63
Tir-induced actin remodeling triggers expression of CXCL1 in enterocytes and neutrophil recruitment during Citrobacter rodentium infection Running title: Tir-mediated neutrophil recruitment Valerie F. Crepin * , Maryam Habibzay, Izabela Glegola-Madejska, Marianne Guenot, James W. Collins and Gad Frankel * MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College, London, UK For Correspondence: Valerie Crepin, CMBI, Flowers Building, Imperial College, London SW7 2AZ. Telephone: +44 20 75943070; Email: [email protected] & Telephone: +44 20 75945253; Email: [email protected] k 1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Tir-induced actin remodeling triggers expression of CXCL1 in enterocytes

and neutrophil recruitment during Citrobacter rodentium infection

Running title: Tir-mediated neutrophil recruitment

Valerie F. Crepin*, Maryam Habibzay, Izabela Glegola-Madejska, Marianne Guenot, James

W. Collins and Gad Frankel*

MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial

College, London, UK

For Correspondence: Valerie Crepin, CMBI, Flowers Building, Imperial College, London

SW7 2AZ. Telephone: +44 20 75943070; Email: [email protected] &

Telephone: +44 20 75945253; Email: [email protected] k

1

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

Page 2: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Abstract

The hallmarks of enteropathogenic Escherichia coli (EPEC) infection are formation

of attaching and effacing (A/E) lesions on mucosal surfaces and actin-rich pedestals

on cultured cells, both dependent on the type III secretion system effector Tir.

Following translocation into cultured cells and clustering by intimin, Tir Y474 is

phosphorylated leading to recruitment of Nck, activation of N-WASP and actin

polymerization via the Arp2/3 complex. A secondary, weak, actin polymerization

pathway is triggered via an NPY motif (Y454). Importantly, Y454 and Y474 play no

role in A/E lesion formation on mucosal surfaces following infection with the EPEC-

like mouse pathogen Citrobacter rodentium. In this study we investigated the roles of

Tir segments located upstream of Y451 and downstream of Y471 in C. rodentium

colonization and A/E lesion formation. We also tested the role Tir residues Y451 and

Y471 play in host immune responses to C. rodentium infection. We found that

deletion of amino acids 382-462 or 478-547 had no impact on the ability of Tir to

mediate A/E lesion formation, although deletion of amino acids 478-547 affected Tir

translocation. Examination of enterocytes isolated from infected mice revealed that a

C. rodentium expressing Tir_Y451A/Y471A recruited significantly less neutrophils to

the colon and triggered less colonic hyperplasia on day 14 post infection, compared to

infection with the wild type strain. Consistently, enterocytes isolated from mice

infected with C. rodentium expressing Tir_Y451A/Y471A expressed significantly

less CXCL1. These result show that Tir-induced actin remodeling plays a direct role

in modulation of immune responses to C. rodentium infection.

2

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

Page 3: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Introduction

Enteropathogenic Escherichia coli (EPEC) strains are important human pathogens

causing infantile diarrhea in low-income countries (1) Recently, the Global Enteric

Multicenter Study (GEMS), designed to detect the cause of paediatric diarrheal

disease in sub-Saharan Africa and south Asia, found that infection with typical EPEC

is associated with increased risk of fatality in infants aged 0-11 months (2).

Citrobacter rodentium is a mouse-specific pathogen, the etiological agent of

transmissible colonic hyperplasia, and a model EPEC microorganism, as both

pathogens share an infection strategy and virulence factors (3, 4). Host resistance to

C. rodentium infection is mediated by diverse T cell effector responses, including T

cells production of interferon-γ (IFNγ) (5, 6), interleukin 17A (IL17A) (7, 8) or IL22

(9). Expression of the pro-inflammatory cytokine IL-17A leads to recruitment of

neutrophils (10), and the anti-inflammatory cytokine IL-22 up-regulates expression of

antimicrobial peptides (such as REGIIIβ and REGIIIγ) in enterocytes (9, 11).

While colonizing the gut mucosa EPEC and C. rodentium induce attaching and

effacing (A/E) lesions. These are characterized by extensive remodeling of the gut

epithelium leading to elongation and effacement of the brush border (BB) microvilli,

intimate bacterial attachment to the enterocyte apical plasma membrane, accumulation

of polymerized actin and formation of elevated pedestal-like structures (4, 12).

Adhesion of EPEC (reviewed in (13)) and C. rodentium (14) to cultured cells triggers

actin polymerization under attached bacteria.

The ability to induce A/E lesions and actin polymerization is encoded within the locus

of enterocyte effacement (LEE) (15), which encodes a type III secretion system

(T3SS) (16), the outer membrane adhesin intimin (17), regulators, chaperones,

translocator and effector proteins (reviewed in (18)). Following initial cell attachment,

3

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

Page 4: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

EPEC and C. rodentium use their T3SS to inject LEE- and non-LEE-encoded

effectors that subvert multiple signaling pathways including apoptosis (the effectors

NleH and NleB), endosomal trafficking (EspG and EspI), Rho GTPases (EspH and

Map), innate immunity (NleC, NleD, NleE and NleF) and actin dynamics (Tir and

EspF) (reviewed in (13)). In particular, following translocation, Tir, which contains

two trans-membrane (TM) helixes, is integrated into the epithelial cell plasma

membrane in a hairpin loop topology (19, 20), exposing an extracellular central

domain that functions as an intimin receptor (21). Infection of cultured epithelial cells

has shown that binding of intimin induces clustering of Tir, which leads to

phosphorylation of a C-terminal tyrosine (20), Y474 in EPEC or Y471 in C.

rodentium, by redundant tyrosine kinases, including Src, Fyn and Abl (22, 23). These

in turn recruit Nck via its SH2 domain which activates the neural Wiskott–Aldrich

syndrome protein (N-WASP) via its SH3 domain. This leads to recruitment of the

Arp2/3 complex which triggers actin polymerization underneath the attached bacteria

(reviewed in (24)). Campellone and Leong (25) have shown that TirEPEC can promote

weak actin polymerization in an Nck-independent manner, involving the C-terminal

Tir tyrosine residue Y454 (or Y451 in C. rodentium), which is present in the context

of a conserved Asn-Pro-Tyr (NPY) motif (26). The NPY motif recruits the adaptor

protein insulin receptor tyrosine kinase substrate (IRTKS) and/or the insulin receptor

substrate protein of 53 kDa (IRSp53) (27, 28). In EPEC belonging to lineage 2, the

weak Tir NPY-mediated actin polymerization pathway is amplified by the bacterial

effector TccP2/EspFM, which also activates N-WASP (29).

Although it was widely believed that the Tir-induced actin signaling pathways

observed during infection of cultured cells were responsible for A/E lesion formation

on mucosal surfaces, Deng et al (30) provided some initial indications that this might

4

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

Page 5: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

not be the case as complementation of a tir C. rodentium mutant with a plasmid

encoding Tir Y471F restored A/E lesion formation in vivo. Moreover, infection of

human in vitro organ cultures (IVOC) with EPEC expressing Tir_Y474F or

Tir_Y454F/Y474F also resulted in A/E lesions (31). In addition, we have

subsequently reported that incorporation of Y451A and Y471A double substitutions

into C. rodentium chromosomal tir abrogated actin polymerization in cultured cells

but had no effect on the level of colonization and A/E lesion formation in the mouse

model (14). Importantly, Ritchie et al. (32) and Mallick et al. (33, 34) have shown

that of A/E pathogens expressing tir mutant unable to trigger actin polymerization in

vitro were attenuated in mucosal colonization in vivo. The aim of this study was to

further investigate the role of the C-terminus of Tir during C. rodentium infection in

vivo.

5

92

93

94

95

96

97

98

99

100

101

102

103

Page 6: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Material and methods

Bacterial strains and growth conditions

The bacterial strains, plasmids and primers used in this study are listed in Table 1.

Bacteria were grown in Luria–Bertani (LB) medium, M9 minimum media (35) or in

Dulbecco's modified Eagle's medium (DMEM) supplemented with kanamycin

(50 mg ml−1), ampicillin (100 mg ml−1) and nalidixic acid (50 mg ml−1) as required.

Introduction of site-directed tir mutants into the C. rodentium chromosome

We used the lambda red-based mutagenesis system (36) to introduce site-directed tir

alterations into the endogenous chromosomal tir gene, together with a kanamycin

cassette, in the tir-cesT intergenic region for 3′ mutagenesis as described before (14).

Deletion of the DNA segment encoding amino acids 478-547 (478-547) within the

tir gene was made by inverse-PCR on pICC433 (encoding Tir_Y451/Y471) and

pICC438 (encoding Tir_Y451A/Y471A) templates, previously described in (14),

using primer pair [Tir-P478DSV-stop-EcoRI-Rv] and [down-Tir-EcoRI-Fw]. The

inverse-PCR product was then digested with EcoRI and the aphT gene cloned into the

tir-cesT intergenic region to confer kanamycin resistance, resulting in plasmids

pICC1842 and pICC1843, respectively (Table 1).

Deletion of the DNA segment encoding amino acids 382-462 (382-462) within tir

was made by overlapping-PCR. C. rodentium genomic DNA was used to amplify tir

base pairs 664-1164 using primer pair [Tir-upTM1-Fw] and [Tir-down-TM2-Rv]. The

primer pair [Tir-down-TM2-Y471-Fw] and [Tir-EcoRI-Rv] was used to amplify tir

base pairs 1383-1644 from pICC433 (encoding Tir_Y471) and pICC438 (encoding

Tir_Y471A) templates. The two PCR fragments ([664-1164] and [1383-1644/Y471])

and ([664-1164] and [1383-1644/Y471A]) were PCR-overlapped, EcoRI digested and

6

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

Page 7: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

ligated to tir-cesT intergenic region (PCR-amplified as previously described in Crepin

et al 2010, using primers [EcoRI-(tir-cesT)-Fw/NcesT-Rv]. The ligated PCR product

was then re-amplified using primers [Tir-upTM1-Fw] and [NcesT-Rv] and cloned into

pGEMT vector. The constructs were digested with EcoRI and the aphT gene cloned

into the tir-cesT intergenic region to confer kanamycin resistance, resulting in

plasmids pICC1844 and pICC1845, respectively (Table 1). All plasmid derivatives

were checked by DNA sequencing.

The various deletions 478-547_Y451/Y471 and 478-547_Y451A/Y471A; 382-

462_Y471 and 382-462_Y471A were PCR amplified from pICC1842 and

pICC1843, using primers [NcesT-Rv /Tir-Up-YY-Fw] and from pICC1844 and

pICC1845, using primers [NcesT-Rv/Tir-upTM1-Fw], respectively. The PCR

products were electroporated into wild type C. rodentium expressing the lambda red

recombinase from pKD46 plasmid (36). The presence of the mutation was confirmed

by PCR and DNA sequencing amongst the kanamycin resistant clones.

Cell culture

Swiss 3T3 cell line was grown, maintained and infected with the different C.

rodentium strains, at a MOI of 50, as described (14). Cells were washed 6 h post

infection with phosphate-buffered saline (PBS), fixed for 15 min in 4%

paraformaldehyde, permeabilized with 0.1% Triton for 4 min. Phalloidin-Tetramethyl

Rhodamine Iso-Thiocyanate (TRITC) (Sigma) was used to stain F-actin, while

bacterial DNA was counterstained with Hoechst 33342. Tir was stained using rabbit

anti-Tir EHEC (37, 38), recognizing the N-terminal domain, and carbocyanine-2-

conjugated donkey anti-rabbit IgG (Jackson ImmunoResearch Europe) secondary

antibody. Images were acquired using an AxioCam MRm monochrome camera and

7

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

Page 8: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

processed using AxioVision (Carl Zeiss MicroImaging GmbH, Germany).

Tir translocation by C. rodentium was qualitatively assessed by calculating the ratio

between the number of total visible bacterial nuclei and the number of bacteria which

show Tir staining concentrated in a straight line at the interface between the

bacterium. A minimum of 100 bacteria were counted by experiment and the

experiment was performed twice.

Oral infection of mice

Pathogen-free female 18–20g C57Bl/6 mice were purchased from Charles River. All

animals were housed in individually HEPA-filtered cages with sterile bedding and

free access to sterilized food and water. All animal experiments were performed in

accordance with the Animals Scientific Procedures (Act 1986) and were approved by

the local Ethical Review Committee. Infections were performed twice using four to

eight mice per group. Mice inoculated with mock mutant and nonsense mutant strains

were included in every experiment. Mice inoculated with wild-type strain and

uninfected mice were included in parallel with mutant strains.

Mice were inoculated by oral gavage with 200μl of overnight LB-grown C. rodentium

suspension in PBS (≈ 5 × 109 cfu). The number of viable bacteria used as inoculum

was determined by retrospective plating onto LB agar containing antibiotics. Stool

samples were recovered aseptically at various time points after inoculation and the

number of viable bacteria per gram of stool was determined by plating onto LB agar

(39). At day 7 and 14 post inoculum, the mice were culled and the colonic tissues

were collected for further analyses.

Sample collection and colonic crypt hyperplasia measurement

8

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

Page 9: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Segments of the terminal colon (0.5cm) of each mouse were collected, flushed and

fixed in 10% neutral buffered formalin. Formalin fixed tissues were then processed,

paraffin-embedded, sectioned at 5μm and stained with haematoxylin and eosin (H&E)

using standard techniques. H&E stained tissues were evaluated for colonic crypt

hyperplasia microscopically without knowledge of the treatment condition used in the

study and the length of at least 20 well-oriented crypts from each section from all of

the mice per treatment group (n=4-6) were evaluated. H&E stained tissues were

imaged with an Axio Lab.A1 microscope (Carl Zeiss MicroImaging GmbH,

Germany), images were acquired using an Axio Cam ERc5s colour camera, and

computer-processed using AxioVision (Carl Zeiss MicroImaging GmbH, Germany).

Additional colonic segments were embedded in optimal cutting temperature (OCT)

medium (Raymond A Lamb Limited, UK) and frozen in dry-ice/ethanol slush for

further cryo-sectioning. Cryo-sections were then fixed in 3%-paraformaldehyde

(PFA) in PBS as previously described (38, 40) and immuno-stained using primary

antibodies at a 1/50 dilution, chicken anti-intimin (14), Ly-6G (RB6-8C5, Santa Cruz)

and E-Cadherin (CD324, BD Biosciences). Secondary antibodies were used at 1/100

dilution, Cy3 (103-165-175, Jackson Immunoresearch), Alexa 488 (712-546-150,

Jackson Immunoresearch) and Cy5 (715-175-150, Jackson Immunoresearch),

respectively. Images were acquired using an AxioCam MRm monochrome camera

and processed using AxioVision (Carl Zeiss MicroImaging GmbH, Germany).

Extraction of enterocytes and immunostaining

Four cm segment of the terminal colon was cut longitudinally and placed in 4ml

enterocyte dissociation buffer (1x Hanks’ balanced salt solution without Mg & Ca

9

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

Page 10: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

containing 10mM HEPES, 1mM EDTA and 5μl/ml 2-mercaptoethanol) and

incubated at 37C, shaking, for 40 min. Left over tissue was removed by

centrifugation (1900g for 5 min) before the samples for each group were pooled

together and fixed with 1% formaldehyde. Fixed enterocytes (CD45-CD326+) were

analyzed for purity by flow cytometry using leukocyte marker CD45 and epithelial

cell marker CD326 (EpCAM). For immunofluorescence staining, fixed enterocytes

were permeabilized with 0.1% Triton and blocked with 1% bovine serum albumin in

PBS. Enterocytes were stained with polyclonal rabbit anti-Tir EHEC (GB1320,

SK1786) for 20 mins followed by 30 min incubation with secondary donkey anti-

rabbit IgG (H+L) Alexa488, phalloidin-TRITC (Sigma, P1951) was used for actin and

DAPI (Invitrogen, D3571) to visualize the nucleus. Tir staining was visualized with

an Axio Imager M1 microscope (Carl Zeiss MicroImaging GmbH, Germany), images

were acquired using an AxioCam MRm monochrome camera, and computer-

processed using AxioVision (Carl Zeiss MicroImaging GmbH, Germany).

Isolation of mRNA and Q-RT-PCR

mRNA of enterocytes was isolated using an RNeasy minikit according to the

manufacturer’s instructions (Qiagen). Samples were treated with RQ1 DNase-1

(Promega) at 37°C for 10 min, followed by 15 min at 72°C. Reverse transcription

(RT)-PCR was carried out by adding RT M-MLV (Promega M170B), RT buffer

(Promega), random primers (Promega), RNasin (Promega), dNTP (10mM) and RNase

free water to the DNase treated RNA extract, incubated at 37°C for 1 hour followed

by 10 min at 72°C and cooled samples were stored at -20°C. CXCL1 (KC) and

glyceraldehyde-3-phosphate dehydrogenase (GAPDH) cDNAs were amplified with

primer pairs mCXCL1-F / mCXCL1-R and mGAPDH-F / mGAPDH-R (Table 1), by

10

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

Page 11: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Q-RT-PCR using the 7300 Applied Biosystems instrument under standard cycle

conditions for Fast SYBR Green master mix. Changes in gene expression levels were

analyzed relative to the control levels (PBS samples), with GAPDH as a standard,

using the ΔΔCT method.

Sample collection for flow cytometry

Four cm segment of the terminal colon was cut, opened longitudinally and rinsed in

sterile PBS and placed in 4ml of RPMI-1640 supplemented with 10% fetal bovine

serum (FBS), penicillin/streptomycin (P/S), GlutaMAX, DNase (Roche,

10104159001) and liberase (Roche, 540112001) in a C-Mac tube (Miltenyi Biotec)

followed by tissue dissociation using gentleMACS dissociator (Miltenyi Biotec). The

tissue was homogenized using ‘intestine’ setting followed by incubation at 37°C, 5%

CO2 for 30 min in a shaking incubator and a final dissociation step was performed

using ‘Lung 2’ setting. The digested preparation was disrupted to a single cell

suspension by passage through a 70μm sieve (BD labware/falcon, USA Cat. No:

352350) and suspended in RPMI-1640 supplemented with 10% FBS and P/S at 0.5-

1x106 cells/ml.

11

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

Page 12: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Extracellular antigen analysis

Cells were stained for surface markers as indicated in PBS containing 1% bovine

serum albumin with 0.5% sodium azide (PBA) for 30 min at 4°C and fixed with IC

fixation buffer (eBioscience). Prior to primary antibody staining, all cells were

blocked for Fc receptors (FcR) using mouse FcR blocking reagent (Miltenyi biotec)

for 10 min at 4°C. Antibodies were purchased from BD Pharmingen or eBioscience.

Data acquired on a BD Fortessa III and 20,000 lymphocytes or myeloid events were

analyzed with the FlowJO (Tress star) analysis program. Data is shown as a

percentage of myeloid or lymphocyte gates. Myeloid and lymphocyte gates are

determined by their position on the forward and side scatter plots generated by the

cytometer. Fluorescence minus one (FMO) control was included for each fluorescent

marker, the expression of a particular marker was calculated by subtracting FMO

fluorescence values from fluorescent antibody levels.

Total live cells were assessed by trypan blue exclusion. Forward and side scatter gates

on Flowjo software were used to gate myeloid cells, the percentage of this gate was

used to determine total number of myeloid cells. The myeloid cells gate was further

analysed to finalise the percentage of neutrophils (CD11b+Ly6G+) and establish the

total number of neutrophils in each mouse colon.

Mouse intestinal in vitro organ cultures

Mouse intestinal in vitro organ culture (mIVOC) model was used to assess A/E lesion

formation caused by C. rodentium expressing Tir Y451A/Y471A/478-547 as

described by Girard et al 2009 (41). Briefly, segments from the terminal colon were

inoculated with 50l of the appropriate overnight bacterial culture, corresponding to

approximately 107 colony forming units (cfu), and incubated at 37°C in 5% CO2

12

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

Page 13: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

atmosphere on a see-saw rocker (18 cycle min-1) for 8 h. Explants were gently rinsed

with PBS and fixed in 2.5% glutaraldehyde for electron microscopy analysis.

13

272

273

274

Page 14: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Electron microscopy

Additional explants/tissue cultured cell samples were processed for electron

microscopy, as previously described (38). Samples for scanning electron microscopy

(SEM) were examined at an accelerating voltage of 25 kV using a JEOL JSM-5300

scanning electron microscope (JEOL (UK) Ltd., Herts, United Kingdom). Samples for

transmission electron microscopy (TEM) were observed using a Phillips 201

transmission electron microscope at an accelerating voltage of 60kV (Philips, United

Kingdom).

Statistical analysis

Results are presented as a line plot (colonization) with the mean and its standard

deviation. The non-parametric Mann–Whitney test and the non-parametric Kruskal–

Wallis test with Bonferroni's corrected a posteriori comparisons were used to conduct

pairwise and global statistical analysis, respectively, using commercially available

GraphPad InStat v3.06 software (GraphPad Software, San Diego, CA, USA). Mann-

Whitney compared to PBS controls (or as indicated in the figure) was used for data

obtained by flow cytometry using GraphPad Prism software. A P = 0.05 was

considered significant.

14

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

Page 15: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Results

Construction of the 3’ tir chromosomal deletion mutants

Previously, using a method that allows expression of tir mutants from the C.

rodentium chromosome, we have introduced point mutations which have shown that

A/E lesion formation in vivo is independent of Tir residues Y451 and Y471 (14). In

this study, we used this technique to introduce deletions at the 3’ end of tir (Fig. 1).

While inserting a kanamycin cassette coupled to the mutated tir into the tir-cesT

intergenic region, we deleted Tir residues 382-462 (Tir382-462), removing the 80

amino acids downstream of the distal TM helix and Tir residues 478-547 (Tir478-

547), removing the entire segment downstream of Y471 phosphorylation site (Fig. 1).

Tir382-462 was made in the context of either Y471 or Y471A (Tir382-462_Y471

and Tir382-462_Y471A), while Tir478-547 was made in the context of either

Y451/Y471 or Y451A/Y471A (Tir478-547 _Y451/Y471 and Tir478-

547_Y451A/Y471A). As a positive control we used a mock mutant tir-cesT (TirC-

ctrl) in which the kanamycin cassettes was introduced into the intergenic region which

did not affect the tir coding sequence and as a negative control, a nonsense codon

introduced at Tir position 33 (Tir1−33stop) (14). Growth curves in minimal and rich

media confirmed that the mutants and parental wild-type strains had identical growth

rates (data not shown).

Testing the carboxy terminal Tir deletions during infection of cultured cells

We characterized the behavior of the Tir derivatives in vitro, following infection of

Swiss 3T3 fibroblast cells with the C. rodentium mutants as described before (14).

This revealed that, as expected, C. rodentium expressing either Tir382-462_Y471A

15

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

Page 16: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

or Tir478-547_Y451A/Y471A failed to induce actin polymerization (data not

shown). In contrast, infection of Swiss 3T3 cells with C. rodentium expressing

Tir382-462_Y471 or Tir478-547_Y451/Y471 revealed robust actin polymerization

under all the adherent bacteria that show Tir translocation (Fig. 2A). Importantly,

while Tir382-462 was translocated as efficiently as the wild type control Tir,

Tir478-547 translocated in low efficiency, with Tir staining seen within 50% of the

adherent bacteria (Fig. 2B). Interestingly, C. rodentium expressing Tir478-

547_Y451/Y471 produced longer pedestals. These results suggest that Tir residues

382-462 are dispensable for Tir translocation and actin polymerization, while the

carboxy terminus of Tir plays a role in translocation and hence indirectly in the

efficiency (both in terms of frequency and length) of actin pedestals formed in vitro.

Testing the carboxy terminal Tir deletions during mouse infection

We next investigated the impact of the C-terminal Tir deletions on colonization of C.

rodentium in vivo by enumerating colony-forming units per gram of stools (cfu g-1)

collected daily following oral inoculation of C57BL/6 mice for 8 days. This has

shown that C. rodentium expressing either Tir382-462_Y471 or Tir382-

462_Y471A colonized the mice similarly to the control strain expressing TirC-ctrl

(Fig. 3A) (variations between groups seen on day 2 post infection are common during

C. rodentium infection and has not biological relevance; the differences seen on day 3

post infection are not significant). Transmission electron microscopy (TEM) revealed

typical A/E lesions in colons infected with C. rodentium expressing either Tir382-

462_Y471 or Tir382-462_Y471A (Fig. 3B). In contrast, C. rodentium expressing

Tir1−33stop was rapidly cleared and failed to initiate an infection, reaching

background level as soon as day 3 post infection (Fig. 3A). These results show that

16

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

Page 17: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Tir segment 382-462 is dispensable for colonization and that C. rodentium expressing

Tir382-462_Y471 or Tir382-462_Y471A are capable of forming A/E lesions in

vivo.

We then tested the phenotype of C. rodentium expressing Tir478-547. Consistent

with the in vitro data, this mutant exhibited an intermediate phenotype, reaching a

colonization level of 106/g of stool on day 3 post infection, which persisted in this

level until day 8 (Fig. 3A). The level of colonization was 100 fold lower than that

seen in mice infected with C. rodentium expressing TirC-ctrl or Tir382-462 but 104

fold higher than mice infected with Tir1−33stop. We observed no difference in

colonization between mice infected with Tir478-547_Y451/Y471 or Tir478-

547_Y451A/Y471A (Fig. 3A). However, as colonization was below the detection

level of TEM, we performed a mouse IVOC infection using C. rodentium expressing

Tir78-547_Y451A-Y471A. Examination of the samples by SEM revealed A/E

lesions, similar to those formed by the wild type strains, suggesting that Tir segment

478-547 is dispensable for A/E lesion formation on mucosal surfaces but does play a

role in colonization (Fig. 3B), probably due to its role in Tir translocation.

Tir-induces actin polymerization on enterocytes in vivo

As the results thus far have shown that C. rodentium expressing Tir deletions

(residues 382-462 and 478-547) and substitutions (Y451 and Y471) was able to form

A/E lesions, we tested whether Tir-induced actin polymerization on enterocytes in

vivo. For this, mice were infected with wild type C. rodentium or C. rodentium

expressing Tir_Y451A/Y471A (Fig. 1) and enterocytes were isolated at the peak of

colonization at day 7 post infection. Enterocytes isolated from naïve mice as a control

17

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

Page 18: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

exhibited a typical ‘crown’ staining pattern of the BB microvilli (Fig. 4). Enterocytes

isolated from infected mice showed good level of C. rodentium adhesion, with

multiple bacteria on individual enterocytes. Tir was detected underneath both attached

wild type C. rodentium and C. rodentium expressing Tir_Y451A/Y471A (Fig. 4).

However, while intense actin staining was seen at the site of wild type C. rodentium

infection, enterocytes infected with C. rodentium expressing Tir_Y451A/Y471A

exhibited mainly weak actin polymerization at the site of bacterial attachment. This

suggests that Tir induces actin polymerization on enterocyte in a process involving the

tyrosine residues.

Recruitment of immune cells to the C. rodentium infection site

As actin remodeling during infection can trigger immune responses (42), we next

investigated if Tir Y451 and Y471 modulate host immune responses. For this, groups

of 4-6 mice were infected with wild type and mutant C. rodentium and recruitment of

immune cells were analyzed by flow cytometry of homogenized colons at days 7

(peak of colonization) and day 14 (peak of pathology) post infection. All the tested

strains were shed at equivalent levels (Fig. 5A). The flow cytometry analysis has

shown that in comparison to the PBS mock-infected control mice (baseline readout),

infection with either the wild type or mutant C. rodentium resulted in equivalent

recruitment of macrophages, CD4+ T cells, and B cells on day 7 (data not shown) and

14 (Fig. 5B-D) post infection. In contrast, significantly less neutrophils were recruited

to the colon following infection with C. rodentium expressing Tir_Y451A/Y471A

compared with the positive control mice infected with C. rodentium expressing wild

type Tir, or mice infected with the single Tir tyrosine mutants Y451A or Y471A (Fig.

5E-F). Similarly, although shed at equivalent levels (Fig. 5A), neutrophils recruitment

18

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

Page 19: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

was observed following infection with C. rodentium expressing Tir382-462_Y471

but not after infection with C. rodentium expressing Tir382-462_Y471A (Fig. 5G-

H).

As C. rodentium and Tir mainly interact with enterocytes, we next determined if the

Tir tyrosine residues play a role in expression of pro-inflammatory chemokines. For

this we isolated colonic enterocytes from mice infected with the different C.

rodentium strains. The purity of the enterocytes preparation was confirmed by flow

cytometry analysis following staining with the leukocyte marker CD45 and epithelial

cell marker CD326 (EpCAM), revealing low level of contamination (Fig. 6A). Using

the purified enterocytes in Q-RT-PCR for the chemokines CXCL1 (KC) and CXCL2

(MIP2-alpha) revealed reduced expression in mice infected with C. rodentium

expressing TirY451A-Y471A (Fig. 6B). The attenuated inflammatory responses

triggered by C. rodentium expressing Tir_Y451A/Y471A were mirrored by a

significantly reduced colonic hyperplasia (Fig. 7A and B) as well as neutrophil

staining with Ly-6G antibodies (Fig. 7C). These results reveal a novel in vivo role for

the Tir tyrosine residues TirY451 and Y471, which are also implicated in actin

polymerization during C. rodentium infection (Fig. 4).

19

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

Page 20: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Discussion

In this study we found that C. rodentium expressing Tir382-462 colonized the

mouse gastrointestinal tract and produced A/E lesion whether in the context of Y471

or Y471A. This result shows that amino acids 382-462 are dispensable for Tir

activity. We also tested a chromosomal deletion of Tir residues 478-547. This has

shown that C. rodentium expressing either Tir478-547_Y451/Y471 or Tir478-

547_Y451A/Y471A behaved similarly, showing an intermediate colonization level

between the wild type and the Tir1-33stop strains. The fact that C. rodentium

expressing either Tir478-547_Y451/Y471 or Tir478-547_Y451A/Y471A showed

104 fold higher colonization level than mice infected with Tir1-33stop suggests that

Tir is at least partially active. Due to the low level of colonization we were unable to

determine if these strains could form A/E lesions in vivo using TEM but we could

confirm A/E lesions by SEM. A previous report demonstrated that a 6 amino acid

sequence (TYARLA) at position 519-524 within the carboxy-terminal region was

required for efficient secretion and translocation, but not for stability, of Tir-EHEC

(43). The carboxy terminus of Tir C. rodentium contain an equivalent 6 amino acid

sequence (TYALLA), which is consistent with the low translocation efficiency, seen

by immuno-fluorescence staining following infection of Swiss 3T3 cells with C.

rodentium expressing Tir478-547_Y451/471, and the intermediate in vivo phenotype

of this strain.

Immunostaining of enterocytes isolated from naïve mice revealed good preservation

of the BB microvilli. In contrast, individual enterocytes isolated from infected mice

were covered with adherent C. rodentium and exhibited effaced BB microvilli. Tir

was detected at equivalent intensity at the site of bacterial attachment, whether in the

context of wild type Tir or Tir_Y451A/Y471A. Importantly, actin staining was

20

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

Page 21: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

considerably brighter under attached C. rodentium expressing wild type Tir. This

could potentially explain the competitive advantage of wild type C. rodentium over C.

rodentium expressing Tir_Y451A/Y471A during mixed infection (14). However, it is

important to note that while, as expected, no Nck was recruited to C. rodentium

expressing Tir_Y451A/Y471A in vivo (14) or to human intestinal biopsies infected

with EPEC expressing the Tir mutant (31), N-WASP was detected underneath the

attached mutant strains, which could explain the faint actin polymerization seen in the

enterocytes isolated from mice infected with C. rodentium expressing

Tir_Y451A/Y471A. Taken together these data suggest the existence of as yet

undetermined Tir actin polymerization pathway in mucosal surfaces.

If the Tir tyrosine residues do not play a role in A/E lesion formation, the question

remains, what function do they have during infection of mucosal surfaces? As EPEC

and C. rodentium interact intermediately with enterocytes, we hypothesized that Tir-

induced actin polymerization might contribute to signaling to the underlying immune

system. To test this, we infected mice with C. rodentium expressing wild type Tir and

Tir tyrosine mutants and compared recruitment of immune cells in homogenized

colons. This revealed no difference in recruitment of macrophages, T cells or B cells

at either 7 or 14 days post infection. In contrast, significantly reduced level of

recruited neutrophils was seen at day 14 following infection with C. rodentium

expressing Tir_Y451A/Y471A or Tir382-462_Y471A, compared to infection with

C. rodentium expressing wild type Tir or Tir382-462_Y471. Moreover, mice

infected with C. rodentium expressing Tir_Y451A/Y471A presented significantly

reduced levels of colonic hyperplasia. Importantly, infection with C. rodentium

expressing single tyrosine Tir mutant (Y451A or Y471A), resulted in neutrophil

recruitment equivalent to that seen following infection with wild type C. rodentium.

21

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

Page 22: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

This phenotype was mirrored following testing for CXCL1 and CXCL2 expression by

Q-RT-PCR on enterocytes purified from C. rodentium infected mice. Similarly, (44)

reported that following EPEC infection both Tir Y454 and Y474 are needed for

efficient nuclear translocation of the transcription factor serum response factor (SRF)

co-factor MAL and transcription of SRF target genes. CXCL1 and CXCL2 signal via

CXCR2 to activate neutrophils and subsequently promote mucosal influx of

neutrophils (45). Importantly, although neutrophils contribute to host defense against

infection (10), wild type C. rodentium and C. rodentium expressing mutant Tir

colonized at equivalent levels. These results suggest that while Tir contains redundant

mechanisms leading to neutrophil recruitment, each relying of one of the two

tyrosines, the host immune response can compensate for the lack of neutrophils late

during infection and clear the pathogen.

Tir EPEC and C. rodentium have further two distal tyrosines (Y480_Y508 and

Y483_Y511, respectively), which comprise an immunoreceptor tyrosine-based

inhibition motif (ITIM). Smith et al. (46) reported that following EPEC infection Tir

residues Y483 and Y511 recruit the host inositol phosphatase SHIP2. Moreover, the

pedestals formed by EPEC expressing Tir Y483F_Y511F were significantly longer

than those formed by EPEC expressing wild type Tir, which is consistent with the

longer pedestals we observed following infection with C. rodentium expressing

Tir478-547_Y451/Y471, which lacks residues Y480_Y508. Recently Yan et al have

shown that infection with EPEC expressing Tir Y483F_Y511F resulted in elevated

levels of IL6 and TNF mRNA in splenic cells and enhanced bacterial clearance (47).

They have shown that phosphorylation of the Tir ITIM leads to the recruitment of

both SHP1 and SHP2 and inhibition of TRAF6 autoubiquitination, which helps the

bacteria to suppress and evade the host innate immune response (47, 48). This feature

22

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

478

479

480

481

482

Page 23: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

of Tir could provide an alternative explanation for why C. rodentium expressing Tir

Tir478-547_Y451/Y471 or Tir478-547_Y451A/Y471A did not colonize the colon

at a wild type level.

Subversion of the actin cytoskeleton by bacterial virulence factors has been shown to

be an important mediator of immune signaling. For example, the Salmonella Rho

GTPase GEF SopE has been shown to activate the pattern recognition receptors

(PRRs) NOD1 (49), while NOD2 is regulated by Rac1 (50). NOD1 and NOD2, could

be found at the plasma membrane in association with F-actin, which is needed for

downstream activation of NF-kB signaling (42, 51). Recently, Bielig et al have shown

that the actin depolymerization factors (ADF)/cofilin phosphatase SSH1 is an

essential component of the NOD1 pathway, which plays a role in activation of NF-kB

and cell responses to Shigella infection (52). Indeed, depletion of SSH1 mRNA

resulted in reduced production of IL8 and IL6 following infection of HeLa cells with

S. flexneri strain M90T. Consistently, our data show that sensing Tir-induced actin

remodeling triggers host responses to C. rodentium infection.

C. rodentium translocates multiple effectors that contribute to coordinated

cytoskeleton remodeling (including Map that activates Cdc42 and Rac1 (53), EspM

that activates RhoA (54), EspT that activates Rac1 (55) and EspJ that inhibits Src

kinases (56) which play a role in Tir tyrosine phosphorylation) and subversion of

innate immune responses, including NF-kB (e.g. NleC, NleD, NleE, NleB and NleF

(13, 57-62)). Importantly, the difference in neutrophil recruitment seen at day 14

between wild type C. rodentium and C. rodentium expressing Tir_Y451A/Y471A is

at the time when both infections are close to being cleared. Future studies will aim at

unraveling the mechanism by which Tir induces expression of CXCL1 and CXCL2 in

enterocytes late during infection in the broader context of the other type III secreted

23

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

Page 24: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

effectors that modulate inflammatory responses.

24

508

509

Page 25: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Acknowledgements

This study was supported by a grant from the Wellcome trust and the BBSRC.

25

510

511

512

Page 26: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

References

1. Chen HD, Frankel G. 2005. Enteropathogenic Escherichia coli: unravelling

pathogenesis. FEMS Microbiol Rev 29:83-98.

2. Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH,

Panchalingam S, Wu Y, Sow SO, Sur D, Breiman RF, Faruque AS, Zaidi

AK, Saha D, Alonso PL, Tamboura B, Sanogo D, Onwuchekwa U, Manna

B, Ramamurthy T, Kanungo S, Ochieng JB, Omore R, Oundo JO,

Hossain A, Das SK, Ahmed S, Qureshi S, Quadri F, Adegbola RA,

Antonio M, Hossain MJ, Akinsola A, Mandomando I, Nhampossa T,

Acacio S, Biswas K, O'Reilly CE, Mintz ED, Berkeley LY, Muhsen K,

Sommerfelt H, Robins-Browne RM, Levine MM. 2013. Burden and

aetiology of diarrhoeal disease in infants and young children in developing

countries (the Global Enteric Multicenter Study, GEMS): a prospective, case-

control study. Lancet 382:209-222.

3. Collins JW, Keeney KM, Crepin VF, Rathinam VA, Fitzgerald KA,

Finlay BB, Frankel G. 2014. Citrobacter rodentium: infection, inflammation

and the microbiota. Nat Rev Microbiol 12:612-623.

4. Mundy R, MacDonald TT, Dougan G, Frankel G, Wiles S. 2005.

Citrobacter rodentium of mice and man. Cell Microbiol 7:1697-1706.

5. Higgins LM, Frankel G, Douce G, Dougan G, MacDonald TT. 1999.

Citrobacter rodentium infection in mice elicits a mucosal Th1 cytokine

response and lesions similar to those in murine inflammatory bowel disease.

Infect Immun 67:3031-3039.

26

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

Page 27: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

6. Shiomi H, Masuda A, Nishiumi S, Nishida M, Takagawa T, Shiomi Y,

Kutsumi H, Blumberg RS, Azuma T, Yoshida M. 2010. Gamma interferon

produced by antigen-specific CD4+ T cells regulates the mucosal immune

responses to Citrobacter rodentium infection. Infect Immun 78:2653-2666.

7. Geddes K, Rubino SJ, Magalhaes JG, Streutker C, Le Bourhis L, Cho

JH, Robertson SJ, Kim CJ, Kaul R, Philpott DJ, Girardin SE. 2011.

Identification of an innate T helper type 17 response to intestinal bacterial

pathogens. Nat Med 17:837-844.

8. Cua DJ, Tato CM. 2010. Innate IL-17-producing cells: the sentinels of the

immune system. Nat Rev Immunol 10:479-489.

9. Zheng Y, Valdez PA, Danilenko DM, Hu Y, Sa SM, Gong Q, Abbas AR,

Modrusan Z, Ghilardi N, de Sauvage FJ, Ouyang W. 2008. Interleukin-22

mediates early host defense against attaching and effacing bacterial pathogens.

Nature Medicine 14:282-289.

10. Spehlmann ME, Dann SM, Hruz P, Hanson E, McCole DF, Eckmann L.

2009. CXCR2-dependent mucosal neutrophil influx protects against colitis-

associated diarrhea caused by an attaching/effacing lesion-forming bacterial

pathogen. J Immunol 183:3332-3343.

11. Manta C, Heupel E, Radulovic K, Rossini V, Garbi N, Riedel CU, Niess

JH. 2013. CX(3)CR1(+) macrophages support IL-22 production by innate

lymphoid cells during infection with Citrobacter rodentium. Mucosal

Immunol 6:177-188.

12. Knutton S, Lloyd DR, McNeish AS. 1987. Adhesion of enteropathogenic

Escherichia coli to human intestinal enterocytes and cultured human intestinal

mucosa. Infect Immun 55:69-77.

27

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

Page 28: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

13. Wong AR, Pearson JS, Bright MD, Munera D, Robinson KS, Lee SF,

Frankel G, Hartland EL. 2011. Enteropathogenic and enterohaemorrhagic

Escherichia coli: even more subversive elements. Mol Microbiol 80:1420-

1438.

14. Crepin VF, Girard F, Schuller S, Phillips AD, Mousnier A, Frankel G.

2010. Dissecting the role of the Tir:Nck and Tir:IRTKS/IRSp53 signalling

pathways in vivo. Mol Microbiol 75:308-323.

15. McDaniel TK, Jarvis KG, Donnenberg MS, Kaper JB. 1995. A genetic

locus of enterocyte effacement conserved among diverse enterobacterial

pathogens. Proc Natl Acad Sci USA 92:1664-1668.

16. Jarvis KG, Girón JA, Jerse AE, McDaniel TK, Donnenberg MS, Kaper

JB. 1995. Enteropathogenic Escherichia coli contains a putative type III

secretion system necessary for the export of proteins involved in attaching-

effacing lesions formation. Proc Natl Acad Sci USA 92:7996-8000.

17. Jerse AE, Yu J, Tall BD, Kaper JB. 1990. A genetic locus of

enteropathogenic Escherichia coli necessary for the production of attaching

and effacing lesions on tissue culture cells. Proc Natl Acad Sci USA 87:7839-

7843.

18. Garmendia J, Frankel G, Crepin VF. 2005. Enteropathogenic and

enterohemorrhagic Escherichia coli infections: translocation, translocation,

translocation. Infect Immun 73:2573-2585.

19. Hartland EL, Batchelor M, Delahay RM, Hale C, Matthews S, Dougan G,

Knutton S, Connerton I, Frankel G. 1999. Binding of intimin from

enteropathogenic Escherichia coli to Tir and to host cells. Mol Microbiol

32:151-158.

28

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

Page 29: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

20. Kenny B. 1999. Phosphorylation of tyrosine 474 of the enteropathogenic

Escherichia coli (EPEC) Tir receptor molecule is essential for actin nucleating

activity and is preceded by additional host modifications. Mol Microbiol

31:1229-1241.

21. Kenny B, DeVinney R, Stein M, Reinscheid DJ, Frey EA, Finlay BB.

1997. Enteropathogenic E. coli (EPEC) transfers its receptor for intimate

adherence into mammalian cells. Cell 91:511-520.

22. Swimm A, Bommarius B, Li Y, Cheng D, Reeves P, Sherman M, Veach D,

Bornmann W, Kalman D. 2004. Enteropathogenic Escherichia coli use

redundant tyrosine kinases to form actin pedestals. Mol Biol Cell 15:3520-

3529.

23. Swimm A, Bommarius B, Reeves P, Sherman M, Kalman D. 2004.

Complex kinase requirements for EPEC pedestal formation. Nat Cell Biol

6:795; author reply 795-796.

24. Caron E, Crepin VF, Simpson N, Knutton S, Garmendia J, Frankel G.

2006. Subversion of actin dynamics by EPEC and EHEC. Curr Opin

Microbiol 9:40-45.

25. Campellone KG, Leong JM. 2005. Nck-independent actin assembly is

mediated by two phosphorylated tyrosines within enteropathogenic

Escherichia coli Tir. Mol Microbiol 56:416-432.

26. Brady MJ, Campellone KG, Ghildiyal M, Leong JM. 2007.

Enterohaemorrhagic and enteropathogenic Escherichia coli Tir proteins

trigger a common Nck-independent actin assembly pathway. Cell Microbiol

9:2242-2253.

29

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

609

610

Page 30: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

27. Vingadassalom D, Kazlauskas A, Skehan B, Cheng HC, Magoun L,

Robbins D, Rosen MK, Saksela K, Leong JM. 2009. Insulin receptor

tyrosine kinase substrate links the E. coli O157:H7 actin assembly effectors

Tir and EspF(U) during pedestal formation. Proc Natl Acad Sci U S A

106:6754-6759.

28. Weiss SM, Ladwein M, Schmidt D, Ehinger J, Lommel S, Stading K,

Beutling U, Disanza A, Frank R, Jansch L, Scita G, Gunzer F, Rottner K,

Stradal TE. 2009. IRSp53 links the enterohemorrhagic E. coli effectors Tir

and EspFU for actin pedestal formation. Cell Host Microbe 5:244-258.

29. Whale AD, Hernandes RT, Ooka T, Beutin L, Schuller S, Garmendia J,

Crowther L, Vieira MA, Ogura Y, Krause G, Phillips AD, Gomes TA,

Hayashi T, Frankel G. 2007. TccP2-mediated subversion of actin dynamics

by EPEC 2 - a distinct evolutionary lineage of enteropathogenic Escherichia

coli. Microbiology 153:1743-1755.

30. Deng W, Vallance BA, Li Y, Puente JL, Finlay BB. 2003. Citrobacter

rodentium translocated intimin receptor (Tir) is an essential virulence factor

needed for actin condensation, intestinal colonization and colonic hyperplasia

in mice. Mol Microbiol 48:95-115.

31. Schuller S, Chong Y, Lewin J, Kenny B, Frankel G, Phillips AD. 2007. Tir

phosphorylation and Nck/N-WASP recruitment by enteropathogenic and

enterohaemorrhagic Escherichia coli during ex vivo colonization of human

intestinal mucosa is different to cell culture models. Cell Microbiol 9:1352-

1364.

32. Ritchie JM, Brady MJ, Riley KN, Ho TD, Campellone KG, Herman IM,

Donohue-Rolfe A, Tzipori S, Waldor MK, Leong JM. 2008. EspFU, a type

30

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

Page 31: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

III-translocated effector of actin assembly, fosters epithelial association and

late-stage intestinal colonization by E. coli O157:H7. Cell Microbiol 10:836-

847.

33. Mallick EM, Garber JJ, Vanguri VK, Balasubramanian S, Blood T,

Clark S, Vingadassalom D, Louissaint C, McCormick B, Snapper SB,

Leong JM. 2014. The ability of an attaching and effacing pathogen to trigger

localized actin assembly contributes to virulence by promoting mucosal

attachment. Cell Microbiol 16:1405-1424.

34. Mallick EM, McBee ME, Vanguri VK, Melton-Celsa AR, Schlieper K,

Karalius BJ, O'Brien AD, Butterton JR, Leong JM, Schauer DB. 2012. A

novel murine infection model for Shiga toxin-producing Escherichia coli. J

Clin Invest 122:4012-4024.

35. Mundy R, Petrovska L, Smollett K, Simpson N, Wilson RK, Yu J, Tu X,

Rosenshine I, Clare S, Dougan G, Frankel G. 2004. Identification of a novel

Citrobacter rodentium type III secreted protein, EspI, and roles of this and

other secreted proteins in infection. Infect Immun 72:2288-2302.

36. Datsenko KA, Wanner BL. 2000. One-step inactivation of chromosomal

genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A

97:6640-6645.

37. Batchelor M, Guignot J, Patel A, Cummings N, Cleary J, Knutton S,

Holden DW, Connerton I, Frankel G. 2004. Involvement of the intermediate

filament protein cytokeratin-18 in actin pedestal formation during EPEC

infection. EMBO Rep 5:104-110.

38. Girard F, Dziva F, van Diemen P, Phillips AD, Stevens MP, Frankel G.

2007. Adherence of enterohemorrhagic Escherichia coli O157, O26, and O111

31

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

Page 32: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

strains to bovine intestinal explants ex vivo. Appl Environ Microbiol 73:3084-

3090.

39. Wiles S, Dougan G, Frankel G. 2005. Emergence of a 'hyperinfectious'

bacterial state after passage of Citrobacter rodentium through the host

gastrointestinal tract. Cell Microbiol 7:1163-1172.

40. Girard F, Frankel G, Phillips AD, Cooley W, Weyer U, Dugdale AH,

Woodward MJ, La Ragione RM. 2008. Interaction of enterohemorrhagic

Escherichia coli O157:H7 with mouse intestinal mucosa. FEMS Microbiol

Lett 283:196-202.

41. Girard F, Crepin VF, Frankel G. 2009. Modelling of infection by

enteropathogenic Escherichia coli strains in lineages 2 and 4 ex vivo and in

vivo by using Citrobacter rodentium expressing TccP. Infect Immun 77:1304-

1314.

42. Kufer TA, Kremmer E, Adam AC, Philpott DJ, Sansonetti PJ. 2008. The

pattern-recognition molecule Nod1 is localized at the plasma membrane at

sites of bacterial interaction. Cell Microbiol 10:477-486.

43. Allen-Vercoe E, Toh MC, Waddell B, Ho H, DeVinney R. 2005. A

carboxy-terminal domain of Tir from enterohemorrhagic Escherichia coli

O157:H7 (EHEC O157:H7) required for efficient type III secretion. FEMS

Microbiol Lett 243:355-364.

44. Heath RJ, Leong JM, Visegrady B, Machesky LM, Xavier RJ. 2011.

Bacterial and host determinants of MAL activation upon EPEC infection: the

roles of Tir, ABRA, and FLRT3. PLoS Pathog 7:e1001332.

45. Kolaczkowska E, Kubes P. 2013. Neutrophil recruitment and function in

health and inflammation. Nat Rev Immunol 13:159-175.

32

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

685

Page 33: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

46. Smith K, Humphreys D, Hume PJ, Koronakis V. 2010. Enteropathogenic

Escherichia coli recruits the cellular inositol phosphatase SHIP2 to regulate

actin-pedestal formation. Cell Host Microbe 7:13-24.

47. Yan D, Wang X, Luo L, Cao X, Ge B. 2012. Inhibition of TLR signaling by

a bacterial protein containing immunoreceptor tyrosine-based inhibitory

motifs. Nat Immunol 13:1063-1071.

48. Yan D, Quan H, Wang L, Liu F, Liu H, Chen J, Cao X, Ge B. 2013.

Enteropathogenic Escherichia coli Tir recruits cellular SHP-2 through ITIM

motifs to suppress host immune response. Cell Signal 25:1887-1894.

49. Keestra AM, Winter MG, Auburger JJ, Frassle SP, Xavier MN, Winter

SE, Kim A, Poon V, Ravesloot MM, Waldenmaier JFT, Tsolis RM,

Eigenheer RA, Baumler AJ. 2013. Manipulation of small Rho GTPases is a

pathogen-induced process detected by NOD1. Nature 496:233-+.

50. Legrand-Poels S, Kustermans G, Bex F, Kremmer E, Kufer TA, Piette J.

2007. Modulation of Nod2-dependent NF-kappaB signaling by the actin

cytoskeleton. J Cell Sci 120:1299-1310.

51. Kufer TA. 2008. Signal transduction pathways used by NLR-type innate

immune receptors. Mol Biosyst 4:380-386.

52. Bielig H, Lautz K, Braun PR, Menning M, Machuy N, Brugmann C,

Barisic S, Eisler SA, Andree M, Zurek B, Kashkar H, Sansonetti PJ,

Hausser A, Meyer TF, Kufer TA. 2014. The cofilin phosphatase slingshot

homolog 1 (SSH1) links NOD1 signaling to actin remodeling. PLoS Pathog

10:e1004351.

33

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

704

705

706

707

708

Page 34: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

53. Berger CN, Crepin VF, Jepson MA, Arbeloa A, Frankel G. 2009. The

mechanisms used by enteropathogenic Escherichia coli to control filopodia

dynamics. Cell Microbiol 11:309-322.

54. Arbeloa A, Garnett J, Lillington J, Bulgin RR, Berger CN, Lea SM,

Matthews S, Frankel G. 2010. EspM2 is a RhoA guanine nucleotide

exchange factor. Cell Microbiol 12:654-664.

55. Bulgin R, Arbeloa A, Goulding D, Dougan G, Crepin VF, Raymond B,

Frankel G. 2009. The T3SS effector EspT defines a new category of invasive

enteropathogenic E. coli (EPEC) which form intracellular actin pedestals.

PLoS Pathog 5:e1000683.

56. Young JC, Clements A, Lang AE, Garnett JA, Munera D, Arbeloa A,

Pearson J, Hartland EL, Matthews SJ, Mousnier A, Barry DJ, Way M,

Schlosser A, Aktories K, Frankel G. 2014. The Escherichia coli effector

EspJ blocks Src kinase activity via amidation and ADP ribosylation. Nat

Commun 5:5887.

57. Li S, Zhang L, Yao Q, Li L, Dong N, Rong J, Gao W, Ding X, Sun L,

Chen X, Chen S, Shao F. 2013. Pathogen blocks host death receptor

signalling by arginine GlcNAcylation of death domains. Nature 501:242-246.

58. Pearson JS, Riedmaier P, Marches O, Frankel G, Hartland EL. 2011. A

type III effector protease NleC from enteropathogenic Escherichia coli targets

NF-kappaB for degradation. Mol Microbiol 80:219-230.

59. Pearson JS, Giogha C, Ong SY, Kennedy CL, Kelly M, Robinson KS,

Lung TW, Mansell A, Riedmaier P, Oates CV, Zaid A, Muhlen S, Crepin

VF, Marches O, Ang CS, Williamson NA, O'Reilly LA, Bankovacki A,

Nachbur U, Infusini G, Webb AI, Silke J, Strasser A, Frankel G,

34

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

733

Page 35: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Hartland EL. 2013. A type III effector antagonizes death receptor signalling

during bacterial gut infection. Nature 501:247-251.

60. Newton HJ, Pearson JS, Badea L, Kelly M, Lucas M, Holloway G,

Wagstaff KM, Dunstone MA, Sloan J, Whisstock JC, Kaper JB, Robins-

Browne RM, Jans DA, Frankel G, Phillips AD, Coulson BS, Hartland EL.

2010. The type III effectors NleE and NleB from enteropathogenic E. coli and

OspZ from Shigella block nuclear translocation of NF-kappaB p65. PLoS

Pathog 6:e1000898.

61. Baruch K, Gur-Arie L, Nadler C, Koby S, Yerushalmi G, Ben-Neriah Y,

Yogev O, Shaulian E, Guttman C, Zarivach R, Rosenshine I. 2011.

Metalloprotease type III effectors that specifically cleave JNK and NF-

kappaB. EMBO J 30:221-231.

62. Pallett MA, Berger CN, Pearson JS, Hartland EL, Frankel G. 2014. The

type III secretion effector NleF of enteropathogenic Escherichia coli activates

NF-kappaB early during infection. Infect Immun 82:4878-4888.

63. Wiles S, Clare S, Harker J, Huett A, Young D, Dougan G, Frankel G.

2004. Organ specificity, colonization and clearance dynamics in vivo

following oral challenges with the murine pathogen Citrobacter rodentium.

Cell Microbiol 6:963-972.

64. Galan JE, Ginocchio C, Costeas P. 1992. Molecular and functional

characterization of the Salmonella invasion gene invA: homology of InvA to

members of a new protein family. J Bacteriol 174:4338-4349.

65. Raymond B, Crepin VF, Collins JW, Frankel G. 2011. The WxxxE effector

EspT triggers expression of immune mediators in an Erk/JNK and NF-

kappaB-dependent manner. Cell Microbiol 13:1881-1893.

35

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

753

754

755

756

757

758

Page 36: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Figure legends

Fig.1 Schematic representing the different Tir variants used in this study. The two

transmembrane helices, upstream and downstream of the intimin-binding domain, are

represented as black boxes and include amino acids 231-257 and 360-382,

respectively. Tyrosine residues Y451 and Y471 are shown in black and the

substitutions Y451A and Y471A in grey. Deletions are represented as dotted lines.

Fig. 2. (A) Immunofluoresence of Swiss 3T3 cells infected with C. rodentium. Tir

(green) was detected under adherent C. rodentium expressing either TirC-ctrl,

Tir382-462_Y451 or Tir478-547_Y451/Y471. Polymerized actin (red) was

observed under Tir staining of all adherent bacteria. Bar = 5 m. Infected cells were

also analyzed by SEM. Bar = 10 m. (B) Tir translocation was qualitatively assess by

determining the percentage of adherent bacteria showing translocated Tir staining.

While C. rodentium expressing either TirC-ctrl or Tir382-462_Y451 showed no

difference in their Tir translocation efficiency, C. rodentium expressing Tir478-

547_Y451/Y471 translocated Tir significantly less effectively.

Fig. 3. (A) Colonization dynamics to the peak of C. rodentium infection (day8 post

infection). C57Bl/6 mice inoculated with C. rodentium expressing TirC-ctrl, Tir382-

462_Y451 or Tir382-462_Y451A exhibit similar colonization dynamics. Mice

infected with C. rodentium expressing Tir478-547_Y451/Y471 or Tir478-

547_Y451A/Y471A showed a reduced colonization compared to strains expressing

TirC-ctrl, Tir382-462_Y451 or Tir382-462_Y451A. No difference was observed

whether the Tir variants were made in the context of either Y451/Y471 or

36

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

780

781

782

Page 37: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

Y451A/Y471A. All strains colonized significantly better than mice infected with C.

rodentium expressing Tir1-33stop. ** P<0.005, *** P<0.001, Kruskal-Wallis test

comparing C. rodentium expressing TirC-ctrl and Tir478-547_Y451/Y471 or

Tir478-547_Y451A/Y471A. (B) Transmission and SEM of mice colonic epithelium

infected with C. rodentium expressing TirC-ctrl, Tir382-462_Y451A or Tir478-

547_Y451A/Y471A. Local effacement of the brush border microvilli and intimately

adherent bacteria (arrow), typical of A/E lesions, were observed following inoculation

of mice with any of the C. rodentium strains. Intact brush border microvilli were

observed in tissue extracted from uninfected mice. Bar = 1m or 5m.

Fig. 4. Colonic enterocytes were isolated from mice infected with wild-type C.

rodentium or C. rodentium expressing Tir_Y451A/Y471A, at day 7 post infection.

Enterocytes isolated from uninfected mice were used as controls. Tir (green) was

detected underneath attached C. rodentium bacteria; intense actin staining was seen at

the site of wild type C. rodentium infection, while weak staining was observed at the

attachment site of enterocytes infected with C. rodentium expressing

Tir_Y451A/Y471A. Enterocytes isolated from uninfected mice showed typical brush

border actin staining (arrow). Bar = 5m.

Fig. 5. At day 14 post infection, mice infected with C. rodentium expressing TirC-ctrl,

Tir_Y451A, Tir_Y471A, Tir_Y451A/Y471A, TirΔ382-462_Y471 or TirΔ382-

462_Y471A showed equivalent bacterial shedding (A). Colonic tissues from mice

infected with C. rodentium expressing TirC-ctrl, Tir_Y451A, Tir_Y471A,

Tir_Y451A/Y471A or PBS mock-infected mice were harvested 14 days post infection

37

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

802

803

804

805

806

Page 38: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

and processed for FACS analyses. No difference in recruitment of (B) macrophages

(CD11b+Ly6G-F4/80+), (C) T helper cells (CD4+CD8-) and (D) B cells

(B220+CD3-) was observed in tissue infected with C. rodentium expressing the Tir

variants. Results are from two independent experiments with 5-6 mice per group. (E-

F) Neutrophils recruitment was significantly lower in tissue isolated from mice

infected with C. rodentium expressing Tir_Y451A/Y471A compared to Tir wild type

or single tyrosine mutant. (G-H) Neutrophils were also analyzed in colonic tissue of

mice infected with C. rodentium expressing either TirΔ382-462_Y471 or TirΔ382-

462_Y471A. Reduced neutrophils recruitment was observed specifically in tissue

infected with C. rodentium expressing TirΔ382-462_Y471A.

Fig. 6. (A) Enterocytes were isolated from colonic tissue of mice infected with C.

rodentium or mocked-infected with PBS and assessed for purity using flow

cytometry. Samples were stained for the leukocyte marker CD45 and the epithelial

cell marker CD326 (EpCAM). 80-90% of the cells were labelled CD326+CD45-,

which constituted the enterocyte population of the sample. Enterocytes were isolated

from colonic tissue of mice infected with C. rodentium expressing TirC-ctrl,

Tir_Y451A, Tir_Y471A or Tir_Y451A/Y471A and the expression of the neutrophil

chemoattractant CXCL1 (B) and CXCL2 (C) was measured by Q-RT-PCR (result is

from two independent enterocyte isolation experiments, n=4 per group). A reduced

level of CXCL1 and CXCL2 mRNA was observed in tissue infected with C.

rodentium expressing TirΔ382-462_Y471A. Data presented relative to GAPDH and

PBS control. *P<0.05, Students t test.

Fig. 7. (A) Representative H&E section of colonic tissue from mice (n=4) infected

38

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

828

829

830

831

Page 39: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

with C. rodentium expressing TirC-ctrl or Tir_Y451A/Y471A and PBS mock-

infected mice, at day 14 post infection. (B) Measurements of crypt length reveal

significantly reduced level of colonic hyperplasia on day 14 post infection in mice

infected with C. rodentium expressing Tir_Y451A/Y471A compared to mice infected

with C. rodentium expressing TirC-ctrl, ***P<0.0001. (C) Frozen colonic sections

were stained with antibodies against E-Cadherin (tissue contrast), Ly-6G (neutrophils)

or hoechst (nuclei). Representative immunofluorescence showing more Ly-6G

positive neutrophils stain (pink) in colonic section of mice infected with C. rodentium

expressing TirC-ctrl than mice infected with C. rodentium expressing

Tir_Y451A/Y471A or the PBS mock-infected control mice (Bar= 100m).

39

832

833

834

835

836

837

838

839

840

841

842

Page 40: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

 Table 1. Strains, plasmids and primers used in this study

 Description Reference

Strains

ICC169 Wild type C. rodentium O152 serotype (63)

ICC294 C. rodentium expressing TirC-ctrl (14)

ICC295 C. rodentium expressing Tir1-33stop (14)

ICC297 C. rodentium expressing Tir_Y451A (14)

ICC298 C. rodentium expressing Tir_Y471A (14)

ICC301 C. rodentium expressing Tir_Y451A/471A (14)

ICC1168 C. rodentium expressing Tir 382-462_Y471Δ This study

ICC1169 C. rodentium expressing Tir 382-462_Y471AΔ This study

ICC1170 C. rodentium expressing Tir 478-547_Y451/Y471Δ This study

ICC1171 C. rodentium expressing Tir 478-547_Y451A/Y471AΔ This study

Plasmids

pGEMT Cloning vector Promega

pKD46 Coding for the lambda Red recombinase (36)

pSB315 Plasmid coding for the kanamycin resistance aphT cassette (64)

pICC433 pGEMT vector containing the 3’ end of tirCITRO (bp 1067-1644), the aphT

cassette, tir-cesT intergenic region and the 5’ end of cesT (bp 1-388)

(14)

pICC438 pICC433 containing tirCITRO Y451A/Y471A mutation (14)

pICC1842 pGEMT vector containing the 3’ end of tirCITRO (bp 1067-1434), from which This study

40

843

844

845

Page 41: Imperial College Londonspiral.imperial.ac.uk/bitstream/10044/1/24750/2/IAI00291... · Web viewEscherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A 97: 6640-6645. 37

amino acids 478-547 have been deleted, the aphT cassette, tir-cesT intergenic

region and the 5’ end of cesT (bp 1-388)

pICC1843 pICC1842 containing tirCITRO Y451A/Y471A mutation This study

pICC1844 pGEMT vector containing the 3’ end of tirCITRO (bp 664-1644), in which amino

acids 382-462 have been deleted, the aphT cassette, tir-cesT intergenic region

and the 5’ end of cesT (bp 1-388)

This study

pICC1845 pICC1844 containing tirCITRO Y471A mutation This study

Primer name Nucleotide sequence

Tir-P478DSV-stop-EcoRI-Rv 5’-ccggaattcttaaacagaatcaggatccggagcgacttcatc-3’ This study

down-Tir-EcoRI-Fw 5’-ccggaattcatatataatgggtattttgttggggggg-3’ This study

Tir-down-TM2-Y471-Fw 5’-atgctccatagacgaaattcgcttctcgctccagaagag-3’ This study

Tir-EcoRI-Rv 5’-ccggaattcttagacgaaacgttcaactccc-3’ This study

Tir-upTM1-Fw 5’-acaacttcaagtgttcgttcag-3’ This study

Tir-down-TM2-Rv 5’-atttcgtctatggagcatagcc-3’ This study

EcoRI-(tir-cesT)-Fw 5’-gattatgtaataccaggtacagg-3' (14)

NcesT-Rv 5’-gcagccctagcatcacaaacagacggcgcgacaag-3’ (14)

Tir-Up-YY-Fw 5’-tggatctctcatcaggtattgg-3’ This study

mCXCL1-F (KC-F) 5’-tggctgggattcacctcaagaaca-3’ (65)

mCXCL1-R (KC-R) 5’-tgtggctatgacttcggtttgggt-3’ (65)

mGAPDH-F 5’-tcaacagcaactcccactcttcca-3’ This study

mGAPDH-R 5’-accctgttgctgtagccgtattca-3’ This study

41

846