Activation of RAW264.7 Macrophages by Low Molecular

Sep 27, 2018 - ...
0 downloads 0 Views 1MB Size
Subscriber access provided by UNIVERSITY OF TOLEDO LIBRARIES

Bioactive Constituents, Metabolites, and Functions

Immune activation of RAW264.7 macrophages by low molecular weight fucoidan extracted from New Zealand Undaria pinnatifida Bi Decheng, Boming Yu, Qingguo Han, Jun Lu, William Lindsey White, Qiuxian Lai, Nan Cai, Wenqi Luo, Liang Gu, Sheng Li, Hong Xu, Zhangli Hu, Shao-Ping Nie, and Xu Xu J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.8b03698 • Publication Date (Web): 27 Sep 2018 Downloaded from http://pubs.acs.org on September 28, 2018

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 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 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.

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 33

Journal of Agricultural and Food Chemistry

1

Immune activation of RAW264.7 macrophages by low molecular weight

2

fucoidan extracted from New Zealand Undaria pinnatifida

3

Decheng Bi a,1, Boming Yu a,1, Qingguo Han a,1, Jun Lu a,b,*, William Lindsey White b,

4

Qiuxian Lai a, Nan Cai a, Wenqi Luo a, Liang Gu a, Sheng Li a, Hong Xu a, Zhangli

5

Hu a, Shaoping Nie c, Xu Xu a,*

6

a

7

Bioresources and Ecology, Shenzhen University, Shenzhen 518060, PR China.

8

b

9

and Environmental Sciences, Auckland University of Technology, Auckland 1142,

College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Marine

School of Science and School of Interprofessional Health Studies, Faculty of Health

10

New Zealand.

11

c

12

Nanchang 330047, Jiangxi, China.

13

*

14

+86-755-26534274, +64-9-9219175

15

E-mail addresses: [email protected] (X. Xu) and [email protected] (J. Lu)

16

1

State Key Laboratory of Food Science and Technology, Nanchang University,

Corresponding authors. Tel.: +86-755-26534977, +64-9-9219999x7381; fax:

These authors contributed equally to this work.

17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

18

Abstract

19

Fucoidan, a sulfated polysaccharide extracted from brown seaweeds, has been

20

shown to possess various bioactivities. In particular, low molecular weight fucoidan

21

(LMWF) has been shown to have better bioactivities. In this study, a LMWF (< 10

22

kDa) was extracted from New Zealand Undaria pinnatifida and investigated for its

23

immune modulation effects. LMWF at a concentration range from 1 to 50 µg/ml

24

exerted an effective immune activation in RAW264.7 macrophages. LMWF treatment

25

promoted significant NO release, iNOS expression and TNF-α and IL-6 secretion in a

26

concentration-dependent manner. It also significantly stimulated the activation of

27

NF-κB and MAPK signalling pathways, and specific inhibitors of NF-κB and MAPK

28

pathways diminish the stimulation, confirming the activation pathways. These results

29

indicate that LMWF possesses potential health benefits through immune-stimulation,

30

which may lead to future pharmaceutical development.

31 32

Keywords: low molecular weight fucoidan; Undaria pinnatifida; immune activation;

33

nitric oxide, New Zealand.

34 35

ACS Paragon Plus Environment

Page 2 of 33

Page 3 of 33

Journal of Agricultural and Food Chemistry

36

Introduction

37

The innate immune system of mammalian is mediated by phagocytes such as

38

macrophages and neutrophils, which is the first line of defence against pathogens.1

39

Macrophage, an important component of the innate and adaptive immune system,

40

plays a vital role in host defence against infection and tumor through inflammation

41

response,2 in which pro-inflammatory mediators are working together.3 The

42

inflammatory mediator production in RAW264.7 macrophages have been reported to

43

be regulated by NF-κB and MAPK signalling pathways.4 NF-κB (p65:p50 dimer)

44

migrates within the cell matrix and binds to the IκB in resting cells. After stimulated

45

by various stimulants, the IκB is phosphorylated and releases p65:p50 dimers. Then,

46

p65:p50 dimers pass through the nucleopore to enter the nucleus and bind the target

47

gene to enhance its transcription.5,6 MAPKs are comprised of three protein kinase

48

subtypes: p38, c-Jun JNK and ERK, and are regulated by Ras. These kinases control

49

many physiological processes, such as cell differentiation, proliferation and immune

50

response.7

51

Marine seaweed polysaccharides, including fucoidan, alginate and carrageenan,

52

have been studied in diverse research fields including drug and functional food

53

development.8,9 Fucoidan mainly exists in the cytoderm and intercellular space of

54

edible brown seaweeds where it occupies about 40% dry weight of the alga cell walls

55

and approximately 16% dry weight of the whole seaweed.10,11.

56

Brown seaweeds such as Ascophyllum nodosum, Fucus vesiculosus, Laminaria

57

angustata and Undaria pinnatifida are often used for the preparation of fucoidan.8

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 4 of 33

58

Fucoidan is mainly composed of fucose and also contains some galactose, xylose,

59

uronic acids and even protein in various quantities.12 It is proposed that the fucose

60

units are linked via either (1-3)-glycosidic bonds, or alternating (1-3)- and

61

(1-4)-glycosidic bonds. Sulphate groups are mainly at O-2 or O-4 positions of the

62

fucose backbone. There are single fucoside or short fuco-oligosaccharide side chains

63

at C-2 or C-4 position of the backbone. Acetylation may occur at the O-2 position of

64

the fucose unit.13 While fucoidan used to be considered as a polysaccharide consists

65

of a fucose-only-backbone, some authors now argue that the term fucoidan should

66

include

67

(1-2)-β-D-mannose backbones.13 Since fucoidan has a structure similar to heparin, it

68

has potent activities of anticoagulation and antithrombosis.14

sulphated

galactofucans

which

have

(1-6)-β-D-galactose

and/or

69

It has been reported that fucoidan has numerous other bioactivities such as

70

antitumor,15 anti-inflammatory,16 antiviral,13 and immunomodulatory17 activities.

71

However, it is difficult for the human body to utilize fucoidan due to its high

72

molecular weight (MW) which would affect the absorption.18 Cho et al. have reported

73

that low MW fucoidan (LMWF) extracted from Korea U. pinnatifida has more

74

effective anticancer activity compared with high MW fucoidan (HMWF).19 As

75

reported, fucoidan can induce nitric oxide (NO) production via p38 MAPK and

76

NF-κB-dependent signalling pathways.20 Furthermore, enzyme-modified Korea

77

Hizikia fusiforme, of which the main component is LMWF, has shown a more

78

significant promoting effect of NO and tumor necrosis factor-α (TNF-α) production

79

and phagocytotic activity of RAW264.7 cells than normal extracts.21 It is known that

ACS Paragon Plus Environment

Page 5 of 33

Journal of Agricultural and Food Chemistry

80

fucoidan varies significantly between source species, the environment the source

81

seaweeds were cultivated in or harvested from, and the time of the harvest during the

82

year. No two isolated fucoidans are exactly the same even if they are extracted from

83

the same seaweed species; they are all unique in their structure, composition and

84

bioactivities.13

85

In this research, we assessed the immunomodulatory function of a LMWF

86

extracted from New Zealand U. pinnatifida (which has never been studied for its

87

effects on the immune system) on murine macrophage RAW264.7 cells in terms of

88

NO and cytokine production and the NF-κB and MAPK signalling pathways

89

activation.

90

Experimental Section

91

Materials

92

Lipopolysaccharides (LPS), Polymyxin B (PMB), NG-nitro-L-arginine methyl

93

ester (L-NAME) and Pyrrolidine dithiocarbamate (PDTC) were purchased from

94

Sigma-Aldrich (St. Louis, MO, USA). Inhibitors SB 20358, SP 600125 and PD 98059

95

were obtained from Selleck (Shanghai, China). Cell counting kit (CCK)-8 and

96

radioimmunoprecipitation assay (RIPA) buffer were obtained from Beyotime (Jiangsu,

97

China). Antibodies against inducible nitric oxide synthase (iNOS), IκB-α,

98

phosphor-IκB-α (p-IκB-α), p65, p-p65, p38, p-p38, JNK, p-JNK, ERK and p-ERK

99

and horseradish peroxidase (HPR)-conjugated secondary antibody were purchased

100

from Cell Signaling Technology (Beverly, MA, USA). Antibodies against α-tubulin

101

and GAPDH were obtained from Proteintech (Hubei, China).

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

102

Page 6 of 33

Preparation of the fucoidan and its fractions

103

Fucoidan was prepared from New Zealand U. pinnatifida as described previously.22

104

In brief, seaweed was soaked in 70ºC hot water overnight. After centrifugation, 2%

105

calcium chloride was added to remove alginate. Then, anhydrous ethanol was added

106

to precipitate fucoidan. Fucoidan was freeze-dried to obtain powder, packed under

107

nitrogen, sealed in foil pack, and stored in dark and room temperature (RT) prior to

108

further analyses.

109

The crude fucoidan’s chemical composition has been analyzed. The total

110

carbohydrate content of the fucoidan samples was quantified by the phenol-sulfuric

111

acid method using fucose as a standard. The sulphate content was determined by the

112

BaCl2-gelatin method with K2SO4 as the standard after hydrolyzing the samples in

113

1M HCl at 105˚C for 12 hours. Uronic acid was measured by the carbazole-sulfuric

114

acid method using glucuronic acid as the standard. The protein content was estimated

115

by a Bicinchoninic acid protein assay kit (Beyotime Biotech., Jiangsu, China). The

116

separation and quantification of monosaccharide constituents of the fucoidan

117

polysaccharides

118

chromatography coupled with a pulsed amperometric detector (HPAEC-PAD)

119

(Thermo Fisher Scientific, Waltham, MA, USA ).

were

determined

by

high-performance

anion

exchange

120

LMWF was obtained via dialysis. Fucoidan was dissolved in water to make 1%

121

solution. The solution was put into a 10,000 Dalton dialysis bag and kept at 4 oC for

122

three days. The solution out of the bag was collected and freeze-dried to obtain

123

300

197

kDa), middle molecular weight fucoidan (MMWF, 10-300 kDa) and LMWF (< 10

198

kDa) (Fig. 1A). Using dialysis bag to separate fucoidan into different molecular

199

fraction recovered 90% of the fucoidan, with 16% 300 kDa

200

fractions. Fractions were re-run with HPGPC and the resulting chromatograms were

201

overlaid in Figure 1A. The LMWF contains 2.76±0.31% sugar, 1.42±0.03% uronic

202

acid, 7.04±0.47% amino acids, and 16.62±1.31% sulfate.

203

Endotoxin pollution assay of fucoidan

204

Endotoxin LPS, a vital cytoderm constituent of the Gram-negative bacteria, is an

205

effective trigger of inflammatory mediators.25,26 We used PMB, an LPS inhibitor, to

206

assess whether there was any endotoxin pollution in fucoidan. RAW264.7 cells were

207

treated with 100 ng/mL of LPS or 50 µg/mL of fucoidan (Crude, HMWF and LMWF

208

fucoidan) with or without PMB (2.5 µg/mL), and NO release in the medium was

ACS Paragon Plus Environment

Page 10 of 33

Page 11 of 33

Journal of Agricultural and Food Chemistry

209

measured. NO secretion in LMWF-treated cells was not impacted by PMB. However,

210

NO secretions in Crude and HMWF treatments were inhibited by PMB significantly

211

(Fig. 1B). These results suggested that the endotoxin contamination might exist in the

212

Crude and HMWF fractions, but not in the LMWF fraction, and the observed

213

LMWF-triggered NO release was the result of activation of the macrophage by

214

LMWF itself. Therefore, we further studied the macrophage activation induced by

215

LMWF instead of by Crude and HMWF.

216

LMWF triggers NO release in RAW264.7 macrophages.

217

Up to 50 µg/mL of LMWF showed no cytotoxicity on RAW264.7 macrophages

218

(Fig. 2A). Next, we investigated NO release triggered by LMWF in RAW264.7

219

macrophages. As presented in Fig. 2B and 2C, NO production was induced by

220

LMWF in time- and concentration-dependent manners and the NO production in

221

macrophage reached a higher level while treated with 50 µg/mL of LMWF for 24 h.

222

LMWF enhances iNOS levels in RAW264.7 macrophages.

223

NO production in macrophages is from L-arginine and regulated by iNOS.27 LMWF

224

treatment significantly up-regulated both the mRNA and protein expression of iNOS

225

in RAW264.7 macrophages in a concentration-dependent manner (Fig. 3A and B).

226

Furthermore, NO production in LMWF-triggered RAW264.7 macrophages was

227

attenuated by NOS inhibitor L-NAME (Fig. 3C).

228

LMWF induces TNF-α and IL-6 secretion in RAW264.7 macrophages.

229

In the innate immune system, pro-inflammatory mediators secreted by

230

macrophages serve vital roles in defence against pathogens.3 TNF-α and IL-6

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 12 of 33

231

secretions in LMWF-treated RAW264.7 macrophages were measured using an

232

ELISA kit. As presented in Fig. 4A and B, TNF-α and IL-6 productions were elevated

233

dose-dependently with LMWF stimulation. We also determined the mRNA level of

234

TNF-α and IL-6 in LMWF-treated RAW264.7 macrophages using RT-PCR analysis.

235

As shown in Fig. 4C, LMWF treatment significantly up-regulated the mRNA

236

expression of TNF-α and IL-6 in a concentration-dependent manner.

237

LMWF activates the NF-κB and MAPK signalling pathways in RAW264.7

238

macrophages.

239

NF-κB

and

MAPK

signalling

pathways

are

the

major

regulators

of

240

pro-inflammatory mediator production in activated macrophages.28,29 The NF-κB and

241

MAPK signalling pathways activation were analyzed using Western blot analysis

242

after RAW264.7 macrophages had been stimulated by LMWF or LPS for 15, 30 and

243

60 min. As shown in Fig. 5A, the levels of phosphorylated IκB-α and p65 induced by

244

LMWF in RAW264.7 macrophages were in a dose-dependent manner. However, the

245

p-IκB-α and p-p65 levels in macrophage treated with LMWF for 60 min were lower

246

than that for 30 min (Fig. 5A). As for the MAPK signalling pathway, LMWF rapidly

247

induced the phosphorylation of p38 in time- and concentration-dependent manners

248

(Fig. 5B). Interestingly, LMWF induced phosphorylation of JNK showed a significant

249

change after 30 min stimulation, however, the p-ERK levels show a weak change

250

until 60 min stimulation (Fig. 5B).

251

In order to confirm whether the activation of NF-κB and MAPK signalling

252

pathways was associated with NO, TNF-α and IL-6 production in RAW264.7

ACS Paragon Plus Environment

Page 13 of 33

Journal of Agricultural and Food Chemistry

253

macrophages stimulated by LMWF, inhibitors of NF-κB, p38, JNK and ERK were

254

used. It was demonstrated that NO, TNF-α and IL-6 secretion in LMWF-induced

255

RAW264.7 macrophages was attenuated by NF-κB inhibitor PDTC, p38 inhibitor SB

256

20358 or JNK inhibitor SP 600125 (Fig. 5C, D and E); while there was no effect of

257

ERK inhibitor PD 98095 on NO and TNF-α production (Fig. 5C and D). However,

258

the PD 98095 could attenuate the IL-6 production in LMWF-triggered RAW264.7

259

cells, it suggests that the weak activation of ERK MAPK induced by LMWF play an

260

important role in the IL-6 secretion. Above results suggest that LMWF from New

261

Zealand U. pinnatifida activated macrophages mainly via NF-κB and MAPK

262

signalling pathways.

263

Discussion

264

This is the first study to investigate the immune activation and its mechanism of

265

LMWF from New Zealand U. pinnatifida. As reported, fucoidan has various

266

bioactivities, and these bioactivities are associated with the molecular weight,24

267

monosaccharide composition, the degree of sulphation,30 glycosidic linkages, the

268

degree of branching and substitution, and chain conformation.31 It has been reported

269

that fucoidan has the ability to activate immunity.20,32 However, structure and

270

bioactivity of fucoidan vary significantly owing to the source species, cultivated

271

environment, time of harvesting and method of extraction.33 Recent studies have

272

discovered that LMWFs from Korea Hizikia fusiforme and Iran Sargassum

273

angustifolium have a much stronger ability on promoting NO secretion in RAW264.7

274

macrophages.21,32 Another study finds that fucoidan purchased from Sigma-Aldrich

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 14 of 33

275

can promote NO release in RAW264.7 macrophages through p38 MAPK and NF-κB

276

signalling pathways.20 However, the species from which this fucoidan was extracted

277

has not been mentioned. There are three types of fucoidan available from

278

Sigma-Aldrich, extracted from Fucus vesiculosus, U. pinnatifida and Macrocystis

279

pyrifera, respectively. One report shows that fucoidan provided by Sigma-Aldrich

280

(Fucus vesiculosus) does not have a strong immune activation ability.8 Another study

281

suggests

282

immunomodulatory function than that derived from U. pinnatifida, both from

283

Sigma-Aldrich.34 As mentioned before, various fucoidans extracted from different

284

species have different bioactivities.33 Therefore, it is important to study the

285

immunomodulatory effect of fucoidan extracted from different species and areas. In

286

this study, we prepared an LMWF (