Accepted Manuscript Activation of the NLRP3 inflammasome by cellular labile iron Kyohei Nakamura, Toru Kawakami, Naoki Yamamoto, Miyu Tomizawa, Tohru Fujiwara, Tomonori Ishii, Hideo Harigae, Kouetsu Ogasawara PII:

S0301-472X(15)00730-4

DOI:

10.1016/j.exphem.2015.11.002

Reference:

EXPHEM 3329

To appear in:

Experimental Hematology

Received Date: 10 April 2015 Revised Date:

28 October 2015

Accepted Date: 1 November 2015

Please cite this article as: Nakamura K, Kawakami T, Yamamoto N, Tomizawa M, Fujiwara T, Ishii T, Harigae H, Ogasawara K, Activation of the NLRP3 inflammasome by cellular labile iron, Experimental Hematology (2015), doi: 10.1016/j.exphem.2015.11.002. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

15.51.Exp Hematology.Nakamura K et al. 1

ACCEPTED MANUSCRIPT

Immunobiology and immunotherapy

2

Title: Activation of the NLRP3 inflammasome by cellular labile iron.

3

Kyohei Nakamura1,2, Toru Kawakami1,3, Naoki Yamamoto1, Miyu Tomizawa1,

4

Tohru Fujiwara2, Tomonori Ishii2, Hideo Harigae2, and Kouetsu Ogasawara*1

5

1 Department of Immunobiology, Institute of Development, Aging, and Cancer,

6

Tohoku University, Sendai, Japan

7

2 Department of Hematology and Rheumatology, Tohoku University Graduate

8

School of Medicine, Sendai, Japan

9

3 Department of Thoracic Surgery, Institute of Development, Aging and Cancer,

TE D

M AN U

SC

RI PT

1

Tohoku University, Sendai, Japan

11

Address corresponding to K.O; Department of Immunobiology, Institute of

12

Development, Aging and Cancer, Tohoku University, Sendai, 980-8575, Japan;

13

Phone: 81-22-717-8579, Fax: 81-22-717-8452, Email: [email protected]

14

200 words in Abstract, 1892 words in Text, 906 words in Method, 41 References, 5

15

Figures

16

AC C

EP

10

15.51.Exp Hematology.Nakamura K et al. 2

ACCEPTED MANUSCRIPT 17

Abstract

18

Cellular labile iron, which contains chelatable redox-active Fe2+, has been implicated

19

in iron-mediated cellular toxicity leading to multiple organ dysfunction.

20

homeostasis is controlled by monocytes/macrophages through their iron recycling

21

and storage capacities. Furthermore, iron sequestration by monocytes/macrophages

22

is regulated by pro-inflammatory cytokines including IL-1, highlighting the importance

23

of these cells in the crosstalk between inflammation and iron homeostasis. However,

24

a role for cellular labile iron in monocytes/macrophages-mediated inflammatory

25

responses has not been defined. Here we show that cellular labile iron activates the

26

NLRP3 inflammasome in human monocytes. Stimulation of LPS-primed PBMCs with

27

ferric ammonium citrates (FAC) increases the level of cellular Fe2+ levels in

28

monocytes and induces production of IL-1β in a dose-dependent manner. This FAC-

29

induced IL-1β production is caspase-1-dependent and is significantly inhibited by an

30

Fe2+-specific chelator. FAC consistently induced IL-1β secretion in THP1 cells but

31

not in NLRP3-deficient THP1 cells indicating a requirement for the NLRP3

32

inflammasome. Additionally, the inflammasome activation is mediated by potassium

33

efflux,

34

permeabilization. Thus, these results suggest that monocytes/macrophages not only

35

sequestrate iron during inflammation, but also mediate inflammation in response to

36

cellular labile iron, which provides novel insights into the role of iron in chronic

37

inflammation.

38

AC C

EP

TE D

M AN U

SC

RI PT

Iron

ROS-mediated

mitochondrial

dysfunction,

and

lysosomal

membrane

15.51.Exp Hematology.Nakamura K et al. 3

ACCEPTED MANUSCRIPT Introduction

40

Iron is the major transition metal in the body and plays essential roles in synthesis of

41

DNA, mitochondrial ATP, and heme1. Under physiological conditions, the majority of

42

circulating iron exists in complex with transferrin, which maintains iron in a soluble

43

and non-toxic form. The transferrin-iron complex is taken up by cells via receptor-

44

mediated endocytosis and is then utilized for biological processes including

45

hematopoiesis.

46

overwhelmed, non-transferrin bound iron (NTBI), which is bound mainly by citrate,

47

appears in the plasma such as is seen in patients with iron overload conditions

48

including hemochromatosis, thalassemia, and myelodysplasia2. NTBI is rapidly taken

49

up by cells through unknown mechanisms and is subsequently incorporated into the

50

chelatable, redox-active iron pools called labile iron pools, leading to generation of

51

reactive oxygen species (ROS) through the Fenton reaction3. The oxidative stress

52

generated by cellular labile iron is thought to play a central role in the pathogenesis

53

of organ dysfunction associated with iron overload, including that of the heart, liver,

54

and endocrine systems.

55

responses remain unclear.

SC

RI PT

39

EP

TE D

M AN U

However, when the iron-binding capacity of transferrin is

AC C

However, the effects of cellular labile iron on immune

56

Monocytes/macrophages play a crucial role in systemic iron homeostasis where

57

they phagocytize and degrade damaged or senescent erythrocytes and recycle

58

heme-associated-iron4.

59

monocytes/macrophages5 or released into plasma via the iron export protein,

60

ferroportin6. Under inflammatory conditions, pro-inflammatory cytokines including IL-

The recycled iron can either be stored as ferritin in

15.51.Exp Hematology.Nakamura K et al. 4

ACCEPTED MANUSCRIPT

6 and IL-1 stimulate production of hepcidin from hepatocytes, which strongly inhibits

62

the expression of ferroportin on monocytes/macrophages7 leading to iron retention

63

within these cells. Thus, the hepcidin-ferroportin axis is responsible for anemia of

64

chronic inflammation8.

RI PT

61

The pro-inflammatory cytokine IL-1β is predominantly secreted from monocytes

66

/macrophages through a cytosolic protein complex called the inflammasome9. The

67

NOD-like receptor family, pyrin domain containing 3 (NLRP3) protein, is the best

68

characterized subtype, and plays a pivotal role in host defense against infection as

69

well

70

atherosclerosis11, diabetes12, and Alzheimer’s13.

71

assembled in response to various danger signals, leading to activation of caspase-1,

72

which, in turn, catalytically cleaves the inactive IL-1β precursor protein (pro-IL-1β)

73

into biologically-active IL-1β

74

activation of the NLRP3 inflammasome, including cytosolic potassium efflux14,

75

lysosomal damage15, and ROS-dependent and -independent mitochondrial

76

dysfunction16,17.′ Given that excess cellular labile iron could be toxic for many

77

mammalian cells due to its redox-activity, this may be recognized as a danger signal

78

in monocytes/macrophages. In this study we addressed whether cellular labile iron

79

activates the inflammasome.

80

in

a

variety

of

sterile

inflammatory

diseases

including

gout10,

The NLRP3 inflammasome is

TE D

as

M AN U

SC

65

AC C

EP

Several mechanisms have been proposed for

15.51.Exp Hematology.Nakamura K et al. 5

ACCEPTED MANUSCRIPT Results

82

Cellular labile iron induces IL-1β production in human monocytes.

83

Non-transferrin bound Fe3+ can rapidly enter cells and increases intracellular

84

chelatable iron, known as labile iron pools, which contain redox-active Fe2+. We first

85

investigated whether excess cellular labile iron induces IL-1β production. To this end,

86

we treated LPS-primed PBMCs with ferric ammonium citrate (FAC) as a source of

87

non-transferrin bound Fe3+, and evaluated the levels of IL-1β in culture supernatants.

88

FAC induced IL-1β production in a dose-dependent manner in LPS-primed PBMCs

89

(Figure 1A) and in LPS-primed monocytes (Figure 1B). FAC also decreased cell

90

viability of monocytes in a dose dependent manner (Figure 1C). To evaluate the

91

labile Fe2+ pools within FAC-treated monocytes, we performed calcein-acetoxymethyl

92

ester (calcein-AM) experiments18. We observed that FAC increased the amount of

93

labile Fe2+ within monocytes in a dose-dependent manner (Figure 1D). Furthermore,

94

FAC-induced IL-1β production was inhibited not only by a Fe3+-specific chelator,

95

deferiprone (L1), but also by a Fe2+-specific chelator, 2,2'-bipyridyl (BIP) (Figure 1E),

96

whereas these chelators did not inhibit the IL-1β production by nigericin (Figure 1F).

97

Taken together, cellular Fe2+, namely cellular labile iron, is involved in FAC-induced

98

IL-1β production.

99

Cellular labile iron activates the NLRP3 inflammasome.

100

AC C

EP

TE D

M AN U

SC

RI PT

81

The activation of caspase-1, formally known as IL-1-converting enzyme, is required

15.51.Exp Hematology.Nakamura K et al. 6

ACCEPTED MANUSCRIPT

for processing of pro-IL-1β into the mature form of IL-1β19. As shown in Figure 2A,

102

IL-1β production is significantly inhibited by the pan-caspase inhibitor Z-VAD-FMK.

103

In addition, IL-1β production is completely abrogated by the Z-WEHD-FMK caspase-

104

1 inhibitor, indicating that caspase-1 is required for FAC-induced IL-1β production. In

105

human monocytes, active caspase-1 constitutively exists, which allows monocytes to

106

produce IL-1β in response to priming signal alone20. Consistently, we found that LPS

107

alone stimulated the production of IL-1β in a caspase-1-dependent manner, whereas

108

FAC markedly augmented IL-1β production (Figure 1A, 1B and 2A). To confirm the

109

involvement of the inflammasome-mediated caspase-1 activation, we next performed

110

western blot analysis. As shown in Figure 2B, the mature form of IL-1β and activated

111

form of caspase-1 (p10 subunit) were detectable in culture supernatant, indicating

112

that inflammasome-mediated caspase-1 activation occurs in response to cellular iron.

113

FAC stimulation alone failed to increase the mRNA of IL-1B, suggesting that it chiefly

114

act as an activation signal, but not priming signal (Figure 2C).

TE D

M AN U

SC

RI PT

101

Of the members of the inflammasome family, the NLRP3 inflammasome in

116

particular is known to be activated in response to various sterile stimuli21. Thus, we

117

hypothesized that cellular labile iron specifically activates the NLRP3 inflammasome.

118

To this end, we incubated LPS-primed PBMCs with the NLRP3 inflammasome

119

inhibitor glyburide22 and found significant inhibition of FAC-induced IL-1β production

120

(Figure 3A).

121

activation14,23 and as expected, FAC-induced IL-1β production was completely

122

abrogated in high potassium media (Figure 3A). To further define the involvement of

AC C

EP

115

Potassium efflux is a common trigger of NLRP3 inflammasome

15.51.Exp Hematology.Nakamura K et al. 7

ACCEPTED MANUSCRIPT

NLRP3, we used PMA-differentiated macrophage-like THP1 cells and THP1 cells

124

deficient for NLRP3 (THP1-defNLRP3 cells) (Figure3B). As shown in Figure 3C,

125

FAC treatment increased the level of cellular labile Fe2+ pools in both control-

126

transfected THP1 cells and THP1-defNLRP3 cells; however, FAC-induced IL-1β

127

production was found in PMA-differentiated THP1 cells, but not in THP1-defNLRP3

128

cells (Figure 3D).

129

activates the NLRP3 inflammasome.

130

Cellular labile iron induces ROS production and mitochondrial dysfunction

131

leading to NLRP3 inflammasome activation.

132

Several studies have shown that ROS-dependent and -independent mitochondrial

133

dysfunction activates the NLRP3 inflammasome16,17. Because cellular labile iron is

134

known to catalyze ROS production3, it is possible that ROS-mediated mitochondrial

135

dysfunction is involved in NLRP3 inflammasome activation mediated by cellular labile

136

iron. To address this possibility, we evaluated the ROS generation in FAC-treated

137

monocytes.

138

dependent manner (Figure 4A and 4B).

139

percentage of monocytes with decreased mitochondrial membrane potential (Figure

140

4C and 4D).

141

FAC-induced IL-1β production (Figure 4E)′ and mitochondrial dysfunction (Figure

142

4F).

143

involved in the activation of the NLRP3 inflammasome by cellular labile iron.

RI PT

123

EP

TE D

M AN U

SC

Taken together, these results indicated that cellular labile iron

AC C

We found that FAC treatment increased the ROS level in a doseSimilarly, FAC treatment increased the

Furthermore, pretreatment with N-acetyl-L-cysteine (NAC) inhibited

Collectively, we show that ROS-dependent mitochondrial dysfunction is

15.51.Exp Hematology.Nakamura K et al. 8

ACCEPTED MANUSCRIPT

ROS-mediated lysosomal membrane permeabilization is involved in cellular

145

labile iron-mediated activation of the NLRP3 inflammasome

146

The NLRP3 inflammasome has been reported to be activated by lysosomal damage

147

following phagocytosis of crystalline and particulate materials11,24. ′ Thus, we

148

investigated the involvement of phagocytosis in FAC-induced IL-1β production. We

149

found that cytochalasine D (CytoD), an inhibitor of actin polymerization, failed to

150

inhibit IL-1β production from FAC-treated LPS-primed PBMCs (Figure 5A).

151

Furthermore, CytoD did not inhibit the increase in cellular Fe2+ in FAC-treated

152

monocytes (Figure 5B). These results indicate that actin-mediated phagocytosis is

153

not required for the generation of cellular Fe2+ pools or NLRP3 inflammasome

154

activation. In contrast, FAC-induced IL-1β production was significantly inhibited by

155

the cathepsin B-specific inhibitor, CA-074ME (Figure 5A), suggesting the involvement

156

of phagocytosis-independent lysosomal damage. ROS is known to induce lysosomal

157

membrane permeabilization (LMP), leading to release of cathepsin B25,26. Thus, we

158

evaluated whether cellular labile iron induces LMP through ROS generation by using

159

PMA-differentiated THP1 cells stained with LysoTracker Red.

160

dramatically induced LMP in THP1 cells (seen as decreased fluorescence)

161

pretreatment with NAC partially attenuated FAC-induced LMP (Figure 5C).

162

results suggest that cellular labile iron activates the NLRP3 inflammasome through

163

ROS-dependent LMP and subsequent release of cathepsin B.

164

AC C

EP

TE D

M AN U

SC

RI PT

144

Although FAC

These

15.51.Exp Hematology.Nakamura K et al. 9

ACCEPTED MANUSCRIPT Discussion′ ′

166

In this study, we demonstrate that excess cellular labile iron activates the NLRP3

167

inflammasome leading to secretion of IL-1β in human monocytes. The generation of

168

ROS has been considered to be an upstream event for NLRP3 inflammasome

169

activation in response to a wide range of stimuli16,27. Moreover, cellular labile iron is

170

known to be a potent inducer of ROS through the Fenton reaction28. Consistently,

171

we found that cellular labile iron activates the NLRP3 inflammasome through ROS-

172

dependent mitochondrial dysfunction.

173

activated in response to lysosomal damage following phagocytosis of crystalline and

174

particulate materials including silica, alum, and cholesterol crystals11,24; however, we

175

found that phagocytosis was not required for activation of the NLRP3 inflammasome

176

by cellular iron. Instead, ROS-dependent LMP was involved in this process. Multiple

177

pathways have been implicated for NTBI uptake such as those including stimulator of

178

Fe transport (SFT), DMT-1, ZIP14, and the L-type Ca2+ channel29,30, which might

179

contribute to generation of cellular labile Fe2+ pools in monocytes. Since our results

180

are based on in vitro study, using PBMCs and THP1 cell line with chemical inhibitors,

181

there are limitations in our study. However, our results suggest a novel link between

182

iron and inflammation.

SC

RI PT

165

AC C

EP

TE D

M AN U

The NLRP3 inflammasome is reportedly

183

IL-1β together with IL-6 plays key roles in iron homeostasis during inflammation. In

184

response to IL-1β and IL-6, hepatocytes produce hepcidin, which downregulates the

185

iron exporter channel ferroportin on monocytes/macrophages leading to iron

186

retention within these cells7,8. The role of cellular iron during innate immune reposes

15.51.Exp Hematology.Nakamura K et al. 10

ACCEPTED MANUSCRIPT

has been studied using mice with a myeloid lineage-specific ferroportin deficiency.

188

These mice show iron accumulation in reticuloendothelial macrophages of liver,

189

spleen and bone marrow, and produce higher levels of TNF-α in response to LPS

190

than do control mice31. Conversely, low intracellular iron levels impair TRAM/TRIF-

191

dependent TLR4 signaling in murine macrophages32.

192

therapy

193

encephalomyelitis33 and animal models of rheumatoid arthritis34, highlighting the

194

importance of iron in inflammatory responses. However, the possibility that excess

195

cellular iron may be recognized by an intracellular danger sensor, namely, the

196

inflammasome, had not previously been addressed.

197

production of hepcidin from hepatocytes, iron-mediated NLRP3 inflammasome

198

activation, together with the hepcidin-ferroportin axis, might comprise a positive

199

feedback loop that augments inflammation. Further study is necessary to understand

200

the crosstalk between hepcidin-ferroportin axis and the NLRP3 inflammasome

201

activation by cellular iron.

202

Iron-mediated organ dysfunction is a main cause of the mortality and morbidity in

203

patients with thalassemia major, which causes iron overload due to increased

204

intestinal iron absorption and/or transfusion-dependency35.

205

levels in patients with thalassemia major are higher compared to asymptomatic

206

carriers and heathy controls36.

207

replacement therapy stimulates production of pro-inflammatory cytokines including

208

IL-1β in both healthy subjects and patients receiving hemodialysis37,38. Our results

disease

progression

in

Moreover, iron chelation

experimental

autoimmune

M AN U

SC

ameliorates

RI PT

187

AC C

EP

TE D

Given that IL-1β stimulates

Notably, serum IL-1β

Moreover, several studies have found that iron

15.51.Exp Hematology.Nakamura K et al. 11

ACCEPTED MANUSCRIPT

raise a possibility that NLRP3 inflammasome activation might be involved in the

210

production of pro-inflammatory cytokines in patients with iron-overload. However, it

211

should be noted that plasma NTBI level in patients with iron overload patients are

212

less than 20 µM39, which is considerably lower than the concentration we used in this

213

study. Given that iron deposition in reticuloendothelial system is commonly found in

214

patients with iron overload, NLRP3 inflammasome activation might occur in

215

reticuloendothelial cells that are exposed to locally high concentration of iron.

216

Because inflammasome

217

fibrosis40,41, it is imperative to investigate the role of inflammasome activation in iron-

218

mediated

219

endocrinopathies. Overall, activation of the NLRP3 inflammasome by cellular labile

220

iron might have broad implications for iron overload and chronic inflammation and

221

future study of this system is warranted.

dysfunction

M AN U

plays a critical role for inflammation-associated organ

including

AC C

EP

TE D

organ

SC

RI PT

209

liver

cirrhosis,

cardiomyopathy

and

15.51.Exp Hematology.Nakamura K et al. 12

ACCEPTED MANUSCRIPT Methods

223

Human blood samples

224

Human peripheral blood mononuclear cells (PBMCs) were isolated from healthy

225

donors with informed consent. The study was approved by the Ethical Committee of

226

Tohoku University Graduate School of Medicine.

227

Isolation and stimulation of mononuclear cells

228

The

229

SEPARATE-L (Muto chemical, Tokyo, Japan). Monocytes were purified by negative

230

selection using the EasySep™ Human Monocyte Enrichment Kit (STEMCELL

231

Technologies, Vancouver, Canada). These cells were cultured in RPMI1640 media

232

supplemented with 10% fetal bovine serum, 1 mM sodium pyrubate, 2 mM glutamine,

233

100 U/mL penicillin, and 100 µg/mL streptomycin. After 4 hr priming with 10 ng/ml

234

LPS (from Salmonella Minnesota R595, Enzo Life sciences, Farmingdale, NY), cells

235

were stimulated with ferric ammonium citrate (FAC) (Wako, Osaka, Japan) at the

236

indicated doses for 4 hr or nigericin(20µM, Invivogen, San Diego, CA) for 30 min.

237

For the inhibition assay, LPS-primed PBMCs were pretreated with the following

238

reagents for 30 min before stimulation: Fe3+ chelator deferiprone (400 µM; Sigma),

239

Fe2+-specific chelator 2,2'-bipyridine (400 µM; Sigma), pan caspase Inhibitor Z-VAD-

240

FMK, caspase1/ICE inhibitor Z-WEHD-FMK (100 µM; R&D Systems, Minneapolis,

241

MN), glyburide (200 µM; Sigma-Aldrich), N-acetyl-L-cysteine

was

SC

M AN U

fraction

isolated

by

density-gradient

centrifugation

using

AC C

EP

TE D

PBMC

RI PT

222

(5 mM or 25 mM;

15.51.Exp Hematology.Nakamura K et al. 13

ACCEPTED MANUSCRIPT

Wako), cytochalasin D (10 µM; Wako) and CA074-ME (40 nM; Enzo). The amount of

243

IL-1β in cell culture supernatants was measured with a Human IL-1β ELISA kit

244

(eBioscience, San Diego, CA) according to the manufacturer’s instructions.′ Cell

245

viability was measured by propidium iodide (2µg/ml, Sigma), using FACSCantoII (BD

246

biosciences, San Jose, CA).

RI PT

242

SC

247

Cell lines and transfectants

249

THP1 cells were cultured in RPMI1640 media supplemented with 10% fetal bovine

250

serum, 1 mM sodium pyrubate, 2 mM glutamine, 100 U/mL penicillin, and 100 µg/mL

251

streptomycin).

252

purchased from Invivogen. These cells were differentiated by phorbol 12-myristate

253

13-acetate (PMA) (5 ng/ml; Sigma-Aldrich) 48 h before stimulation on 12 well plate

254

(106 cells / well). After overnight priming with LPS (100 µg/ml), cells were stimulated

255

with the indicated doses of FAC for 6 h.

256

supernatants was measured.

EP

TE D

THP1 cells with reduced NLRP3 activity (THP1-defNLRP3) were

The amount of IL-1β in cell culture

AC C

257

M AN U

248

258

Immunoblot analysis

259

LPS (10 ng/mL)-primed PBMCs were stimulated with the indicated doses of FAC for

260

4 hr. As a positive control, LPS-primed PBMCs were stimulated with ATP (2.5mM,

15.51.Exp Hematology.Nakamura K et al. 14

ACCEPTED MANUSCRIPT

Sigma) for 30 min. Culture supernatants and total cell lysates were collected and

262

then clarified by centrifugation. Proteins in culture supernatants were precipitated

263

with Strataclean Resin (Stratagene, La Jolla, CA). Anti-human caspase-1 p10 Ab (C-

264

20, Santa Cruz), anti-human NLRP3 mAb (D2P5E, Cell signaling technology, Boston,

265

MA), anti-IL-1β mAb (D3U3EH, Cell signaling technology), and anti-α tubulin antibody

266

(DM1A, Santa Cruz) were used for detection.

267

Quantification of intracellular labile iron pools

268

Intracellular labile Fe2+ pools were quantitated using the iron-sensing fluorescent

269

probe, calcein-acetoxy-methyl-ester (calcein-AM) as previously reported18. Briefly,

270

FAC-loaded cells were stained with calcein-AM (250 nM, Dojin Chemical Co,Tokyo,

271

Japan) for 15 min. These cells were washed twice with PBS, and then treated with

272

or without 2,2′-bipyridyl (400 µM, Sigma) for 1 hr.

273

defined as the difference of MFI (FL-1 channel) in the presence or absence of 2,2’-

274

bipyridyl, using FACSCantoII.

275

Quantitative PCR

276

LPS-primed or unprimed PBMCs were stimulated with or without FAC (20mM) for 4hr.

277

Total RNA from PBMCs was extracted using the RNeasy lipid tissue kit (QIAGEN,

278

Hilden, Germany) after stimulation. Total RNA was reverse transcribed into cDNA

279

using SuperScript III with Oligo (dT)12–18 (Invitrogen, Carlsbad, CA, USA). RT-PCR

280

was performed in a DNA chromo 4 (Bio-Rad Laboratories, Hercules, CA, USA).

Intracellular Fe2+ levels were

AC C

EP

TE D

M AN U

SC

RI PT

261

15.51.Exp Hematology.Nakamura K et al. 15

ACCEPTED MANUSCRIPT 281

Following PCR primers were used: IL-1B: sense 5

282

TGA C-3

283

sense 5′-GGA GAG ACC TTT ATG AGA AAG CAA-3′, antisense 5′-GCT GTC TTC

284

CTG GCA TAT CAC A-3′; ACTB: sense 5′-ATT GCC GAC AGG ATG CAG AA -3′,

285

antisense 5′-GCT GAT CCA CAT CTG CTG GAA -3′. SYBR green (QIAGEN) was

286

used for quantification of the amplified DNA. Data were analyzed by Opticon Monitor

287

version 3 software (Bio-Rad laboratories), according to manufacturer’s instruction.

288

ROS detection assay

289

PBMCs (5×105) were stimulated with the indicated doses of FAC for 4 hr, and then

290

cells were stained with APC-conjugated anti-CD14 mAb (Biolegend). The level of

291

ROS in CD14-gated population was measured by total ROS detection kit (Enzo life

292

sciences) according to the manufacturer’s instruction. Mean fluorointensity (MFI) of

293

the FL-1 channel in the CD14-gated population was determined by FACSCantoII.

294

Mitochondrial membrane potential assay

295

To evaluate mitochondrial damage, JC-10 assay (Abcam, Cambridge, UK) was used

296

according to manufacturer’s instruction. Briefly, PBMCs (5×105) were treated with

297

different doses of FAC for 4 hr. After staining with APC-conjugated anti-CD14 mAb,

298

cells were stained with JC-10 dye loading solution for 30 min. The percentage of

299

CD14-positive cells with depolarized mitochondria (green fluorescence) was

300

determined by FACSCantoII.

-GTC GGA GAT TCG TAG CTG GAT-3

; NLRP3:

AC C

EP

TE D

M AN U

SC

RI PT

, antisense 5

-TTA CAG TGG CAA TGA GGA

15.51.Exp Hematology.Nakamura K et al. 16

ACCEPTED MANUSCRIPT Fluorescence microscopy′ ′ ′

302

To evaluate lysosomal membrane permeabilization, PMA-differentiated THP1 cells

303

were loaded with Lysotracker Red (200 nM; Invivogen) and Hoechst 33342 (1 µg/ml;

304

Dojin Chemical Co) for 15 min. After being washed twice with PBS, THP1 cells

305

pretreated with or without 25 mM NAC were incubated with various doses of FAC for

306

the indicated periods. Fluorescent images were acquired using an Olympus IX81

307

microscope (Olympus, Tokyo, Japan) with a 20× objective lens. Merged images

308

were obtained by using Lumina Vision software (Mitani Corporation, Fukui, Japan).

309

Statistical analysis

310

Statistical analysis was performed using the unpaired two-tailed Student's t-test: *P

Activation of the NLRP3 inflammasome by cellular labile iron.

Cellular labile iron, which contains chelatable redox-active Fe(2+), has been implicated in iron-mediated cellular toxicity leading to multiple organ ...
563B Sizes 2 Downloads 18 Views