MCB Accepted Manuscript Posted Online 27 April 2015 Mol. Cell. Biol. doi:10.1128/MCB.01498-14 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Noncanonical effects of IRF9 in intestinal inflammation: more than type I and type III

2

interferons

3 4

Isabella Rauch*†/, Felix Rosebrock*†, Eva Hainzl#, Susanne Heider§, Andrea Majoros*,

5

Sebastian Wienerroither*, Birgit Strobl#, Silvia Stockinger#, Lukas Kenner§, Mathias Müller#

6

and Thomas Decker*

7 8

*Department of Microbiology and Immunobiology, Max F. Perutz Laboratories, University of

9

Vienna, Austria

10

§Ludwig Boltzmann Institute for Cancer Research, Medical University of Vienna, Vienna,

11

Austria

12

#Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna,

13

Austria

14 15



16

/

17

Pathogenesis, University of California, Berkeley, California, USA

these authors contributed equally

current affiliation: Department of Molecular & Cell Biology, Division of Immunology &

18 19 20

Running title: IFNλ and IRF9 in colitis

21 22 23

address correspondence to

24

Thomas Decker

25

Phone 00431427754605

26

Fax 0043142779546

27

[email protected]

28 29 30 31 32 33 34 1

35

Abstract

36

The ISGF3 transcription factor with its Stat1, Stat2 and IRF9 subunits is employed for

37

transcriptional responses downstream of receptors for type I interferons (IFN-I) that include

38

IFNα and IFNβ and type III interferons (IFN-III), also called IFNλ. Here we show in a

39

murine model of dextran sodium sulfate (DSS)-induced colitis that IRF9 deficiency protects

40

animals whereas the combined loss of IFN-I and IFN-III receptors worsens their condition.

41

We explain the different phenotypes by demonstrating a function of IRF9 in a noncanonical

42

transcriptional complex with Stat1, apart from IFN-I and IFN-III signaling. Together

43

Stat1/IRF9 produce a proinflammatory activity that overrides the benefits of the IFN-III

44

response on intestinal epithelial cells. Our results further suggest that the CXCL10 chemokine

45

gene is an important mediator of this proinflammatory activity. We thus establish IFNλ as

46

potentially anti-colitogenic cytokine and propose an important role for IRF9 as component of

47

noncanonical Stat complexes in the development of colitis.

48

2

49

Introduction

50

Interferons (IFN) are subdivided into three distinct types, type I IFN (IFN-I, mainly IFNα/β),

51

type II IFN (IFN-II or IFNγ) and type III IFN (IFN-III or IFNλ/IL28/IL29). Collectively IFN

52

are potent regulators of immune responses to pathogens (1, 2). In addition they contribute to

53

autoimmunity-related or other types of sterile inflammation (3-5). Biological responses to

54

IFN require transcription of a large number of IFN-induced genes (ISG), regulated by signal

55

transducers and activators of transcription (Stat) and interferon regulatory factors (IRF). In

56

their canonical signaling pathways the type I and type III IFN receptors stimulate the

57

assembly of the IFN-stimulated gene factor -3 (ISGF3) complex that contains tyrosine-

58

phosphorylated Stat 1 and 2 in association with IRF9, whereas the IFNγ receptor employs

59

Janus tyrosine kinases (Jaks) to produce the gamma-interferon-stimulated factor (GAF), a

60

homodimer of tyrosine-phosphorylated Stat1 (6). In addition, non-canonical complexes of

61

Stats1/2 and IRF9 can be formed in response to IFN-I or IFNγ signaling and contribute to

62

gene-selectivity of the transcriptional response (7-12). Stat1 homodimers bind to gamma

63

interferon activated sequences (GAS) in target promoters whereas ISGF3 binds to IFN

64

stimulated response elements (ISRE), which can be found in a large number of antimicrobial

65

and antiviral genes. Noncanonical complexes containing IRF9 would similarly be expected to

66

associate with the ISRE, consistent with the DNA-binding specificity of this subunit.

67

In keeping with the common deployment of ISGF3, immunological activities of IFN-I and

68

IFN-III appear to be very similar. However, the IFN-I receptor (IFNAR) is expressed on

69

virtually all nucleated cells, whereas IFN-III receptor expression in mice is restricted to

70

epithelial tissues (13). It is not entirely clear yet, whether IFN-I and III lead to completely

71

identical signaling outputs, as their receptors may differ in their ability to activate Stats other

72

than 1 and 2 or the MAPK pathway (14, 15).

73

Inflammatory bowel disease (IBD) is a health problem affecting a rising number of

74

individuals especially in the western world. A multi-step process initiates this chronic disease,

75

with a disturbance of the epithelial layer as an early event (16). Host factors as well as gut

76

microbiota have been implicated in the development and maintenance of IBD, with specific

77

innate and adaptive immune signaling pathways as well as specific bacterial species such as

78

members of the Enterobacteriaceae shown to be associated with the disease (17, 18). IFN-I

79

both reduce or enhance colitis in animal models, depending on the intensity of inflammation

80

and whether the acute or resolution phases are examined (5, 19). Clinical studies testing IFN-I

81

in the treatment of IBD support this view as they led to conflicting results concerning their

82

therapeutic potential (5, 20). Loss of IFNγ signaling exacerbated disease in some reported 3

83

animal studies (21, 22) while it was protective in others (23, 24). Blockade of IFNγ caused a

84

fairly subtle improvement of symptoms in human IBD patients (25, 26). IFN-III have not

85

been studied in the context of colitis. However, such studies appear of high priority in the

86

light of recent reports showing they control rotavirus infections by targeting the intestinal

87

epithelium (27). This study further demonstrated that polarized intestinal epithelial cells (IEC)

88

respond to IFN-III from the basolateral and apical but to IFN-I only from the apical side. The

89

authors suggested IFN-III might be more relevant for immune signaling in IEC than IFN-I.

90

Earlier in vitro studies showed that IFN-III decrease proliferation and induce antiviral proteins

91

in human IEC lines (28). Also, infection of IEC lines with various Gram-positive bacteria

92

induced production of IFN-III (29).

93

The aim of our study was to determine the effect of simultaneous elimination of IFN-I and

94

IFN-III responses by inducing colitis with the chemical dextran sodium sulfate (DSS) in mice

95

lacking the ISGF3 subunit IRF9. We report that mice were strongly protected from colitis

96

when deficient for IRF9. Surprisingly, the opposite effect was observed after combined

97

deletion of IFN-I and IFN-III receptors. Our results suggest that the procolitogenic activity of

98

IRF9 results from its participation in a noncanonical complex with Stat1 independently of

99

IFN-I and IFN-III receptor signaling. In addition, they support the notion that IFN-III prevent

100

damage of the gut mucosa after DSS treatment and might provide a novel therapeutic option.

101 102

4

103

Material and Methods

104 105

Mice, animal experiments. Animal experiments were approved by the University of

106

Veterinary Medicine Vienna institutional ethics committee and carried out in accordance with

107

protocols approved by the Austrian law (BMWF-66.006/002-II/10b/2010).

108

Mice lacking functional type III IFN receptors (IL28rα-/-) were provided by Bristol-Myers-

109

Squibb Company, New Jersey, USA. B6.A2G-Mx1 wild-type mice carrying intact Mx1

110

alleles (Mx1), B6.A2G- Mx1-IL28rα-/- mice lacking functional type III IFN receptors (Mx1-

111

IL28rα-/-), and B6.A2G-Mx1-IL28rα-/-Ifnar1-/- double-knockout mice (Mx1-IL28rα-/-Ifnar1-/-;

112

30) lacking functional receptors for both type I and type III IFN were provided by Peter

113

Stäheli (Freiburg, Germany). The mice were backcrossed to C57BL/6 mice as previously

114

described for Balb/c (31). C57BL/6N mice, Ifnar1-/-, Irf9-/- (32), Stat1-/- (33) and Stat2-/- mice

115

(34) backcrossed for more than 10 generations on a C57BL/6N background were housed in

116

the same specific pathogen free (SPF) facility under identical conditions according to

117

recommendations of the Federation of European Laboratory Animal Science Association,

118

additionally monitored for being norovirus negative. Colitis experiments where performed in

119

individually ventilated cage isolators.

120

To induce colitis, mice were provided with 2% dextran sodium sulfate (DSS, molecular

121

weight: 36–50 kDa, MP Biomedicals) in autoclaved drinking water ad libitum for 7 days,

122

after which they received DSS-free autoclaved drinking water for 2 days. Mice were weighed

123

daily and sacrificed on day 0 (untreated), 5 or 10 of the colitis protocol. Upon sacrifice the

124

intestine was removed and flushed with phosphate-buffered saline (pH 7.2). The colon was

125

halved lengthwise and one half was snap frozen while the other half was fixed in 2%

126

paraformaldehyde, prepared as a swiss role, and embedded in paraffin. Sections of whole

127

intestine were stained with H&E using a standard protocol and blind-scored by a pathologist

128

using an established method that evaluates inflammation severity, crypt damage,

129

inflammation extent and percent tissue involvement (35).

130

Immunohistochemistry. Tissue sections were rehydrated and boiled 20 min in a citric buffer

131

(pH 6) to unmask antigens. For proliferation assessment slides where incubated with rabbit

132

polyclonal anti-Ki67 antibody (NovoCastra) diluted 1:1000 and to evaluate apoptosis with

133

rabbit polyclonal anti-cleaved Caspase-3 antibody (Cell Signaling) diluted 1:200. Epitope

134

binding was revealed with peroxidase-conjugated secondary antibodies. Sections were

135

counterstained with hematoxylin and imaged on an epifluorescence microscope (Zeiss

136

AxioImager). IHC images were photographed and quantified with the HistoQUEST software 5

137

(TissueGnostics GmbH). At least 3 pictures per colon were taken and the percentage of

138

positive stained cells per number of nucleated cells was calculated.

139

RNA isolation, cDNA synthesis and qPCR. For RNA preparation colon tissue was

140

homogenized / BMDM were lysed in 700 µl RA1 buffer of the NucleoSpin II RNA isolation

141

kit (Macherey and Nagel) and processed according to the manufacturer’s protocol. cDNA was

142

prepared as described (36). PCRs were performed with 60°C annealing temperature on an

143

Eppendorf cycler. Relative expression of every sample was calculated to the housekeeping

144

gene OAZ1. Primer sequences are summarized in supplemental table 1.

145

Chromatin immunoprecipitation (ChIP) and ChIP-Seq. ChIP was performed as described

146

(37). 2µl (anti-Stat1, anti-Stat2, Santa Cruz) or 4ml (anti-IRF9, Santa Cruz) antiserum was

147

used per 50µg Chromatin. ChIP data were normalized to and expressed as a percentage of

148

input. Primers used for qPCR of the Cxcl10 enhancer regions (E1 and E2) and IFN response

149

region of Mx1 are listed in table S1. Next generation sequencing was carried out by the

150

Vienna Biocenter Campus Support Facilities (CSF). 5-10 ng of DNA precipitate was used for

151

the generation of sequencing libraries using the KAPA library preparation kit for Illumina

152

systems. Libraries were quantified with a Bioanalyzer dsDNA 1000 assay kit (Agilent) and a

153

QPCR NGS library quantification kit (KAPA). Cluster generation and sequencing was

154

performed with a HiSeq 2000 system with a read length of 100 nucleotides according to the

155

manufacturer's guidelines (Illumina).

156

In the re-ChIP experiments (12), the immune complexes were eluted by adding 10 mM

157

dithiothreitol (DTT) and incubating for 40min at room temperature. The samples were diluted

158

40-fold and reimmunoprecipitated.

159

Cell isolation and treatment. For IEC isolation, colons where washed in cold PBS

160

containing 50µg/ml gentamycin, cut into pieces and incubated for 10 min in 15mM

161

EDTA/PBS at 37°C shaking. After 1 g sedimentation to remove cell sheets, supernatants

162

where collected, spun down, washed in PBS and resuspended in buffer RA1 for RNA

163

isolation. For western blot, cells where resuspended in RPMI 1640 with or without 5ng/ml

164

IFNγ (eBioscience) for 15min and processed as described (36).

165

For lamina propria mononuclear cell (LPMC) isolation, colons where incubated in RPMI

166

1640 containing gentamycin and 5mM EDTA to fully remove IEC 3 times 15 min at 37°C

167

shaking. Remaining tissue pieces where washed in RPMI and incubated for 90 min in RPMI

168

containing 15mM HEPES and 0.2 mg/ml type VIII collagenase (Sigma). The digest was

169

filtered through a 70µm cell strainer, spun down, washed in PBS and resuspended in buffer

170

RA1. These fractions contained >50% LPMC. 6

171

Bone marrow-derived macrophages (BMDMs) were differentiated from bone marrow isolated

172

from femurs and tibias of 6-8 week old mice. Cells were cultured in DMEM (Sigma-Aldrich)

173

supplemented with 10 % of FCS (Sigma-Aldrich), 10 % of L929-cell conditioned medium as

174

a source of CSF-1 and 100 units/ml penicillin, 100 µg/ml streptomycin (Sigma-Aldrich) as

175

previously described (36). The culture contained >99 % of F4/80+ cells. BMDMs were

176

treated with 5 ng/ml of IFNγ (Affymetrix - eBioscience).

177

CXCL10 determination. Frozen tissue was homogenized in PBS containing proteinase

178

inhibitors (Phenylmethylsulfonylfluorid, Roche Complete Protease Inhibitor Cocktail), 1mM

179

vanadate and 1mM DTT. The homogenate was freeze-thawed twice. Total protein in the

180

supernatants was determined by BCA assay (Pierce) and CXCL10 levels where determined

181

using FlowCytomix kit (eBioscience) on 25µl supernatant according to the manufacturer’s

182

instructions. The CXCL10 amount was normalized to total protein.

183

Western blot. Organs or isolated IEC where homogenized in Frackelton buffer containing

184

protease and phosphatase inhibitors and centrifuged at 12000 g. The supernatant was used for

185

western blotting as described (36). Total Stat1 was detected using mAb to the Stat1 N

186

terminus (BD Biosciences) at a dilution of 1:1000. Tyrosine 701 phosphorylated Stat1 was

187

detected using a 1:1000 diluted antibody (Cell Signaling). As a loading control, the blot was

188

probed with Abs to the Erk1 and Erk2 kinases (panErk) (BD Transduction Laboratories) at a

189

dilution of 1:2000.

190

Statistics. Statistical analysis was done using the Mann Whitney U test, considering p

Noncanonical Effects of IRF9 in Intestinal Inflammation: More than Type I and Type III Interferons.

The interferon (IFN)-stimulated gene factor 3 (ISGF3) transcription factor with its Stat1, Stat2, and interferon regulatory factor 9 (IRF9) subunits i...
4MB Sizes 0 Downloads 10 Views