Article Type: Original Article

2

Received date: 12/17/2013

3

Revised date: 06/04/2014

4

Accepted date: 06/12/2014

5

Title:

Accepted Article

1

6

Functional relevance of NLRP3 inflammasome-mediated IL-1 during acute allergic airway inflammation1

7

Short title:

Anakinra ameliorates allergic airway inflammation

8

Authors:

Manuel Ritter1,2, Kathrin Straubinger2, Stephanie Schmidt2, Dirk H. Busch2,3,

9

Stefanie Hagner4, Holger Garn4, Clarissa Prazeres da Costa2* and Laura E. Layland1,2*

10

1

11

Affiliations:

Institute of Medical Microbiology, Immunology and Parasitology (IMMIP),

12

University Clinic Bonn, Bonn, Germany; 2Institute of Medical Microbiology, Immunology and

13

Hygiene (MIH), Technische Universität München, Munich, Germany; 3Clinical Cooperation

14

Groups ‘‘Antigen-specific Immunotherapy’’ and “Immune Monitoring”, Helmholtz Center

15

Munich (Neuherberg) and Technische Universität München, Munich, Germany; 4Institute of

16

Laboratory Medicine and Pathobiochemistry, Medical Faculty, Philipps-University Marburg,

17

Marburg, Germany.

18 19

*equal contribution

20 21

Corresponding Authors

22

Dr. L. E. Layland

23

Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Clinic

24

Bonn, Sigmund Freud Strasse 25, 53105, Bonn, Germany.

25

Tel: +49 228 287 11387

26

[email protected];

27 28

Dr. C. Prazeres da Costa

29

Institute of Medical Microbiology, Immunology and Hygiene (MIH), Technische Universität

30

München, Trogerstrasse 30, 81675, Munich, Germany; This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process, which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1111/cei.12400

1 This article is protected by copyright. All rights reserved.

Tel: +49 89 4140 4130

32

[email protected]

Accepted Article

31

33 34

Keywords

35

Allergy, Anakinra, IL-1, IL-1 receptor antagonist, NLRP3-inflammasome.

36

Abbreviations: AAI: allergic airway inflammation; AHR: Airway hyper-responsiveness; ASC:

37

apoptosis-associated speck-like protein containing a caspase recruitment domain; BAL:

38

bronchoalveloar lavage; GINA: Global Initiative of Asthma; IL-1R: IL-1 receptor; IL-1Ra: IL-1

39

receptor antagonist; NLRP3: nucleotide oligomerization domain (NOD)-like receptor family,

40

pyrin domain 3; OVA: ovalbumin; RT: room temperature.

41 42

2 This article is protected by copyright. All rights reserved.

Accepted Article

43 44

Summary

45

Overall asthmatic symptoms can be controlled with diverse therapeutic agents. However,

46

certain symptomatic individuals remain at risk for serious morbidity and mortality which

47

prompts the identification of novel therapeutic targets and treatment strategies. Thus, using

48

an adjuvant-free TH2 murine model we have deciphered the role of IL-1 signalling during

49

allergic airway inflammation (AAI). Since functional IL-1 depends on inflammasome

50

activation we first studied asthmatic manifestations in specific inflammasome-deficient

51

(NLRP3-/- and ASC-/-) and IL-1 receptor type 1-/- (IL-1R1-/-) mice on the BALB/c background. To

52

verify the onset of disease we assessed cellular infiltration in the bronchial regions, lung

53

pathology, airway hyper-responsiveness and OVA-specific immune responses. In the absence

54

of NLRP3 inflammasome-mediated IL-1 release all symptoms of AAI were reduced except

55

OVA-specific immunoglobulin levels. To address whether manipulating IL-1 signalling

56

reduced asthmatic development we administered the IL-1R antagonist Anakinra (Kineret®)

57

during critical immunological time-points: sensitization or challenge. Amelioration of

58

asthmatic symptoms was only observed when Anakinra was administered during OVA

59

challenge. Our findings indicate that blocking IL-1 signalling could be a potential

60

complementary therapy for allergic airway inflammation.

61

3 This article is protected by copyright. All rights reserved.

Accepted Article

62 63

Introduction

64

Over the last decade, the importance of inflammasome mediated release of IL-1 has been

65

shown in a variety of models and moreover, has provided the basis for the treatment of

66

several auto-inflammatory diseases such as rheumatoid arthritis, gout and type 2 diabetes

67

[1-3]. In addition, several studies have demonstrated that IL-1 plays an important role

68

during bronchial asthma development. Indeed, asthmatic patients were shown to harbour

69

elevated levels of IL-1 in their bronchoalveloar lavage (BAL) [4]. Other studies have further

70

reported that this proinflammatory cytokine promotes the infiltration of eosinophils and

71

inflammation within the lung and encourages both mast and T cell activation [5,6]. Currently,

72

300 million people are estimated to be affected by asthma worldwide (GINA 2010) and the

73

prevalence continues to increase. Although current therapeutic strategies, such as

74

corticosteroids and long or short acting beta agonists, can control attacks and disease

75

progression, a quarter of a million people still succumbed to the syndrome in 2009 (GINA

76

2009) [7,8]. Asthma is characterized by dominant TH2 immune responses including enhanced

77

IL-4, IL-5 and IL-13 responses, allergen-specific immunoglobulin production, eosinophilia,

78

airway inflammation, bronchoconstriction and airway hyperresponsiveness (AHR) [9-11].

79

Thus, when assessing the onset of asthma in murine models is it imperative to address both

80

cellular and pathological aspects.

81 82

The secretion of functional bioactive IL-1 however, depends on the inflammasome which

83

mediates, via caspase-1 activation, the cleavage of the inactive cytokine precursor (pro-IL-

84

1 into the active form (IL-1. In general, this multiprotein complex contains proteins such

85

as central oligomerization domain (NACHT), a leucine rich repeat (LRR) domain, apoptosis4 This article is protected by copyright. All rights reserved.

associated speck-like protein containing caspase recruitment domain (ASC) and caspase-1

87

[12-14]. For instance, the well-studied NLRP3 inflammasome plays an essential role in a

88

variety of scenarios and is triggered by numerous pathogens such as parasites [15], fungi

89

[16], bacteria [17] or danger signals such as ATP [18] or crystalline silica and asbestos

90

[19,20]. However, its role during allergic asthma remains somewhat controversial. For

91

example, IL-1 receptor type 1 (IL-1R1)-deficient mice present reduced allergic responses and

92

lung inflammation [21,22] and ovalbumin (OVA)-treated NLRP3-/- mice exhibit severely

93

dampened lung inflammation, OVA-specific immunoglobulins, cytokine responses and

94

eosinophil infiltration [23]. In contrast to the latter, Allen et al. demonstrated that allergic

95

airway inflammation within NLRP3-deficient mice is similar to wildtype control mice [24].

96

Both of these NLRP3 inflammasome studies were performed using C57BL/6 mice which are

97

renowned for their dominant TH1 based immune responses and reduced allergic airway

98

inflammation when compared to TH2-biased BALB/c mice [25-28]. Therefore, we have

99

assessed the role of the NLRP3 inflammasome and functional IL-1 during OVA-induced AAI

100

in a BALB/c murine model and observed altered antigen-specific TH responses, reduced

101

eosinophil infiltration and airway inflammation in mice deficient for NLRP3, ASC and the IL-

102

1R1. To analyze the dynamics of functional IL-1 in more detail, we blocked IL-1 signalling

103

using the IL-1R antagonist (IL-1Ra) Anakinra. The application of this therapy ameliorated

104

allergic airway inflammation only during the challenge phase and implies that blocking IL-1

105

signalling could complement current asthma treatment regimen.

Accepted Article

86

106

5 This article is protected by copyright. All rights reserved.

Accepted Article

107 108

Materials and Methods

109

Mice

110

BALB/c and C57BL/6 wildtype mice were purchased from Harlan® (Borchen, Germany). ASC-,

111

NLRP3- and IL-1R1-deficient mice on BALB/c background (backcrossed at least nine

112

generations) were a kind gift from Prof. Jürg Tschopp (University of Lausanne, Switzerland).

113

Mice were bred under specific pathogen-free (spf) conditions in the animal facilities at the

114

Institute of Medical Microbiology, Immunology and Hygiene (Munich, Germany) in

115

accordance with national and EU guidelines 86/809. Experimental mice were sex- and age-

116

matched and the study was approved by the Regierung von Oberbayern, Munich, Germany

117

(Animal License number Az. 55.2.1.54-2532-67-12).

118 119

OVA-induced allergic airway inflammation model and Anakinra treatment

120

Female BALB/c wildtype, ASC-, NLRP3- and IL-1R1-deficient mice were thrice sensitized

121

subcutaneously (s.c.) in the neck with 10µg ovalbumin (OVA) grade VI (Sigma-Aldrich,

122

Taufkirchen, Germany) or PBS (control mice) on days 0, 7 and 14 without adjuvant and

123

consecutively challenged by aerosol inhalation on days 26-28 with 10µg OVA grade V (Sigma-

124

Aldrich). On day 31 (3 days after the last OVA challenge) mice were sacrificed and assessed

125

for allergic airway development (Supporting information, Fig. S1a). For IL-1 type cytokine

126

blocking experiments mice were treated with consecutive doses of Anakinra (Kineret®;

127

Biovitrum, Stockholm, Sweden) during OVA-sensitization or challenge. For details see

128

schemes in Supporting information, Fig. S1b,c. Experiments were performed using either

129

15mg/kg (i.p), 100mg/kg (i.p.) or 150mg/kg (s.c.) in the neck per day.

130

6 This article is protected by copyright. All rights reserved.

Accepted Article

131 132

Measurement of airway hyperresponsiveness (AHR)

133

Airway hyperresponsiveness (AHR) to methacholine (Sigma-Aldrich) was determined using

134

the Flexivent system (SCIREQ, Montreal, Canada). Following anaesthesia the trachea was

135

intubated with a 1.2 mm tracheal cannula and the lungs mechanically ventilated at a

136

respiratory frequency of 150 breaths per min, a tidal volume of 10ml/kg and a positive end-

137

expiratory pressure of 3ml H2O. After exposing mice to aerosolized PBS to retrieve the

138

baseline value, bronchoconstriction was induced using increasing concentrations (1, 2.5, 5,

139

10, 25 and 50 mg/ml in PBS) of aerosolized methacholine using an ultrasonic nebulizer.

140

Dynamic resistance was recorded over 1 minute intervals (every 5 secs) after exposure to

141

defined doses of methacholine via a standardized inhalation manoeuvre (SnapShot-150)

142

[29,30].

143 144

Analysis of bronchoalveolar lavage

145

Mice were euthanized with Narcoren® (Merial, Halbergmoos, Germany) and lungs were

146

flushed twice with 1ml PBS containing proteinase inhibitor cocktail tablets (Roche

147

Diagnostics, Mannheim, Germany). The obtained BAL was weighed and centrifuged at 230g

148

for 5min (4°C). Resulting supernatants were frozen at -20°C and the cytokine content

149

determined using commercially available ELISA kits (eBiosciences, Frankfurt, Germany). The

150

cell pellets were resuspended in PBS containing 2% FCS and counted. To analyze the

151

composition of immune cells within the BAL, 150µl of cell suspension was centrifuged on

152

glass slides at 400rpm for 5min using the Shandon Cytospin 3 centrifuge (Thermo Scientific®,

153

Hamburg, Germany). Glass slides were dried overnight at room temperature (RT) and

154

stained using the Diff-Quick staining set (Medion Diagnostics, Langen, Germany) according to 7 This article is protected by copyright. All rights reserved.

manufacturer’s instructions. Cell differentiation was performed as described previously

156

[29,30].

Accepted Article

155

157 158

OVA-specific immune responses

159

Erythrocyte-depleted mediastinal lymph node (LLN) cells (2x105) from OVA- or PBS-treated

160

mice were co-cultured in the presence or absence of 20µg/ml OVA grade VI at 37 °C in RPMI

161

1640 medium (PAA, Linz, Austria) containing 5% FCS, penicillin/streptomycin, sodium-

162

pyruvate, non-essential amino acids and -mercaptoethanol (all PAA). After 72 hours culture

163

supernatants were analysed for cytokine levels by ELISA.

164 165

Cytokine and OVA-specific immunoglobulin determination by ELISA

166

To decipher in situ cytokine levels, weighed lung samples were placed in 500µl of RPMI 1640

167

medium (without supplements) and homogenized using the T10 basic Ultra-Turrax®

168

disperser (IKA, Staufen, Germany). Samples were then centrifuged at 16,000g for 10min

169

(4°C) and the resulting supernatant frozen at -20°C. The cytokine content was then

170

determined by ELISA. OVA-specific IgE and IgG1 levels were measured in the sera of

171

individual mice [30]. In brief, 96-well ELISA plates (Nunc , Langenselbold, Germany) were

172

coated overnight (4°C) with either 1µg (for IgE) or 0.1µg (for IgG1) OVA grade V diluted in

173

50mM Tris (Roth®, Karlsruhe, Germany) solution containing 3% BSA (PAA; blocking buffer).

174

After washing and blocking, sera was diluted in blocking buffer (1:200 to 1:100,000 dilutions)

175

and standards of mouse α-OVA IgE or IgG1 antibodies (Biozol, Eching, Germany) were

176

applied in two-fold serial dilutions and incubated overnight (4°C). Subsequently, plates were

177

washed and α-mouse IgE or IgG1 biotinylated detection antibodies (Biozol) were applied and

178

incubated for 2h (RT). After further washing streptavidin-horseradish peroxidase (HRP) 8 This article is protected by copyright. All rights reserved.

conjugate (R&D Systems GmbH, Wiesbaden, Germany) was added and plates were

180

incubated for 30min (RT) in the dark. After a final washing step, BD OptEIA™ TMB substrate

181

(BD, Heidelberg, Germany) was applied and reactions were stopped with 2M H 2SO4. Finally,

182

ODs were determined at 450nm using the SunriseTM ELISA microplate reader (Tecan,

183

Crailsheim, Germany). The concentration of the samples was then calculated according to

184

the standard curve.

Accepted Article

179

185 186

Histochemistry and evaluation of lung inflammation

187

Paraffin embedded sections (3µm) from the left lungs of individual mice were stained with

188

PAS (Periodic acid-Schiff), which allows the detection of goblet cells in lung basilar

189

membranes. Sections were analyzed microscopically for tissue inflammation and goblet cell

190

hyperplasia according to previously described methods [29,30]. In short, tissue inflammation

191

was determined by the degree of visual thickness of the basal membrane which was graded

192

on a scale from 0 to 3 (inflammation score). To determine goblet cell hyperplasia, goblet

193

cells within the basal membrane were counted and results are presented as the percentage

194

of goblet cells per length of basal membrane.

195 196

Dendritic cell generation and in vitro inflammasome assays

197

Bone-marrow derived dendritic cells (BMDC) from wildtype C57BL/6, wildtype BALB/c and

198

ASC-/- BALB/c mice were generated using 10µg/ml GMCSF (Peprotech, USA) as previously

199

described [15]. To assess IL-1 activation, 1x105 BMDC were stimulated in RPMI medium

200

containing 10% FCS and supplements (PAA) for 6 hours with LPS (5ng/ml Invivogen, San

201

Diego, USA) and thereafter with ATP (5mM, Carl Roth, Karlsruhe, Germany) for 1 hour. IL-1

202

in the culture supernatant was measured by ELISA according to manufacturer guidelines 9 This article is protected by copyright. All rights reserved.

(eBiosciences).

Accepted Article

203 204 205

Statistical analysis

206

Statistical differences were analyzed using GraphPad Prism 5 software (San Diego, CA, USA).

207

Parametrically distributed data were analyzed using unpaired t-tests or one-way ANOVA.

208

Parametric data are represented as mean ± SD.

209

10 This article is protected by copyright. All rights reserved.

Accepted Article

210 211

Results

212

Lack of NLRP3 inflammasome reduces the development of allergic airway inflammation

213

Since the majority of inflammasome activation studies have been performed in C57BL/6

214

mice, we first investigated whether bone-marrow derived dendritic cells (BMDCs) generated

215

from BALB/c mice were able to produce IL-1in vitro using a standard assay [15]. As shown

216

in Supporting information, Fig. S2a, BMDC derived from C57BL/6 mice produced high

217

amounts of IL-1 following LPS priming and ATP activation. BALB/c BMDC were also able to

218

secrete sufficient amounts of IL-1which were significantly reduced using ASC-deficient

219

BMDC, confirming that BALB/c mice possess functional inflammasome activation [31,32]. To

220

determine the requirement of the NLRP3 inflammasome on the development of AAI, groups

221

of wildtype (WT), ASC- and NLRP3-deficient BALB/c mice were sensitized with PBS or OVA

222

(see methods and Supporting information, Fig. S1a). The three rounds of sensitization were

223

administered s.c. in the absence of an adjuvant such as aluminium hydroxide (Alum) since it

224

is known that Alum can induce the NLRP3 inflammasome and thus IL-1 secretion [33]. In

225

addition, Conrad et al. demonstrated that this adjuvant-free OVA model can induce AAI in a

226

manner similar to models using adjuvants [29]. In the model applied here, mice were

227

challenged with aerosolic OVA over three consecutive days and analyzed 72 hours thereafter

228

(Supporting information, Fig. S1a). A primary parameter of AAI is the influx of immune cells

229

into the BAL and the induction of asthmatic symptoms can be clearly observed when one

230

compares the cellular content in OVA-WT mice to that found in PBS-WT groups (Fig. 1a-c).

231

On first glance, levels of leucocyte infiltration were comparable between OVA-treated WT

232

and inflammasome-deficient strains (Fig. 1a). However, upon further analysis, both knockout

233

strains showed a significant reduction in the percentage of eosinophils (Fig. 1b) and total 11

This article is protected by copyright. All rights reserved.

numbers of eosinophils were significantly reduced in ASC-deficient mice (Supporting

235

information, Fig. S3a). Inflammasome-deficient mice had significantly higher numbers of

236

macrophages (Fig. 1c and Supporting information, Fig. S3b). In contrast, no neutrophil

237

infiltration could be observed in either wildtype or inflammasome-deficient mice

238

(Supporting information, Fig. 3c). In the control groups of mice, no differences were

239

observed in the aforementioned parameters (Fig. 1a-c and Supporting information, Fig. S3a-

240

c). In terms of AHR, sensitized ASC-/- and NLRP3-/- mice showed a significant reduction in

241

airway resistance (Fig. 1d).

Accepted Article

234

242 243

Lung inflammation and goblet cell hyperplasia are reduced in NLRP3 -/- and ASC-/- mice

244

To verify the finding that NLRP3 inflammasome-deficient mice had reduced experimental

245

asthma, lung sections were analyzed for the degree of inflammation and goblet cell

246

hyperplasia (bright red colouring) [29,30]. When compared to lung sections of PBS-sensitized

247

mice (Fig. 2a-c), strong inflammation could be clearly observed in sections from OVA-

248

sensitized and challenged WT mice (Fig. 2d) and this was visibly reduced in ASC-/- (Fig. 2e)

249

and NLPR3-/- mice (Fig. 2f). To quantify these impressions, the extent of inflammation (Fig.

250

2g) and the amount of goblet cell hyperplasia (Fig. 2h) were measured in each individual

251

mouse. When compared to the pathology found in groups of OVA-WT mice, both

252

parameters were significantly reduced in ASC-/- strains (Fig. 2g,h). NLRP3-/- mice showed a

253

significant reduction in the inflammation score (Fig. 2g) but only a mild reduction in the

254

percentage of goblet cells (Fig. 2h). These data confirm that the absence of inflammasome

255

components dampens OVA-induced AAI.

256 257

NLRP3 inflammasome activation alters TH cytokine responses 12

This article is protected by copyright. All rights reserved.

OVA-induced allergic airway inflammation is associated with strong OVA-specific TH

259

responses. To assess whether the diminished airway responses in inflammasome-deficient

260

mice were reflected in their TH profiles, various cytokines were measured in the BAL and

261

within cell-culture supernatants from mediastinal lymph node (LLN) cells that were re-

262

stimulated ex vivo with OVA. Fig. 3a shows that levels of IL-5 and IL-13 but not IFN- in the

263

BAL were significantly down-regulated in both ASC-/- and NLRP3-/- mice. However, OVA

264

stimulated cells from the local draining lymph node of inflammasome-deficient mice

265

secreted significantly less IFN- but not IL-5, when compared to WT animals. In contrast, IL-

266

10 responses were moderately elevated (non-significantly), indicating possible elevations in

267

regulatory processes in ASC- and NLRP3-deficient mice (Fig. 3b). IL-10 levels within the BAL

268

and IL-13 levels of OVA stimulated lymph node cells were not altered in inflammasome-

269

deficient mice (data not shown). Interestingly, no IL-17 responses could be detected within

270

the BAL or OVA stimulated lymph node cells (data not shown). Differential regulation was

271

not observed in OVA-specific B cell responses since levels of IgE and IgG1 in the sera were

272

comparable in wildtype and inflammasome-deficient mice (Fig. 3c).

Accepted Article

258

273 274

Reduced eosinophilia but not lung inflammation and function in the absence of the IL-1R1

275

Levels of in situ IL-1, normalised to individual lung weight, were determined in the lungs of

276

OVA-exposed WT and inflammasome-deficient mice and surprisingly no differences were

277

observed on the final day of analysis (Fig. 4a). However, directly after the last OVA exposure

278

(day 28), significantly higher levels of IL-1 were determined in the WT group (Fig. 4b). This

279

suggested that IL-1 plays a critical role during the primary inflammation stages of asthma

280

but is rapidly surpassed by other immune mechanisms. In contrast, IL-18 levels were slightly

281

higher in the WT group on day 28 but significantly higher on day 31 (Supporting information, 13

This article is protected by copyright. All rights reserved.

Figs. S2b and c, respectively). To further decipher the role of functional IL-1, we studied AAI

283

development in mice deficient for the IL-1 receptor type 1 (Fig. 4c-i). Within the BAL, no

284

differences were observed between the two groups of mice in terms of total leucocyte

285

numbers (Fig. 4c). However, OVA-sensitized IL-1R1-/- mice demonstrated significantly lower

286

levels of eosinophils (Fig. 4d and Supporting information, Fig. S3d), whereas macrophage

287

numbers were comparable to wildtype mice (Supporting information, Fig. S3e). Again, very

288

few neutrophils could be detected in the BAL (Supporting information, Fig. S3f). With regards

289

to pathology, IL-1R1-/- mice showed no significant reduction in either inflammation score

290

(Fig. 4e) or goblet cell influx (Fig. 4f). Interestingly, PBS-treated IL-1R1-/- mice already

291

presented enhanced lung inflammation and goblet cell hyperplasia when compared to naive

292

WT mice (Fig. 4e,f). Moreover, when compared to PBS groups of WT mice, the inflammation

293

in OVA-treated WT groups was significantly enhanced but this was not the case in IL-1R1-/-

294

mice (Fig. 4e). Airway resistance levels in PBS-treated IL-1R1-/- mice were almost equal to

295

those resulting from OVA-treated IL-1R1-/- mice and both groups were higher than in OVA-

296

treated WT mice (Fig. 4g). Interestingly, levels of IL-18 were equal within BAL of asthmatic

297

wildtype and IL-1R1-deficient mice (Supporting information, Fig. S2d). Levels of IL-13 and

298

IFN- within the BAL were increased in IL-1R1-deficient mice whereas levels of IL-5 were

299

significantly lower (Fig. 4h). As with all other inflammasome-deficient animals, OVA-specific

300

immunoglobulin levels remained unchanged (Fig. 4i). These data show that functional IL-1

301

does play a role during AAI and appears to be important for the initiation of immune

302

responses and maintaining the correct balance of cytokines and the cellular composition

303

within the lung.

Accepted Article

282

304 305

Administration of Anakinra during OVA-challenge reduces the development of AAI 14

This article is protected by copyright. All rights reserved.

Finally, we aimed to decipher whether bio-active IL-1 was critical during the sensitization

307

phase or challenge phase of AAI. IL-1 signalling can be blocked through the administration of

308

drugs, such as the endogenous antagonist IL-1Ra Anakinra (Kineret®). Indeed the use of such

309

drugs has been shown to ameliorate IL-1 mediated diseases such as rheumatoid arthritis or

310

type 2 diabetes [1,3]. Thus, BALB/c mice were treated with different doses of Anakinra

311

during either the sensitization (AnakinraSens) or challenge (AnakinraChall) phases of AAI (see

312

Supporting information, Figs. S1b and c, respectively). Treatment doses of Anakinra were

313

either 15 or 100mg/kg i.p. or 150mg/kg s.c. since the administration of Anakinra in patients

314

is usually applied in this manner. These data are respectively depicted in Supporting

315

information, Figs. S4, S5 and Fig. 5. Interestingly, mice injected with any of the employed

316

doses of Anakinra during challenge had significantly reduced eosinophil numbers in the BAL

317

but this was not observed in mice treated during sensitization (Fig. 5a and Supporting

318

information, Figs. S3g, S4a and S5a). When compared to doses of 15 or 100mg/kg i.p.

319

(Supporting information Figures S4b-c and S5b-c), the increased dose of Anakinra via the s.c.

320

route led to significantly reduced disease parameters such as lung inflammation and goblet

321

cell hyperplasia (Fig. 5b,c). With regards to immune profiles, levels of IL-13 and IFN- but not

322

IL-5, were strongly down-regulated in the AnakinraChall group but not the AnakinraSens group

323

(Fig. 5d c.f. filled squares and grey triangles). These modulations to OVA-specific responses

324

were only partially observed using lower doses of Anakinra (Supporting information, Figs.

325

S4d and S5d). Again, levels of OVA-specific IgG1 and IgE were not altered in any of the

326

applied experimental scenarios (Fig. 5e and Supporting information, Figs. S4e and S5e).

327

Moreover, no differences in macrophage levels and again no neutrophil infiltration could be

328

detected (Supporting information, Figs. S3h,i). However, since Anakinra treatment during

329

the challenge phase significantly reduced several AAI parameters we also measured the

Accepted Article

306

15 This article is protected by copyright. All rights reserved.

effects of Anakinra on airway hyperresponsiveness. Interestingly, when compared to OVA

331

groups airway resistance was significantly reduced in Anakinra-treated OVA groups (c.f.

332

closed squares and closed triangles in Fig. 5f). Indeed, levels in the Anakinra-treated group

333

were comparable to control groups of mice (c.f. PBS (open squares) and OVA+Anakinra

334

(closed triangles). Thus, in contrast to the data sets using IL-1R1-deficient mice which

335

showed no significant differences in lung pathology and function the data presented here

336

reveals that specific blocking of IL-1 signaling during the acute challenge phase leads to

337

reduced inflammatory responses against OVA. Moreover, since the PBS IL-1R1 knockout

338

group showed elevated AHR and moderately increased inflammation we demonstrate that a

339

permanent loss of IL-1 signaling has a fundamental effect at the steady state. In conclusion,

340

the data reveal that specific blocking of IL-1 signalling during the acute challenge phase leads

341

to reduced inflammatory responses against OVA and opens up an avenue for exploration

342

into possible therapeutic strategies.

Accepted Article

330

343

16 This article is protected by copyright. All rights reserved.

Accepted Article

344 345

Discussion

346

In vitro, several agents have been shown to trigger the assembly of the NLRP3

347

inflammasome and the consequent release of functional IL-1[13,15,19,20]. However, the

348

dysregulation of IL-1 can elicit severe consequences, such as the rare inherited

349

inflammasome-mediated pathophysiology of autoinflammatory diseases including Muckle-

350

Wells Syndrome and Familial cold autoinflammatory syndrome [34,35]. In both of these

351

cases, the genetic defect lies in multiple point mutations in the NACHT domain of the NLRP3

352

protein which results in a permanent activation of the NLRP3 inflammasome [36,37]. These

353

conditions are classified as cryopyrin-associated periodic syndromes (CAPS) which are a

354

subset of autoinflammatory disorders [1,33] and are distinguished from autoimmune

355

diseases since they fail to induce antigen-specific T cell responses and high antibody titers

356

[35]. Research correlating NLRP3 inflammasome activation and the pathogenesis of these

357

disorders has exploded over the last decade and further evidence shows a fundamental role

358

in the development of other diseases including cancer [1-3,38]. With regards to the latter,

359

murine models of chronic pulmonary fibrotic disorders using silica and asbestos have

360

correlated IL-1 and the onset of inflammation induced lung cancer [19,39].

361 362

The list of diseases that have been successfully treated with the IL-1Ra Anakinra (Kineret®), a

363

non-glycosylated recombinant form of the naturally occurring IL-1Ra, encompasses not only

364

autoinflammatory diseases such as Sweet Syndrome [40] and Relapsing Polychondritis [41]

365

but illnesses such as gout and type 2 diabetes [1-3]. Within those studies, Anakinra

366

demonstrated high efficacy and no adverse side effects. The data presented here display

367

that administration of Anakinra to OVA-allergic mice ameliorates disease parameters, such 17

This article is protected by copyright. All rights reserved.

as eosinophil infiltration and lung inflammation. We blocked IL-1 signalling during either the

369

sensitization or challenge phases and determined that blocking IL-1 signalling during the

370

challenge phase produced the most beneficial results. In accordance, a recent study

371

demonstrated, that administration of IL1-alpha only during the challenge phase (in contrast

372

to the sensitization phase) had disease aggravating effects [42]. Even though studies have

373

demonstrated that the IL-1 family members IL-18 and IL-33 contribute to allergic airway

374

inflammation [43,44], Anakinra specifically blocks IL-1 and IL-1 and therefore indicates

375

that the observed effects here are independent of these cytokines. However, further studies

376

should be performed to identify whether simultaneous blocking of several IL-1 family

377

members during the challenge phase, such as IL-18 and IL-33, is more effective. Even though

378

Anakinra has a good safety record it requires daily injections due to its short half-life (4-6

379

hours). Alternative agents such as Rilonacept (Regeneron®; IL-1 trap) and Canakinumab

380

(Ilaris®; anti-IL-1 antibody), which have much longer half-lives and specifically block IL-1,

381

are currently being assessed in drug trials [45,46]. Thus, it will be interesting to decipher

382

whether these alternative treatments or new upcoming drugs dampen allergic responses in

383

man.

Accepted Article

368

384 385

As mentioned above, the beneficial effects of Anakinra were observed upon application

386

during the challenge phase. In the studies comparing inflammasome-deficient and wildtype

387

animals, we were initially puzzled that the levels of IL-1 within the lungs were comparable

388

on the final day of analysis (day 31). However, further investigation revealed that in WT

389

mice, levels were significantly higher directly after the last exposure to OVA (day 28). These

390

data indicated that the first inflammatory response depended on NLRP3 inflammasome

391

activation that subsequently led to the release of functional IL-1. This hypothesis was 18

This article is protected by copyright. All rights reserved.

verified to some extent using IL-1R1-deficient mice since these mice also displayed reduced

393

eosinophil infiltration and levels of IL-5 in the BAL. However, they also demonstrated

394

elevated levels of IL-13 and IFN- implying that additional factors play a role in the induction

395

of local TH2 and TH1 responses within the IL-1R1-deficient mice. Another intriguing aspect of

396

these studies was observed in naïve IL-1R1 knockout mice since it appeared that a

397

continuous loss of functional IL-1 signalling in the steady-state had a more fundamental

398

effect since deficient animals showed elevated basal levels of inflammation, goblet cell

399

numbers and moreover showed higher airway resistance than OVA-treated wildtype mice.

400

Such effects were not observed in PBS-sensitized mice that were treated with Anakinra

401

demonstrating that selective blocking of IL-1 signalling for a certain period of time does not

402

influence cellular composition and immune responses per se. In correlation, TH1 and TH2

403

levels in the BAL were reduced in Anakinra-treated (challenge) and inflammasome-deficient

404

asthmatic mice, confirming that NLRP3 inflammasome-mediated IL-1responses are crucial

405

for the induction of local inflammation within the lung. Besides the functional relevance of

406

IL-1, we show here that IL-18 is important during AAI. Indeed, levels of IL-18 were

407

significantly reduced in ASC- and NLPR3-deficient strains whereas levels were comparable

408

between WT and IL-1R1-/- mice. Since IL-18 is an important TH1 inducing proinflammatory

409

cytokine [43] this may explain the reduced IFN- levels observed in asthmatic ASC-/- and

410

NLPR3-/- mice but not IL-1R1-/- animals. With regards to OVA-specific responses of re-

411

stimulated lung lymph node cells only IFN- responses were dampened in inflammasome-

412

deficient mice and interestingly, IL-10 levels were actually elevated, albeit not significantly,

413

demonstrating that inflammasome activation influences TH immune responses [15]. IL-17

414

responses were also measured in BAL and antigen-specific culture assays but no differences

415

between the groups could be observed and moreover, levels were extremely low in any of

Accepted Article

392

19 This article is protected by copyright. All rights reserved.

the assessed experiments (data not shown). In addition, almost no IL-17A and IL-17F mRNA

417

expression within asthmatic lungs of WT and ASC-deficient mice could be determined using

418

quantitative RT-PCR (data not shown). These results are in contrast to previous findings since

419

studies have reported that alongside other cytokines, IL-1 induces TH17 immune responses

420

[47-49] that can influence allergic asthma development [50,51]. Moreover, several studies

421

also reported that IL-17 favours neutrophil recruitment leading to the induction of

422

neutrophilia and lung inflammation [52,53]. Interestingly, we could hardly detect neutrophils

423

within the BAL and the frequency of those cells was extremely low compared to other

424

studies [22,23,30]. This might be a possible explanation as to why no IL-17 responses could

425

be detected in any of our currently described experiments. However, the lack of neutrophils

426

within the lungs of asthmatic BALB/c mice and the total absence of IL-17 responses remains

427

unclear. Further studies should investigate the role of neutrophils in correlation with IL-17

428

responses within allergic airway diseases of BALB/c mice.

Accepted Article

416

429 430

The data presented here demonstrate that NLRP3 inflammasome activation is essential for

431

the initiation of inflammatory responses within the lung and the modulation of TH2 immune

432

responses but is dispensable for immunoglobulin production since no differences were

433

observed in levels of OVA-specific IgG1 and IgE. In contrast, Besnard et al. demonstrated that

434

OVA-specific IgE levels were strictly dependent on NLRP3 inflammasome activation, as well

435

as all the other asthmatic parameters [23]. However, this study was performed with C57BL/6

436

mice, which, in contrast to BALB/c mice develop less pronounced AAI and have dominant

437

TH1 based immune responses [25-28]. Moreover, Allen et al. showed that allergic airway

438

inflammation within NLRP3-/- mice on C57BL/6 background is similar to WT control mice [24].

439

These contrasting findings demonstrate the complexity of allergic asthma development and 20

This article is protected by copyright. All rights reserved.

highlight the necessity to employ further distinct methods to analyse the role of cytokines in

441

addition to complete knockout mouse models. Overall, the mechanisms of inflammasome

442

activation during AAI still remains unclear although the data shown here indicate that the

443

NLRP3 inflammasome may play a larger role in controlling AHR and airway remodeling rather

444

than inflammation in AAI. Nevertheless, it has been reported that ATP levels are increased in

445

asthmatic patients [54] and in the BAL of OVA-treated mice [50]. Interestingly, mice that lack

446

the functional purinergic P2-receptor P2X7 which recognizes extracellular ATP displayed

447

reduced features of acute and chronic asthma [55] and epithelial damage has been shown to

448

release uric acid and ATP which provokes NLRP3 inflammasome activation [49,56,57]. Thus,

449

the possible release of DAMPs (i.e. ATP) during allergen exposure could be responsible for

450

NLRP3 inflammasome activation during AAI. Collectively, the data presented within this work

451

demonstrate that NLRP3 inflammasome activation and IL-1 release play a crucial role in the

452

pathogenesis of experimental allergic asthma. Moreover, we show that administration of

453

Anakinra during the acute challenge phase ameliorates allergic airway inflammation and

454

suggests that blocking IL-1 signalling could provide the basis for novel treatment strategies.

Accepted Article

440

455

21 This article is protected by copyright. All rights reserved.

Accepted Article

456 457

Acknowledgements

458

We thank Sabine Paul (MIH) and Vanessa Krupp and Sandra Arriens (IMMIP) for excellent

459

technical support and Thomas Ruppersberg (Philipps-University Marburg, Germany) for

460

helping with AHR measurements. Moreover, we thank Aubry Tardivel (University of

461

Lausanne, Switzerland) for supplying inflammasome deficient mice. In memoriam of Prof.

462

Jürg Tschopp (University of Lausanne, Switzerland). This works was supported by the SFB

463

(Sonderforschungsbereich) Transregio Tr22. The position of MR was funded by the Else

464

Kröner Fresenius Stiftung (EKFS A47/2010). MR, KS, SS, SH, HG and LEL performed the

465

experiments. MR, LEL, DB, HG and CPdC designed the experiments and provided essential

466

expertise. MR, CPdC and LEL wrote the manuscript.

467 468

Conflict of Interest

469

The authors declare no commercial or financial conflict of interest.

470

22 This article is protected by copyright. All rights reserved.

Accepted Article

471 472

References

473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518

1. 2. 3.

4.

5. 6.

7. 8.

9.

10.

11. 12.

13. 14. 15.

16.

17.

18.

Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 2009; 27:519-550. So A, De Smedt T, Revaz S, Tschopp J. A pilot study of IL-1 inhibition by anakinra in acute gout. Arthritis Res Ther 2007; 9:R28. Larsen CM, Faulenbac M, Vaag A, Vølund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Eng J Med 2007; 356:1517-1526. Tillie-Leblond I, Pugin J, Marquette CH et al. Balance between proinflammatory cytokines and their inhibitors in bronchial lavage from patients with status asthmaticus. Am J Respir Crit Care Med 1999; 159:487-494. Chung KF, Barnes PJ. Cytokines in asthma. Thorax 1999; 54:825-857. Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K. Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol 2005; 32:311-318. Pelaia G, Gallelli L, Renda T, Romeo P, Busceti MT, Grembiale RD, Maselli R. Update on optimal use of omalizumab in management of asthma. J Asthma Allergy 2011; 4:49-59. Dolan CM, Fraher KE, Bleecker ER et al. Design and baseline characteristics of the epidemiology and natural history of asthma: Outcomes and Treatment Regimens (TENOR) study: a large cohort of patients with severe or difficult-to-treat asthma. Ann Allergy Asthma Immunol 2004; 92:32-39. Umetsu DT, McIntire JJ, Akbari O, Macaubas C, DeKruyff RH. Asthma: an epidemic of dysregulated immunity Nat Immunol 2002; 3:715-720. Bousquet J, Jeffery PK, Busse WW, Johnson M, Vignola AM. Asthma. From bronchoconstriction to airways inflammation and remodeling. Am J Respir Crit Care Med 2000; 161:1720-1745. Bergeron C, Boulet LP. Structural changes in airway diseases: characteristics, mechanisms, consequences, and pharmacologic modulation. Chest 2002; 129:1068-1087. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 2002; 10:417426. Tschopp J, Martinon F, Burns K. NALPs: a novel protein family involved in inflammation. Nat Rev Mol Cell Biol; 2003. 4:95-104. Martinon F, Tschopp J. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell 2004; 117:561-574. Ritter M, Gross O, Kays S, Ruland J, Nimmerjahn F, Saijo S, Tschopp J. Schistosoma mansoni triggers Dectin-2, which activates the Nlrp3 inflammasome and alters adaptive immune responses. Proc Natl Acad Sci USA 2010; 107:20459-20464. Hise AG, Tomalka J, Ganesan S, Patel K, Hall BA, Brown GD, Fitzgerald KA. An essential role for the NLRP3 inflammasome in host defense against the human fungal pathogen Candida albicans. Cell Host Microbe 2009; 5:487-497. Duncan JA, Gao X, Huang MT et al. Neisseria gonorrhoeae activates the proteinase cathepsin B to mediate the signaling activities of the NLRP3 and ASC-containing inflammasome. J Immunol 2009; 182:6460-6469. Mariathasan S, Weiss DS, Newton K, McBride J, O'Rourke K, Roose-Girma M, Lee WP et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 2006; 440:228232. 23

This article is protected by copyright. All rights reserved.

19.

Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008; 320:674-677. Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA, Latz E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 2008; 9:847-856. Johnson VJ, Yucesoy B, Luster MI. Prevention of IL-1 signaling attenuates airway hyperresponsiveness and inflammation in a murine model of toluene diisocyanate-induced asthma. J Allergy Clin Immunol 2005; 116:851-858. Schmitz N, Kurrer M, Kopf M. The IL-1 receptor 1 is critical for Th2 cell type airway immune responses in a mild but not in a more severe asthma model. Eur J Immunol 2003; 33:991-1000. Besnard AG, Guillou N, Tschopp J et al. NLRP3 inflammasome is required in murine asthma in the absence of aluminum adjuvant. Allergy 2011; 66:1047-1057. Allen IC, Jania CM, Wilson JE et al. Analysis of NLRP3 in the development of allergic airway disease in mice. J Immunol 2012; 188:2884-2893. Whitehead GS, Walker JK, Berman KG, Foster WM, Schwartz DA. Allergen-induced airway disease is mouse strain dependent. Am J Physiol Lung Cell Mol Physiol 2003; 285:L32-42. Kumar RK, Herbert C, Foster PS. The "classical" ovalbumin challenge model of asthma in mice. Curr Drug Targets 2008; 9:485-494. Zhu W, Gilmour MI. Comparison of allergic lung disease in three mouse strains after systemic or mucosal sensitization with ovalbumin antigen. Immunogenetics 2009; 61:199207. Swedin L, Ellis R, Kemi C, Ryrfeldt A, Inman M, Dahlén SE, Adner M. Comparison of aerosol and intranasal challenge in a mouse model of allergic airway inflammation and hyperresponsiveness. Int Arch Allergy Immunol 2010; 153:249-258. Li H, Nookala S, Re F. Aluminum hydroxide adjuvants activate caspase-1 and induce IL-1beta and IL-18 release. J Immunol 2007; 178:5271-5276. Conrad ML, Yildirim AO, Sonar SS et al. Comparison of adjuvant and adjuvant-free murine experimental asthma models. Clin Exp Allergy 2009; 39:1246-1254. Gross O, Yazdi AS, Thomas CJ et al. Inflammasome activators induce interleukin-1α secretion via distinct pathways with differential requirement for the protease function of caspase-1. Immunity 2012; 36:388-400. Stout-Delgado HW, Vaughan SE, Shirali AC, Jaramillo RJ, Harrod KS. Impaired NLRP3 inflammasome function in elderly mice during influenza infection is rescued by treatment with nigericin. J Immunol 2012; 188:2815-2824. Layland LE, Straubinger K, Ritter M, Loffredo-Verde E, Garn H, Sparwasser T, Prazeres da Costa C. Schistosoma mansoni-mediated suppression of allergic airway inflammation requires patency and Foxp3+ Treg cells. PLoS Negl Trop Dis 2013; 7(8):e2379. Master SL, Simon A, Aksentijevich I, Kastner DL. Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease. Annu Rev Immunol 2009; 27:621-668. Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity 2004; 20:319-325. Aganna E, Martinon F, Hawkins PN et al. Association of mutations in the NALP3/CIAS1/PYPAF1 gene with a broad phenotype including recurrent fever, cold sensitivity, sensorineural deafness, and AA amyloidosis. Arthritis Rheum 2002; 46:2445-2452. McDermott MF, Tschopp J. From inflammasomes to fevers, crystals and hypertension: how basic research explains inflammatory diseases. Trends Mol Med 2007; 13:381-388.

Accepted Article

519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567

20.

21.

22.

23. 24. 25. 26. 27.

28.

29. 30. 31.

32.

33.

34. 35.

36.

37.

24 This article is protected by copyright. All rights reserved.

38.

Menu P, Vince JE. The NLRP3 inflammasome in health and disease: the good, the bad and the ugly. Clin Exp Immunol 2011; 166:1-15. Cassel SL, Eisenbarth SC, Lyer SS et al. The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci USA 2008; 105:9035-9040. Delluc A, Limal N, Puéchal X, Francès C, Piette JC, Cacoub P. Efficacy of anakinra, an IL-1 receptor antagonist, in refractory Sweet syndrome. Ann Rheum Dis 2008; 67:278-279. Vounotrypidis P, Sakellariou GT, Zisopoulos D, Berberidis C. Refractory relapsing polychondritis: rapid and sustained response in the treatment with an IL-1 receptor antagonist (anakinra). Rheumatology 2006; 45:491-492. Caucig P, Teschner D, Dinges S, Maxeiner JH, Reuter S, Finotto S, Taube C, von Stebut E. Dual role of interleukin-1alpha in delayed-type hypersensitivity and airway hyperresponsiveness. Int Arch Allergy Immunol 2010; 152:303-312. Yamagata S, Tomita K, Sato R, Niwa A, Higashino H, Tohda Y. Interleukin-18-deficient mice exhibit diminished chronic inflammation and airway remodelling in ovalbumin-induced asthma model. Clin Exp Immunol 2008; 154:295-304. Lloyd CM. IL-33 family members and asthma - bridging innate and adaptive immune responses. Curr Opin Immunol 2010; 22:800-806. Kapur S, Bonk ME. Rilonacept (Arcalyst), an Interleukin-1 Trap for the Treatment of Cryopyrin-Associated Periodic Syndromes. P T 2009; 34:138-141. Lachmann HJ, Kone-Paut I, Kuemmerle-Deschner JB et al. Use of canakinumab in the cryopyrin-associated periodic syndrome. N Engl J Med 2009; 360:2416-2425. Sutton C, Brereton C, Keogh B, Mills KH, Lavelle EC. A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis. J Exp Med 2006; 203:1685-1691. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17producing human T helper cells. Nat Immunol 2007; 8:942-949. Guo L, Wei G, Zhu J, Liao W, Leonard W J, Zhao K, Paul W. IL-1 family members and STAT activators induce cytokine production by Th2, Th17, and Th1 cells. Proc Natl Acad Sci USA 2009; 106:13463-13468. Besnard AG, Togbe D, Couillin I et al. Inflammasome-IL-1-Th17 response in allergic lung inflammation. J Mol Cell Biol 2012; 4:3-10. Kudo M, Melton AC, Chen C et al. IL-17A produced by αβ T cells drives airway hyperresponsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat Med 2012; 18:547-554. Fogli LK, Sundrud MS, Goel S et al. T cell-derived IL-17 mediates epithelial changes in the airway and drives pulmonary neutrophilia. J Immunol 2013; 191:3100-3111 Park SJ, Lee YC. Interleukin-17 regulation: an attractive therapeutic approach for asthma. Respir Res 2010; 11:78 Idzko M, Hammad H, van Nimwegen M et al. Extracellular ATP triggers and maintains asthmatic airway inflammation by activating dendritic cells. Nat Med 2007; 13:913-919. Müller T, Vieira RP, Grimm M et al. A potential role for P2X7R in allergic airway inflammation in mice and humans. Am J Respir Cell Mol Biol 2011; 44:456-464. Gasse P, Riteau N, Charron S et al. Uric acid is a danger signal activating NALP3 inflammasome in lung injury inflammation and fibrosis. Am J Respir Crit Care Med 2009; 179:903-913. Riteau N, Gasse P, Fauconnier L et al. Extracellular ATP is a danger signal activating P2X7 receptor in lung inflammation and fibrosis. Am J Respir Crit Care Med 2010; 182:774-783.

Accepted Article

568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616

39. 40. 41.

42.

43.

44. 45. 46. 47.

48.

49.

50. 51.

52. 53. 54. 55. 56.

57.

25 This article is protected by copyright. All rights reserved.

Accepted Article

617 618

Figure Legends

619

Fig.

620

hyperresponsiveness. Groups of WT (PBS: n=11; OVA: n=27), ASC-/- (PBS: n=9; OVA: n=15) or

621

NLRP3-/- (PBS: n=6; OVA: n=20) mice were sensitized with either PBS or OVA (days 0, 7, 14)

622

and subsequently challenged with OVA (days 26-28) to induce allergic airway inflammation.

623

On day 31, (a) total leucocytes and the percentage of (b) eosinophils and (c) macrophages

624

were determined within the BAL. (a-c) Symbols show mean ± SD of individual mice from

625

three independent experiments and asterisks show significant differences between the

626

indicated brackets (***p

Functional relevance of NLRP3 inflammasome-mediated interleukin (IL)-1β during acute allergic airway inflammation.

Overall asthmatic symptoms can be controlled with diverse therapeutic agents. However, certain symptomatic individuals remain at risk for serious morb...
1MB Sizes 0 Downloads 3 Views