characterisation of the type iii secretion effector nlef › bitstream › 10044 › 1 ›...

187
1 Characterisation of the type III secretion effector NleF Ph.D. Thesis Submitted to the Department of Life Sciences, Imperial College London, in partial fulfilment of the requirements for the degree of Doctor of Philosophy By Mitchell Pallett CMBI, Imperial College London Supervisor: Prof. G Frankel

Upload: others

Post on 03-Jul-2020

8 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

1

Characterisation of the type III secretion effector NleF

Ph.D. Thesis Submitted to the Department of Life Sciences,

Imperial College London, in partial fulfilment of the requirements for the degree of

Doctor of Philosophy

By Mitchell Pallett

CMBI, Imperial College London Supervisor: Prof. G Frankel

Page 2: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

2

Abstract

The human-specific enteropathogenic Escherichia coli and the mouse-restricted

Citrobacter rodentium colonise the host intestinal mucosa via attaching and effacing

lesions, inducing severe diarrhoeal disease and transmissible murine colonic

hyperplasia, respectively. C. rodentium is widely used as an in vivo model for EPEC

infection, sharing a highly homologous arsenal of virulence factors and a similar

infection strategy with EPEC. EPEC and C. rodentium pathogenesis relies on the

locus of enterocyte effacement-encoded type III secretion system and delivery of

effectors into the host cytosol to subvert host cell signalling including: actin

dynamics, cell trafficking and immune signalling. The role of many effectors during

infection remains unclear. We sought to identify the function of the non-LEE-encoded

effector protein F (NleF). We discovered that EPEC infection of polarised epithelial

cells activated a caspase-4-dependent non-canonical inflammasome response

leading to the processing and secretion of IL-18, which was counteracted in a T3SS-

and NleF-dependent manner. EPEC NleF interacts with the p20 and p10 subunits of

caspase-4, associating with the substrate-binding domain, to inhibit its proteolytic

activity and downstream inflammasome activation. Infection of mice with a C.

rodentium nleF mutant enhanced IL-18 secretion and revealed that NleF is essential

for the inhibition of the caspase-1/11-dependent inflammasome and recruitment of

neutrophils in vivo. We further report that ectopically expressed NleF activated NF-

κB nuclear translocation and the up-regulation of the pro-inflammatory chemokine IL-

8. Infection of HeLa with an EPEC nleF deletion mutant abolished the T3SS-

dependent activation of NF-κB and the expression of IL-8. NleF was identified to act

upstream to IκBα activation and could be inhibited by the TAB2/3 inhibitor NleE1.

However the mechanism of the pro-inflammatory role of NleF remains unclear.

Page 3: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

3

These findings identify novel roles for the T3SS effector NleF, furthering our

understanding of the infection strategy of EPEC and the host epithelial cell

inflammasome response.

Page 4: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

4

Copyright Declaration

The copyright of this thesis rests with the author and is made available under a

Creative Commons Attribution Non-Commercial No Derivatives licence. Researchers

are free to copy, distribute or transmit the thesis on the condition that they attribute it,

that they do not use it for commercial purposes and that they do not alter, transform

or build upon it. For any reuse or redistribution, researchers must make clear to

others the licence terms of this work.

I hereby declare that the work presented in this thesis is my own. Any work

contributing to this study but carried out by others is described below or appropriately

referenced.

The transduction and complementation of the Casp11-/- iMM and infections with

STBL2/LPS treatment, along with the ELISAs, were carried out by Dr. A. Shenoy

(CMBI, Imperial College London). The caspase-4 specific and non-targeting control

shRNA miR30E backbone plasmids were designed by Dr. A. Shenoy and

synthesised by Miss J. Sanchez-garrido (Shenoy group).

Co-purification and characterisation of the NleFE69 and caspase-4 complex was

carried out by Dr. O. Kotik (CMBI, Imperial College London).

The in vivo characterisation of the C. rodentium homolog of NleF was carried out by

Dr. V. Crepin-Sevenou and Dr. M. Habibzay (CMBI, Imperial College London).

Infection and analysis of C. rodentium infection of Casp1/11-/- C57BL/6 mice was

carried out by our collaborators Dr. N. Serafini and Prof. J. Di Santo (Institut Pasteur)

The mutagenesis and generation of the ΔnleF C. rodentium strain was carried out by

Dr. V. Crepin-Sevenou and Mr. G. Frankel (CMBI, Imperial College London)

Page 5: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

5

Table of Contents

Abstract ................................................................................................................... 2

Copyright Declaration ............................................................................................. 4

List of figures and tables ......................................................................................... 9

Abbreviations ........................................................................................................ 11

Chapter 1: Introduction ............................................................................................. 15

1.1 Escherichia Coli .......................................................................................... 16

1.1.1 Pathogenic E. coli ................................................................................. 16

i. ETEC .................................................................................................... 17

ii. DAEC.................................................................................................... 18

iii. EAEC and STEAEC .............................................................................. 19

iv. EIEC ..................................................................................................... 21

v. EPEC .................................................................................................... 23

vi. EHEC .................................................................................................... 25

1.1.2 Citrobacter rodentium ........................................................................... 26

i. The C. rodentium mouse model ........................................................... 27

1.2 Bacterial Secretion Systems ....................................................................... 28

1.2.1 The General Secretory Pathway ........................................................... 29

1.2.2 The Type II and V secretion system ..................................................... 30

1.2.3 The Type I Secretion system ................................................................ 31

1.2.4 The Type VI secretion system .............................................................. 31

1.2.5 The Type IV secretion system .............................................................. 32

1.2.6 The Type III Secretion System ............................................................. 33

i. The Locus of Enterocyte Effacement Pathogenicity Island ................... 34

ii. Biogenesis of the EPEC and the EHEC T3SS ...................................... 34

iii. Regulation of the T3SS ......................................................................... 36

1.3 Effector proteins and Manipulation of host cell signalling ............................ 38

1.4 The Host innate immune response ............................................................. 40

1.4.1 Pattern recognition receptors and host-pathogen detection ................. 40

1.4.2 The Toll-like receptors .......................................................................... 41

1.5 Intracellular detection via PRRs .................................................................. 44

1.5.1 RHLs and DNA sensors........................................................................ 44

1.5.2 The NODs and NOD-like receptors ...................................................... 45

i. NODs: Pathogen Detection and Signalling ........................................... 46

1.6 NLRs and the Inflammasome ...................................................................... 47

1.6.1 The NLRC4 inflammasome .................................................................. 47

i. NLRC4 detection of EPEC E69 and C. rodentium ligands.................... 48

Page 6: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

6

1.6.2 NLRP3 .................................................................................................. 49

i. The canonical inflammasome ............................................................... 50

1.6.3 Caspase-4 ............................................................................................ 51

i. Structural activation of caspase-4 ......................................................... 51

ii. Caspase-4: Homologs and orthologs ................................................... 52

iii. Caspase-4/-5/-11 and the non-canonical inflammasome ...................... 53

iv. The unconventional non-canonical inflammasome in epithelial cells .... 56

1.6.4 The role of the inflammasome in vivo ................................................... 57

1.6.5 The IL-1 cytokine family ........................................................................ 58

i. IL-1α and IL-1β share biological properties .......................................... 58

ii. The functional role of IL-1β in vivo ........................................................ 58

iii. IL-18 and IFNγ inducing factor ............................................................. 59

iv. The functional role of IL-18 in vivo ........................................................ 60

1.7 Manipulation of the inflammatory response by enteric pathogens............... 61

1.7.1 The EPEC anti-inflammatory effectors NleB1, NleC, NleD and NleE1 inhibit NF-κB and JNK signalling........................................................................ 61

1.7.2 Inflammatory signalling during EPEC E69 infection .............................. 63

1.7.3 EspT is a pro-inflammatory effector of atypical EPEC .......................... 64

1.8 NleF of attaching and effacing pathogens ................................................... 65

1.9 Aims and outline .......................................................................................... 67

Chapter 2: Materials and Methods ........................................................................... 68

2.1 Bacterial strains, reagents, growth conditions and plasmids ....................... 69

2.2 Y2H screening and YDH hybrids ................................................................ 69

2.3 Tissue culture .............................................................................................. 70

2.4 Transfection assays .................................................................................... 71

2.4.1 Plasmid transfection of HeLa cells ........................................................ 71

2.4.2 Transfection of short inhibitory RNA ..................................................... 71

2.5 Isolation of RNA and RT-qPCR ................................................................... 72

2.6 Immunofluorescence staining and antibodies ............................................. 72

2.7 Bacterial priming, cell growth analysis and attachment assays ................... 73

2.8 Generation of an EPEC E69 nleF deletion strain ........................................ 74

2.9 Infection of assays ...................................................................................... 74

2.9.1 Infection of the HeLa cell line ................................................................ 74

2.9.2 Infection of iMM .................................................................................... 75

2.9.3 Infection of the Caco-2 cell line ............................................................. 75

2.10 Western blotting and coomassie staining ................................................. 76

2.11 Cytokine secretion analysis and cell death assays .................................. 77

Page 7: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

7

2.12 Affinity –tagged purification and size exclusion ........................................ 78

2.13 Co-expression and purification of the caspase-4/NleFE69 complex .......... 78

2.14 Caspase activity assays ........................................................................... 79

2.15 shRNA construction and retroviral transductions ..................................... 79

2.16 Generation of C. rodentium nleF knockout mutant................................... 80

2.17 C. rodentium in vivo mouse model of infection ........................................ 81

2.18 Sample collection and FACs analysis ...................................................... 81

2.19 Ex-vivo ex-plants ...................................................................................... 82

2.20 Statistical analysis .................................................................................... 83

Results: Chapter 3 - Identification and confirmation of host interacting partners ..... 91

3.1 A Y2H screen identifies caspase-4 as an interacting partner of NleFE69 ..... 92

3.2 NleFE69 specifically interacts with caspase-4 ............................................... 94

3.3 NleFE69 interacts with active caspase-4 dependent on its’ C-terminal motif LQCG .................................................................................................................... 97

3.4 NleFE69 binds and inhibits the catalytic activity of caspase-4/11 ................. 99

3.4.1 Developing a caspase-4/-11 activity assay ........................................... 99

3.4.2 Purification of NleFE69 for in vitro activity assays ................................ 101

3.4.3 NleF of A/E pathogens inhibit the activity of caspase-4/-11 ................ 103

3.5 Preliminary data suggests NleFE69 forms a complex with the active caspase-4 hetero-tetramer ................................................................................................ 104

3.5.1 Co-purification of active caspase-4 and NleFE69 ................................. 105

3.6 Discussion ................................................................................................. 107

Results: Chapter 4 – Caspase-4, the functional ortholog of murine caspase-11 is target by EPEC NleF .............................................................................................. 110

4.1 Caspase-4 is the functional ortholog of murine caspase-11.......................... 111

4.1.1 Caspase-4 can functionally complement caspase-11 to activate the non-canonical inflammasome in response to EPEC infection ................................. 111

4.1 Generation of an nleF deletion EPEC strain ............................................. 114

4.2 NleFE69 inhibits activation and secretion of caspase-4 during infection of polarised intestinal epithelial cells ....................................................................... 116

4.3 Activation of caspase-4 in response to EPEC E69 infection triggers IL-18 processing and secretion .................................................................................... 119

4.4 Discussion ................................................................................................. 122

Results: Chapter 5 – NleFE69 activates NF-κB signalling during EPEC infection .. 127

1.10 NleFE69 localises to the mitochondria and activates NF-κB nuclear translocation ........................................................................................................ 128

1.10.1 NleFE69 localises to the mitochondria dependent on its C-terminal motif LQCG…………………………………………………………………………………128

Page 8: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

8

1.10.2 Ectopically expressed NleFE69 activates nuclear translocation of NF-κB………..…………………………………………………………………………….131

1.11 NleFE69 up-regulates the expression of the chemokine IL-8 ................... 135

1.11.1 NleFE69 induced NF-κB activation is independent of interaction with caspase-4, -8 or -9. .......................................................................................... 136

1.12 NleFE69 activates the nuclear translocation of NF-κB early during EPEC E69 infection ....................................................................................................... 139

1.13 NleFE69 dependent activation of NF-κB is up stream to IκBα degradation……………………………………………………………………………..144

1.14 NleFE69 interacts with human DHFR ....................................................... 145

1.15 Discussion ............................................................................................. 147

1.15.1 Mechanism of NF-κB activation ....................................................... 148

Results: Chapter 6 - NleFCR inhibits IL-18 secretion and neutrophil recruitment in vivo ............................................................................................................................... 152

6.1 NleFCR inhibits IL-18 secretion and neutrophil recruitment in vivo ............. 153

6.1.1 NleFCR is not required for normal colonisation in vivo ......................... 153

6.1.2 C. rodentium delivers NleFCR to inhibit IL-18 secretion ....................... 154

6.2 Deletion of nleF increases colonic neutrophil recruitment during C. rodentium infection .............................................................................................. 155

6.3 Recruitment of neutrophils and IL-18 secretion early during C. rodentium infection is caspase-1/11 dependent. .................................................................. 157

6.4 Discussion ................................................................................................. 158

6.4.1 NleFCR as a pro-inflammatory effector of C. rodentium ....................... 161

6.4.2 The role of NleF in the context of EPEC infection ............................... 163

Chapter 7: Conclusions .......................................................................................... 165

7.1 Overview ................................................................................................... 166

7.2 Conclusions and implications .................................................................... 166

Acknowledgements ............................................................................................. 168

Bibliography ........................................................................................................ 171

Permissions for use of unpublished data ............................................................ 187

Page 9: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

9

List of figures and tables

Chapter 1: Introduction Figure 1.1: Secretion systems of Gram negative bacteria……………........... 30 Figure 1.2: The locus of enterocyte effacement……………………………….. 35 Figure 1.3: The T3SS translocon of EPEC………………………………......... 36 Figure 1.4: EPEC infection is characterised by pedestal and A/E lesion formation in vivo…………………………………………………………………...

40

Figure 1.5: The TLR-4 and TNFR1 intracellular signalling cascades............ 44 Figure 1.6: Structural activation of caspase-4...……………………………….. 53 Figure 1.7: The non-canonical inflammasome model………………..……….. Figure 1.8: EPEC E69 recruits a plethora of effectors to inhibit inflammatory signalling during infection………………………………………… Figure 1.9: Alignment of NleF of pathogenic E. coli and distribution of PAI Prophage 6 (PP6)……………...………………………………………………….

55

63

67 Chapter 2: Material and methods

Table 2.1: Bacterial strains used in this study…………………………………. 85 Table 2.2: Plasmids and Primers used for cloning in this study………….. 86 Table 2.3: cDNA and templates used in this study………………………........ 90 Table 2.4: Primers used for mutagenesis and RT-qPCR in this study……… 91

Chapter 3: Results Table 3.1: A Y2H screen for NleFE69 identifies caspase-4 as a putative host target………………………………………………………………………….

94

Figure 3.1: NleFE6 interacts with full length caspase-4……………………….. 95 Figure 3.2: Caspase-5 shares significant homology with caspase-4 but does not interact with NleFE69..………………………………………………….

97

Figure 3.3: The C-terminal motif LQCG of NleFE69 interacts with active caspase-4.………………………................................................................

99

Figure 3.4: Caspase-4/-11 undergoes self-proteolytic cleavage and oligomerisation to cleave the fluorescent substrate LEVD-AFC.…………..…

101

Figure 3.5: Optimisation of IPTG induced expression of NleFE69 in BL21 Star……………………………………………………………………………….…

102

Figure 3.6: Purification of NleF homologs of A/E pathogens……………..….. 103 Figure 3.7: NleF inhibits caspase-4 and -11 activity in vitro………………..… 104 Figure 3.8: Recombinant NleFE69 forms a complex with the active caspase-4 complex…………………………………………..…………….

107

Chapter 4: Results Figure 4.1: EPEC activates the NLRC4 inflammasome in a T3SS dependent manner…………………………………………………………..…….

113

Figure 4.2: Caspase-4 can functionally complement caspase-11 to trigger the murine non-canonical inflammasome……………………………………….

114

Figure 4.3: Deletion of nleF does not affect EPEC E69 growth kinetics. 116 Figure 4.4: NleFE69 inhibits secretion of active caspase-4 and IL-18 during

Page 10: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

10

infection of IECs……………………………………....................................... 118 Figure 4.5: Deletion of nleF alone does not increase cell death during infection of IECs………………………………………….………………………..

119

Figure 4.6: Caspase-4 is essential for IL-18 processing and secretion during EPEC E69 infection of IECs……………………………………..……….

121

Figure 4.7: Full length caspase-1 is not detectable in the lysates of the Caco-2 clones TC7 or BBE1…………………………………………..…………

122

Chapter 5: Results Figure 5.1: Ectopically expressed NleFE69 localises to the mitochondria dependent on its C-terminal motif LQCG……………………….………………

132

Figure 5.2: Ectopically expressed NleF activates nuclear translocation of p65…………………………………………………………………………..………

134

Figure 5.3: NleFE69 activity is independent of subcellular localization…….. 135 Figure 5.4: Ectopic expression of NleFE69 induces up-regulation the NF-κB promoter dependent chemokine IL-8……………………………………………

136

Figure 5.5: NleFE69 functions independently of caspase-4, -8 or -9 to activation IL-8 expression………………………………………………………...

138

Figure 5.6: Knock-down of caspase-4 and -8 has no effect on the pro-inflammatory activity of NleFE69………………………………………………….

139

Figure 5.7: NleFE69 activates p65 nuclear translocation early during infection…………………………………………………………………………….

141

Figure 5.8: EPEC inhibits exogenous stimulation and T3SS-dependent activation of NF-κB at 4 h post-infection. ………………………………………

144

Figure 5.9: EPEC E69 induces IκBα degradation in an NleFE69 dependent manner during infection…………………………………………………………...

145

Figure 5.10: NleFE69 interacts with DHFR but not DHFRL1………………….. 147 Figure 5.11: EPEC E69 initially activates NF-κB then later inhibits inflammatory signalling in a T3SS dependent manner………………………

149

Chapter 6: Results Figure 6.1: NleFCR is not required for C. rodentium colonisation………….. 154 Figure 6.2: NleFCR inhibits early secretion of IL-18 from colonic tissue in vivo…………………………………………………………………………………..

155

Figure 6.3: NleFCR inhibits early colonic recruitment of neutrophils……….. 157 Figure 6.4: NleFCR dependent inhibition of neutrophil recruitment is via caspase-1/11………………………………………………………………………

158

Page 11: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

11

Abbreviations

A/E Attaching and effacing AAF Aggregative adherence fimbriae ABC ABC binding cassette AD Activation domain AcaA adhesin involved in diffuse Citrobacter adherence Ade Adenine Afa Afimbrial adhesin AggR Aggregative adherence virulence regulator AIDA-I Adhesins involved in diffuse adherence AIM2 Absent in Melanoma 2/ Interferon inducible protein 2 AP-1 Activator Protein 1 ARP2/3 Actin-related protein 2/3 Asc Apoptosis associated speck-like protein containing a CARD BD Binding domain BFP Type IV bundle forming pili CARD Caspase activation and recruitment domain CEACAM Carcinoembryonic antigen-related cell adhesion molecules Ces Chaperone for E. coli secreted protein CF Colonization factor CFA Colonization factor adhesins Cfc Citrobacter colonisation factor CFTR Cystic fibrosis transmembrane conductance regulator CFU Colony forming units cIAP1/2 Inhibitor of apoptosis protein 1/2 homolog C ClyA Cytolysin A CrC1sp Citrobacter rodentium Class I serine protease CREB1 Cyclic-AMP responsive element-binding protein 1 DAF Complement receptor CD55 DAI/ZBP DNA-dependent activator of IRFs DAMP Danger-associated molecular pattern DAP Meso-diaminopimelic acid DAPI 4,6-Diamidino-2-phenylindole DEAC Diffusely enteroadherent E. coli DHFR Dihydrofolate reductase DHFRL1/P4 Dihydrofolate isoform 4 DISC Death inducing signalling complex DNA Deoxyribonucleic acids EAEC Enteroadherent E. coli EAF EPEC adherence factor EAST-1 EAEC heat-stable toxin 1 EatA ETEC autotransporter A EfaI E. coli factor for adherence 1

Page 12: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

12

EhaA EHEC autotransporter encoding gene A EHEC Enterohaemorrhagic E. coli Ehx EHEC toxin EibG E. coli immunoglobulin binding protein G EIEC Enteroinvasive E. coli EPEC Enteropathogenic E. coli EPEC E69 Enteropathogenic E. coli O127:H6 E2348/69 ER Endoplasmic reticulum ERK1/2 Extracellular signal regulated kinase 1/2 Esc E. coli secretion gene Esp E. coli secreted protein ETEC Enterotoxigenic E. coli ETT2 EHEC Type III secretion system 2 ExPEC Extra-intestinal pathogenic E. coli FADD Fas-associated death-domain protein FAK Focal adhesion kinase GEF Guanine nucleotide exchange factor hNAIP Human neuronal apoptosis inhibitory protein HPV-16 Human papillomavirus Type 16 Hra Heat resistant agglutinin HSV-1 Herpes Simplex virus 1 HUS Haemolytic Uremic syndrome ICE Interleukin-1 beta converting enzyme IE Integrating element IEC Intestinal epithelial cell IFN Interferon IκBα NF-κB inhibitor alpha IκK IκB Kinase IL Interleukin ILC Innate lymphoid cell IL-1R Interleukin-1 receptor IL-18R Interleukin-18 receptor inPEC Intestinal pathogenic E. coli IPAF Ice protease-activating factor JNK c-Jun N-terminal kinase Kfc K99-like factor involved in Citrobacter colonisation KO Knockout IMC Inner membrane complex IMP Outer membrane complex iMM Immortalised mouse macrophages IRAK IL-1 receptor-associated kinase IRKTS Insulin receptor tyrosine kinase substrate IRF Interferon regulating factor LDH Lactose dehydrogenase

Page 13: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

13

LEE Locus of enterocyte effacement Ler LEE-encoded regulator Leu Leucine LPB LPS-binding protein LPS Lipopolysaccharide LRR Leucine rich repeat domain LT Heat-liable toxin LTB4 Leukotriene B4 Ly-96 Lymphocyte antigen 96 MAP Mitogen-activated protein MAPK Mitogen-activated protein kinase MDA5 Melanoma associated protein 5 MDP Muramyl-dipeptide MFP Membrane fusion protein MHC Major histocompatibility complex MIP-2 Macrophage inflammatory protein MKK Mitogen-activated protein kinase kinase MyD88 Myeloid differentiation primary response protein 88 NAIP Neuronal apoptosis inhibitory protein NACHT Nucleotide-binding and oligomerisation domain NALP NACHT, LRR and PYD domains-containing protein NBD Nucleotide binding domain NF-κB Nuclear factor Kappa B NK Natural killer cells nle Non-LEE encoded NLR NOD-like receptor NLRC NLR family CARD domain-containing protein NLRP NOD-like receptor family, pyrin domain containing protein NO Nitric oxide NOD Nucleotide-oligomerisation domain NRIR nle regulated inverted repeated N-WASP neural Wiskott–Aldrich syndrome protein OMC Outer-membrane complex OMP Outer-membrane protein pAA Aggregative adherence virulence plasmid PAA Porcine attaching and effacing associated adhesin PAI Pathogenicity island PAMP Pathogen-associated molecular pattern PBS Phosphate buffered saline Pet Plasmid encoded toxin PI Propidium iodide Pic Protein involved in colonisation PKC Protein Kinase C

Page 14: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

14

PMA Phorbol myristate acetate PMNs Polymorphonuclear leukocytes PP Prophage PRR Pattern recognition receptor PYD Pyrin domain qRT-PCR Quantitative Reverse transcription PCR RHL RIG-I-like helicases RIP Receptor-interacting serine/threonine-protein kinase RNA Ribonucleic acid ROS Reactive oxygen species Sat Secreted autotransporter toxin Sec Universal secretory pathway SepA Shigella extracellular protein shRNA Short hairpin RNA SigA Shigella IgA-like protease siRNA Short inhibitory RNA SPATE Serine protease autotransporter of the enterobacteriaceae ST Heat-stable toxin STEC Shiga-toxin producing E coli Stx Shiga-like toxin/ Verotoxin T1SS Type I secretion system T2SS Type II secretion system T3SS Type III secretion system T4CP Type IV coupling protein T4SS Type IV secretion system T5SS Type V secretion system T6SS Type VI secretion system TAB2/3 TGF-β activated and MAP3K7 binding protein 2/3 TAK1 Transforming growth factor-β-activated kinase 1 Tat Two-arginine system TccP Tir cytoskeleton coupling protein Th T helper cell Th17 T helper cell expressing IL-17 Tir Translocated intimin receptor TLR Toll-like receptor TMCH Transmissible murine crypt hyperplasia TNFα Tumour necrosis factor alpha TNFR TNFα receptor TPS Two partner system TRADD Tumour necrosis factor receptor type 1-associated death

domain protein TRAF TNF-receptor associated factor

Page 15: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

15

Chapter 1: Introduction

Page 16: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

16

1.1 Escherichia Coli

Escherichia coli (E. coli), the most abundant Gram negative facultative anaerobe of

the intestinal commensal microflora, mutually and beneficially co-exists with the

human host (Kaper, Nataro & Mobley, 2004). Considered the workhorse of the

molecular biologist it has been extensively exploited to further scientific research and

this has led it to be the foremost studied bacterial species and one of the earliest to

have its genome sequenced. While all E. coli strains share a common backbone of

around 4.1 Mbp, vertical gene transfer and the presence of punctuated lineage

specific elements through horizontal gene transfer and/ or possible deletion has led

to divergence over the past 4.5 million years and the emergence of pathogenic

strains (Donnenberg, 2002). Acquisition of elements associated with virulence are

assigned as O-islands and are detectable by their G + C content, insertion position

(eg. at a tRNA gene) and presence of a flanking integrase or transposase,

responsible for mobilisation (Donnenberg, 2002). Furthermore, pathogenic strains of

E. coli have acquired F-like plasmids associated with virulence and disease which

encode a range of virulence factors including transcriptional regulators, enterotoxins

and adhesins (Donnenberg, 2002). This acquisition of lineage specific elements has

resulted in a range of pathotypes with the ability of E. coli to cause a spectrum of

diseases in a range of hosts.

1.1.1 Pathogenic E. coli

Infection with pathogenic E. coli can be classified into three clinical syndromes:

enteric/diarrhoeal disease, urinary tract infection (UTI) or systemic infection leading

to meningitis (Nataro & Kaper, 1998). Strains that cause diarrhoeal disease are

classified as intestinal pathogenic E. coli (InPEC) while those causing systemic

infection/ UTI are grouped as extra-intestinal pathogenic E. coli (ExPEC; Croxen &

Page 17: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

17

Finlay, 2010). InPEC is a major cause of diarrhoeal disease which accounts for ~1.3

million deaths annually and remains the second leading cause of death in infants

worldwide (Ochoa & Contreras, 2011; Black et al., 2010). Furthermore inPEC is

subdivided into 8 distinct categories dependent upon mechanism of infection and

presence of specific virulence factors including bacterial toxins and colonisation

factors as reviewed by others (Clements et al., 2012; Croxen & Finlay, 2010; Kaper,

Nataro & Mobley, 2004). This includes: enteropathogenic (EPEC),

enterohaemorrhagic (EHEC), enteroinvasive (EIEC), enterotoxigenic (ETEC),

enteroadhesive (EAEC), diffusely adherent E. coli (DAEC) adherent invasive E. coli

(AIEC) and Shiga toxin producing enteroadhesive E. coli (STEAEC). AIEC is

associated with Crohn’s disease and is not a diarrheagenic pathogen and therefore

will not be covered any further (Clements et al., 2012). It should also be noted that

strains can also be classified by their O-antigen (serogroup) and their O-antigen and

flagelllar (serotype); although, a particular serotype may be common among E. coli

of different pathotypes.

i. ETEC

ETEC is the leading cause of diarrhoeal disease in travellers to developing countries

(Kaper, Nataro & Mobley, 2004). ETEC colonizes the surface of the small bowel

mucosa; inducing a watery diarrhoeal disease (Kaper, Nataro & Mobley, 2004).

Colonization is mediated by approximately 23 fibrillar, fimbrial and afimbrial

colonization factors (CFs; Croxen and Canto 2012); which confer host specificity

(Kaper, Nataro & Mobley, 2004), the outer-membrane proteins TibA and Toxigenic

invasion loci A (Tia), the flagellin tip protein EtpA and the porcine attaching and

effacing associated adhesin (Paa) (Clements et al., 2012; Croxen & Finlay, 2010).

ETEC is defined as E .coli that produces one or more of either the heat-liable (LTs)

Page 18: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

18

or heat-stable (STs) toxins; which are responsible for the ETEC-mediated diarrhoea.

LT, which functionally resembles Cholera toxin of Vibrio Cholerae, is an AB5 toxin

that once internalised activates host adenylyl cyclase resulting in an increase in

intracellular cyclic AMP and activation of Protein Kinase A (PKA). Importantly, LT –

dependent activation of PKA inhibits the expression of antimicrobial peptides

(Croxen & Finlay, 2010). Alternatively, STs, bind the cell surface receptor guanylyl

cyclase of the intestinal brush border increasing intracellular cyclic GMP and

decreasing Na+ absorption (Croxen & Finlay, 2010). Both LT and ST, through

manipulation of cGMP and cAMP levels, activate the cystic fibrosis transmembrane

conductance regulator (CFTR) increasing Cl- secretion (Croxen & Finlay, 2010).

Ultimately, manipulation of intracellular levels of cGMP and cAMP dysregulates

epithelial cell fluid transport and induces diarrhoea (Croxen & Finlay, 2010). ETEC

strains also encode the serine protease autotransporter of the enterobacteriaceae

(SPATE) family protein ETEC autotransporter A (EatA) as well as Cytolysin A (ClyA)

and EAEC heat stable toxin 1 (EAST-1). While EAST-1 and ClyA function similarly to

ST, the SPATE EatA cleaves cathepsin G associated substrates and mucin,

accelerating toxin delivery and intestinal fluid build-up (Kumar et al., 2014; Croxen &

Finlay, 2010; Patel et al., 2004).

ii. DAEC

DAEC colonizes the small bowel and urinary tract and although more commonly

associated with urinary tract infections it is also associated with diarrhoeal disease

(Croxen & Finlay, 2010). As the name suggests DAEC is characteristically

distinguished by its diffuse adherence pattern to HEp-2 cells in vitro (Servin, 2005).

DAEC is an extremely heterogeneous group and colonization and pathogenesis of

DAEC relies on the presence of cell surface fimbrial (Dr and F1845) and afimbrial

Page 19: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

19

(Afa) adhesins referred to collectively as the Afa-Dr fimbriae (Servin, 2005). The

Afa/Dr fimbriae bind to the host complement decay-accelerating factor (DAF), type IV

collagen and the Carcinoembryonic antigen-related cell adhesion molecules

(CEACAM) to facilitate tight adherence during infection (Servin, 2005; Berger et al.,

2004). Strains of DAEC are subdivided, by their repertoire of specific adhesins, into

typical or atypical DAEC. Typical DAEC encode the adhesins Afa/ Dr and F1845 and

are able to bind both DAF and CEACAM and are commonly associated with re-

occurring UTIs as well as enteric infection (Servin, 2005). Atypical DAEC express

Afa but no Dr or F1845 adhesins and do not bind DAF (Servin, 2005). A subclass of

atypical DAEC, commonly referred to as DA-EPEC, is associated with infantile

diarrhoea (2-3 yrs) and phenotypically resembles infection with EPEC but

characteristically adheres in a diffuse pattern through the presence of adhesins

involved in diffuse adherence (AIDA-I; Servin, 2005). Pathogenesis of DAEC is

predominately dependent on the expression of the Afa-Dr fimbriae although it does

encode the SPATE Secreted auto-transporter toxin (Sat); which is implicated in tight

junction lesion formation and an increase in cell permeability (Croxen & Finlay,

2010).

iii. EAEC and STEAEC

EAEC are phylogenetically similar to DAEC and were previously sub-classed

together as an adherent non-EPEC/enteroadherent pathogen before being

individually typed as having either diffuse or aggregative adherence (Weintraub,

2007; Nataro et al., 1987). EAEC is linked to outbreaks of watery diarrhoea in

developed countries and persistent diarrhoea in developing countries and is the

second most common cause of Travellers disease after ETEC (Croxen & Finlay,

2010). It colonizes the intestinal mucosa of the colon, where it up-regulates the

Page 20: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

20

secretion of mucus to form an auto-aggregative bacterial mucus-biofilm layer

(Weintraub, 2007). Pathogenesis of EAEC is regulated by the transcriptional

regulator AggR which controls the expression of adherence factors and the type 6

secretion system which is required for biofilm formation (Kaur, Chakraborti & Asea,

2010). Encoded by the aggregative adherence virulence plasmid (pAA) AggR

regulates the expression of Aggregative adherence fimbriae (AAFI/II/II). Each EAEC

isolate expresses only one AAF subtype which through the binding to extracellular

matrix proteins including type IV collagen, fibronectin and lamin is responsible for

initial adherence to the intestinal mucosa and the ‘stacked brick’-like pattern of

adherence (Kaur, Chakraborti & Asea, 2010). Interestingly atypical EAEC do not

possess pAA and therefore do not express AAFI/II/II; however, still adhere in the

characteristic ‘stacked brick’ pattern through a Dr adhesin-like aggregative adhesin

pilin, also regulated by AggR (Kaur, Chakraborti & Asea, 2010). Further colonization

factors identified vary among clinical isolates of EAEC but include the Heat resistant

agglutinin (Hra) 1 and 2, Tia and Dispersin, which potentiates AAF binding

(Clements et al., 2012). Through the secretion of enterotoxins and cytotoxins EAEC

elicits a pro-inflammatory response, mild mucosal damage and persistent watery

diarrhoea. This includes the pAA encoded toxin EAST-1 and the chromosomal

encoded SPATE proteins Protein involved in colonization (Pic), Plasmid encoded

toxin (Pet), Shigella IgA-like protease (SigA), Shigella extracellular protein (SepA), E.

coli secreted protein P (EspP) and Sat as well as the pore forming toxin HlyE and the

Shigella-like she island encoded Shigella enterotoxin 1/2 (ShET1/2; Clements et al.,

2012; Croxen & Finlay, 2010; Kaur, Chakraborti & Asea, 2010; Weintraub, 2007).

Pic, a class II SPATE, is an extracellular serine protease with mucinolytic activity

Page 21: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

21

required for colonization. Furthermore it has recently been identified that Pic cleaves

leukocyte cell surface glycoproteins to interfere with immune signalling

(Weiss & Brockmeyer, 2012; Wiles et al., 2004) Pet, SigA and EspP are class I

SPATEs. Pet and SigA have similar functions and both cleave the actin binding

protein spectrin to induce cytotoxicity and cell detachment while, EspP functions to

degrade proteins of the host complement system to manipulate immune signalling

(Weiss & Brockmeyer, 2012). SepA shares 73% homology and functions similarly to

EatA (Dautin, 2010). ShET1/2 is an AB5 toxin however its role in pathogenesis is still

not fully understood (Croxen & Finlay, 2010).

Further to the enterotoxins described above an emerging pathogenic subclass of

EAEC O104:H4 recently re-classified as Shiga Toxin (Stx) producing

Enteroaggregative E. coli (STEAEC) closely resembles EAEC but, through

acquisition of a stx2-harbouring prophage, encodes the Shiga toxin (Stx) as well as

the IrgA homolog adhesin (Iha) and is responsible for the outbreak in Germany in

2011 (Clements et al., 2012). Stx, also referred to as verocytotoxin, is released in the

colon due to bacterial cell lysis where it induces necrosis of the paneth cells of the

intestinal mucosa or targets the kidneys by entering the blood stream and binding to

the glycolipd globotriaosylceramide (Gb3) on the surface of renal endothelial cells

(Croxen & Finlay, 2010). This induces local cytokine secretion and inflammation

which leads to haemorrhagic colitis and in more severe cases to kidney damage and

the progression to Haemolytic-uremic syndrome (HUS) and bloody diarrhoea.

iv. EIEC

Pathogenesis of EIEC is closely associated with that of Shigella spp., to which it is

genetically closely related. Although disease can lead to dysentery and shigellosis it

Page 22: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

22

is rarely distinguishable from other E. coli pathotypes (Donnenberg, 2002). EIEC and

Shigella spp. share 90% genetic homology and the same O-antigens (Donnenberg,

2013). In fact it is now widely accepted that Shigella spp. are a distant clone of

commensal E. coli which acquired multiple virulence factors and most importantly a

high molecular weight plasmid (pINV) comprised of four separate ancestral plasmids

which is key to the virulence of Shigella spp.

While most pathogenic E. coli are extracellular; EIEC, is an intracellular pathogen

that lacks both flagella and adherence factors (Donnenberg, 2013). Due to the

selective pressure to adapt to the intracellular environment the EIEC pathotype and

infection strategy is uniquely different from the other inEPEC pathotypes. EIEC

exploits the immune system of the host to be preferentially taken up by M cells in the

large intestine where it is transcytosed across the enterocyte barrier and rapidly

phagocytosed by macrophages (Donnenberg, 2013). Rather than be killed EIEC is

able to evade the phagosome and induces a form of inflammatory cell death known

as pyroptosis to escape the macrophage. This gives EIEC the unique advantage to

infect the basolateral surface of the enterocytes. Following invasion it is able to

evade the endocytic vacuole and through the outer-membrane protein VirG it

manipulates the host cell actin cytoskeleton to direct movement in the cytoplasm to

disseminate to neighbouring cells (Kaper, Nataro & Mobley, 2004). The induction of

pyroptosis of resident macrophages, invasion of epithelial cells and subsequent

recruitment of polymorphonuclear leukocytes (PMNs) results in a pro-inflammatory

burst and inflammatory cell damage. This leads to destabilisation of tight junctions

and allows for further infection by exposing the sub-epithelial layer. Although EIEC

encodes the enterotoxins ShET1/2 and the SPATEs SepA and SigA, which are likely

to cause the diarrhoea which proceeds dysentery, a mutant which is unable to

Page 23: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

23

invade enterocytes does not cause disease in the monkey model (Donnenberg,

2013). In fact the invasion, intracellular survival and pathogenesis of EIEC is enabled

by the pINV-encoded type III secretion system (T3SS; see below) which is

homologous to Shigella spp.

v. EPEC

EPEC, the first pathogenic E. coli to be discovered in 1945, colonizes the human

small bowel and gastrointestinal tract causing watery diarrhoea (Wong et al., 2011).

EPEC is a major cause of infantile persistent diarrhoea in low to middle income

countries (Abba et al., 2009).The global enteric multicentre (GEMs), designed to

detect the cause of acute-moderate to severe paediatric (0-59 months) diarrhoeal

disease in sub-Saharan Africa and South Asia concluded that typical EPEC is

associated with increased fatality in children aged 0-11 months (Kotloff et al., 2013).

Moreover, a recent epidemiological study during 2010-12 found that EPEC is a still

an important cause of sporadic infection of infants in England (Sakkejha et al., 2013).

Typical EPEC, with the prototypical isolate being EPEC O127::H6 E2348/69 (EPEC

E69) is characterised by the presence of the 70-100 KDa virulence plasmid EAF

(EPEC adherence factor). EAF encodes for the type IV bundle-forming pilus (BFP)

which through filamentous bacteria-bacteria interaction gives rise to the

characteristic micro-colony formation of EPEC (Kaper, Nataro & Mobley, 2004). BFP

further binds N-acetyl-lactosamine containing receptors for initial adherence of EPEC

to host enterocytes (Croxen & Finlay, 2010). Although adherence, and more

importantly pathogenesis, is significantly abrogated in the absence of BFP

(Donnenberg, 2013; Saldana et al., 2009) both typical and atypical EPEC adherence

is further regulated by the Escherichia coli common pilus (ECP; Saldana et al.,

2009), long polar fimbriae (LFP), the non-fimbrial adhesins Paa, EHEC

Page 24: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

24

autotransporter encoding gene A (EhaA) and Iha as well as intimate attachment

through the T3SS (Clements et al., 2012; Frankel & Phillips, 2008).

EPEC along with the mouse restricted pathogen C. rodentium and EHEC belong to

the family of attaching and effacing (A/E) pathogens which share the common

feature of infection. A/E pathogens are characterised by their ability to induce A/E

lesions and intimate attachment; through host cytoskeletal rearrangement and

formation of a pedestal-like structure from accumulation of polymerised actin

beneath the attached bacteria and by the local effacement and destruction of the

enterocyte brush-border microvilli (Frankel & Phillips, 2008). A/E lesions are

mediated by the 35 Kb Locus of Enterocyte Effacement (LEE) Pathogenicity Island

(PAI) which encodes for a T3SS and the adhesin Intimin. Both of which are essential

for intimate attachment (Elliott et al., 1998). The LEE is regulated by the LEE

encoded regulator (Ler) located on the EAF plasmid (Kaper, Nataro & Mobley,

2004). Due to the presence of the T3SS, the mechanism of infection is more

complex for EPEC in comparison to the pathotypes addressed above. Translocation

of numerous bacterial proteins known as effectors, through the T3SS, into the host

cytoplasm of the gut enterocytes induce the characteristic intestinal A/E lesion and

manipulate host cell signalling to dampen immune and pro-apoptotic signalling to

promote survival and colonization (Croxen & Finlay, 2010). The localised

effacement, inflammation, ion channel disruption and resulting permeabilisation all

contributes to a watery diarrhoeal disease (Kaper, Nataro & Mobley, 2004). In

conjunction with the T3SS EPEC also encodes the 385 KDa Lymphostatin; LifA,

which inhibits the activation of lymphocytes (Kaper, Nataro & Mobley, 2004).

Furthermore EPEC expresses the SPATE EspC which like Pet targets the host

cytoskeleton and focal adhesion proteins to disrupt cell attachment (Clements et al.,

Page 25: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

25

2012; Vidal & Navarro-Garcia, 2008). EPEC E69 shares the common ancestor

EPEC O55:H7 str. with the A/E family member EHEC (Kyle et al., 2012). Whilst both

are LEE positive the EHEC pathotype differs from that of EPEC due to the absence

of the EAF plasmid and acquisition of the lambda-like bacteriophage genes encoding

Stx1/2 and the virulence plasmid pO157 (Kaper, Nataro & Mobley, 2004)

vi. EHEC

Due to Stx, EHEC O157::H7 is the leading cause of paediatric renal failure in

developed countries, with the main environmental reservoir established in dairy

cattle (Nataro & Kaper, 1998). EHEC is also a common cause of food borne

outbreaks in developed countries with the prominent serotype being O157:H7 in the

UK, USA and Japan or O26 and O111, elsewhere (Kaper, Nataro & Mobley, 2004).

The high rate of infectivity in an outbreak is due to the relatively low inoculum level

required to successfully colonize a human host which is fewer than 100 bacteria

(Croxen & Finlay, 2010). Not all strains of EHEC are Stx positive and not all Stx

positive strains are LEE positive. This has given rise to a ninth pathotype that is

extremely heterogeneous and is grouped by the presence of Stx and therefore

named Shiga toxin-producing E. coli (STEC). Interestingly most STEC that are LEE

negative, one exception being O103:H21, do not cause disease in humans,

underlining the importance of the T3SS in pathogenesis and adherence (Croxen &

Finlay, 2010). Furthermore EHEC encodes a second T3SS designated the EHEC

Type III secretion system 2 (ETT2). While the ETT2 is not fully function due to frame

shift mutagenesis or absence of integral structural components of the T3SS its

regulatory factors may impact on T3SS regulation and virulence (Zhou et al., 2014).

Initial adherence of EHEC to enterocytes of the large colon is carried out by an array

of adhesins common among other pathotypes and includes: Paa, Saa, Iha EhaA and

Page 26: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

26

LPF (Clements et al., 2012). EHEC also encodes colonization factors unique among

the inEPEC pathotypes including the LifA-like Toxin B (ToxB) and the E. coli factor

for adherence 1 (Efa1), the outer-membrane protein (OmpA), and the

autotransporter E. coli immunoglobulin binding protein G (EibG) (Clements et al.,

2012; Croxen & Finlay, 2010). EHEC also encodes multiple virulence factors on the

pO157 plasmid. This includes EspP, StcE a proteinase/mucinase which cleaves the

C1 esterase inhibitor of the complement pathway and is essential for colonization of

the large colon (Croxen & Finlay, 2010; Communication; Spandiaou) and the repeats

in toxin (RTX) family member EHEC toxin (Ehx) which is a pore forming haemolysin

and pro-apoptotic toxin (Bielaszewska et al., 2014).

C. rodentium shares 67% gene identity with both EHEC and EPEC. This includes the

LEE pathogenicity island encoding the T3SS and a similar cohort of T3SS effectors

(Collins et al., 2014). C. rodentium is a natural mouse pathogen; allowing the unique

opportunity to study A/E pathogen virulence and pathogenesis within its natural host.

1.1.2 Citrobacter rodentium

C. rodentium is an intestinal pathogen that colonises the murine large colon leading

to weight loss, colitis, soft stools and colonic hyperplasia also known as

transmissible murine crypt hyperplasia (TMCH; Collins et al., 2014). Initial adherence

of C. rodentium is not fully understood however colonization factors unique to C.

rodentium and homologous to those of EPEC/EHEC have been identified. C.

rodentium encodes the Citrobacter colonization factor (Cfc), a Type IV pilus which

shares homology with EPEC BFP (Mundy et al., 2004), a LifA/Efa1 homolog (Mundy

et al., 2005), the fimbrial adhesin K99-like factor involved in Citrobacter colonization

(Kfc), a homolog of the autotransporter AIDI-I adhesin named adhesin involved in

diffuse Citrobacter adherence (AdcA) (Hart et al., 2008) and the adhesin intimin

Page 27: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

27

(Collins et al., 2014). Cfc is essential for colonization and a strain lacking Cfc is

avirulent (Mundy et al., 2003); alternatively, deletion of either Kfc or AdcA has no

observed colonization effect in vivo (Hart et al., 2008). Intimate attachment of C.

rodentium and colonisation of the host is conferred similarly to that of EPEC/EHEC

through intimin and the T3SS dependent A/E lesion formation (Collins et al., 2014).

Through the acquisition of the PAI LEE C. rodentium encodes a T3SS which shares

homology and effectors with both EPEC and EHEC, including the intimin receptor Tir

(Collins et al., 2014). Furthermore C. rodentium encodes two T6SSs designated

CTS1 and CTS2 (Collins et al., 2014). Whether the C. rodentium T6SSs contribute to

virulence and are expressed and/or functional in vivo remains unknown. Non-T3SS

dependent virulence factors that are encoded by C. rodentium include the Class I

SPATE and EspC homolog C. rodentium class-1 serine protease (CrC1sp;

Vijayakumar et al., 2014) and the Class II SPATEs AdcA and the Pic homolog C.

rodentium class-2 serine protease (Cr-C2sp; Ruiz-Perez & Nataro, 2014). CrC1sp,

while predicted to belong to the Class I SPATEs and therefore predicted to cleave

spectrin, surprisingly has an anti-inflammatory and immuno-modulatory role in vivo

(Vijayakumar et al., 2014). A homologous T3SS, similar cohort of enterotoxins and

similar infection strategy make C. rodentium an ideal pathogen to study A/E

virulence in vivo. This has given rise to the C. rodentium mouse model.

i. The C. rodentium mouse model

C. rodentium infects mice through the oral-faecal route and is therefore under

laboratory conditions administered in liquid culture, by oral gavage (o.g.). The

outcome of infection is widely dependent on the mouse strains used. For instance,

C. rodentium infection of C3H/HeJ results in bacterial dissemination and high

mortality by day 10 post infection; in contrast, C57BL/6 mice rarely, if at all, succumb

Page 28: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

28

to infection (Mundy et al., 2005). This is most likely due to the difference and high

diversity of the host microbiota which can influence the susceptibility and immune

response to C. rodentium infection (Collins et al., 2014). In the early stages of

infection of C57BL/6 mice C. rodentium colonizes the caecal patch: the major

lymphoid structure of the caecum and by day 4 the distal colon (Collins et al., 2014).

Colonization of the colon reaches its peak at roughly day 8 post infection. Following

this, C. rodentium is rapidly cleared by the shedding of colonic epithelial

cells/enterocytes and the host mounted CD4+ T-cell dependent humoral immune

response (Collins et al., 2014). By day 14-21 post infection C. rodentium is

completely cleared and no longer detectable in the stool samples. While the

enterotoxins and the colonization factors described above play a significant role in

inEPEC and C. rodentium pathogenesis the T3SS and subsequent A/E lesion

formation is essential for EPEC, C. rodentium and EHEC virulence.

1.2 Bacterial Secretion Systems

Gram negative and positive pathogens use an array of specialised systems to deliver

bacterial toxins and proteins known as effectors into the extracellular milieu and host

cells, to aid in and promote colonization (Tseng, Tyler & Setubal, 2009). In order to

carry out this process, the need to overcome the complexity of transport across the

bacterial cell membranes/cell wall and host cell membrane is essential. Gram

negative bacteria have 6 main classes of dedicated secretion systems, numbered 1-

6, that transport effectors across the inner and outer membrane (As reviewed By

Tseng and Colleagues (Fig. 1.1; Tseng, Tyler & Setubal, 2009). While gram positive

bacteria share some similar systems they also have a unique system known as the

Type VII secretion system (T7SS). The Type II secretion system (T2SS) and Type V

Page 29: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

29

secretion system (TVSS), unlike the other specialised secretion systems, rely on an

initial general secretory pathway for delivery to the periplasmic space before

secretion.

1.2.1 The General Secretory Pathway

There are two general secretory pathways named the universal secretory pathway

(Sec) or Two-arginine (Tat) system (Tseng, Tyler & Setubal, 2009). The Sec system

requires a hydrophobic N-terminal signal sequence that transports proteins in an

unfolded state. The Sec pore is formed of three proteins SecY, E and G and the

system is driven by the cytoplasmic ATPase SecA. SecB, the chaperone protein,

recognises the N-terminal signal sequence and upon binding SecA initiates

translocation across the inner membrane. The Tat system recognises proteins via a

signal sequence motif consisting of the amino acid code SRR-X-FLK. Differing from

the Sec system, proteins translocated by the Tat system are done so in a folded

state across a protein gradient. Proteins are then further secreted by the Sec-

dependent T2SS and T5SS.

Figure 1 1 Secretion systems of Gram negative bacteria. A simple schematic depicting the machinery of the known Gram negative secretion systems adapted from Tseng and colleagues (Tseng, Tyler & Setubal, 2009). HM, host membrane; OM, outer-membrane; IM, inner-membrane; MFP, membrane fusion protein. ATPases coloured yellow, secretion apparatus blue and translocons red.

Page 30: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

30

1.2.2 The Type II and V secretion system

The T2SS machinery is composed of 12-15 secretin subunits. These form the multi-

meric protein pore and also anchor it to the outer-membrane. A further 11-14

uncharacterised membrane proteins are required for T2S. The ETEC virulence factor

LT is delivered in a T2SS dependent manner. A homologous system in EHEC is

required for the secretion of StcE a proteinase/mucinase which is essential for

colonization of the large colon (Communication; Spandiaou). Typical EPEC also

have a functional T2SS; secreted and surface associated lipoprotein from E. coli

(SsIE) is involved in the progression of micro-colonies to mature biofilm formation

(Baldi et al., 2012).

The T5SS is divided into 3 major subclasses: a-c. T5SS proteins consist of an N-

terminal passenger domain carrying the N-terminal signal sequence for Sec

transport and a conserved β-barrel C-terminal domain involved in translocation. In

the T5aSS the C-terminal forms a pore while the N-terminal is cleaved off and

translocated into the extracellular space as a hydrolytic enzyme or toxin, giving rise

to the name auto-transporters. The T5cSS forms a single β-barrel by tri-meric

formation of the secreted protein; alternatively, the T5bSS consists of pairs of

proteins (also known as Two partner system; TPS) where one has the β-barrel and

the other is secreted. The adhesion AIDA-I of DEAC is a T5aSS secreted protein

(Tseng, Tyler & Setubal, 2009). Furthermore EPEC E69 delivers EspC a serine

proteinase that targets Focal adhesion Kinase (FAK) and spectrin and is linked to in

vitro cell cytotoxicity and detachment (Navarro-Garcia et al., 2014). EHEC EspP, a

homolog to EspC, has been linked to the disruption of multiple host pathways

including the complement cascade, haemostasis and biofilm formation (Weiss &

Brockmeyer, 2012). Unlike the T5SS and T2SS the Type I, III, IV and VI systems are

Page 31: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

31

fundamentally single step; transporting effectors directly across the inner and outer-

membrane.

1.2.3 The Type I Secretion system

The T1SS consists of three main domains: the outer-membrane proteins (OMPs),

membrane fusion proteins (MFPs) and the ATP-binding cassette (ABC) transporters.

The OMP TolC forms the outer-membrane pore and connects to the trans-

periplasmic MFPs; the MFPs in turn are structural proteins which span the

periplasmic region creating a tunnel between the OMPs and the ABC-transporters.

The ABC transporter HlyB drives translocation in an ATPase dependent manner and

secretion is dependent on the recognition of a specific leader signal sequence. HlyA,

a pore forming toxin (Donnenberg, 2013), is a well characterised T1SS secretion

system effector that is encoded by uropathogenic E. coli (Tseng, Tyler & Setubal,

2009). C. rodentium encodes multiple putative T1SSs shared by other enteric

pathogens including Salmonella entrica Thyphi (Petty et al., 2010). The T1SS and its

contribution to virulence in A/E pathogens is poorly characterised.

1.2.4 The Type VI secretion system

The type VI secretion system is one of the more recently discovered specialised

secretion systems of Gram negative bacteria. While the system remains poorly

characterised it was identified due to its homology to the T4SS of Legionella

pneumophila and consists of an injectisome based on the T4 bacteriophage-like tail

(Petty et al., 2010). The injectisome of the T6SS is anchored to the outer-membrane

by the membrane complex composed of the inner membrane proteins TssL, TssM

and TagL and the outer-membrane lipoprotein TssJ. T6S is powered by the

cytoplasmic chaperone ATPase ClpV (Felisberto-Rodrigues et al., 2011).

Remarkably this system is not only dedicated to the delivery of effector proteins to

Page 32: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

32

eukaryotic cells to promote full virulence and pathogenesis, but unlike other systems,

it can also deliver effectors to prokaryotic cells in order to out-compete neighbouring

bacteria (Felisberto-Rodrigues et al., 2011; Tseng, Tyler & Setubal, 2009). While

EPEC does not encode a T6SS it is present in EAEC (Felisberto-Rodrigues et al.,

2011), EHEC and C. rodentium (Gueguen & Cascales, 2013; Petty et al., 2010). C.

rodentium encodes two T6SSs on cluster 1 (CTS1) and cluster 2 (CTS2). CTS2

contains a confirmed frameshift in the ATPase TssM (also known as intracellular

multiplication factor F) which is required to form the TssM and TssL complex and

energize secretion of T6SS (Petty et al., 2010). CTS1, which shares high homology

to the T6SS of Enterobacter cancerogenus and also shares similarity to the SPI-6 of

Salmonella enterica, encodes a fully functional T6SS in a chaperone-usher fibrial

biogenesis cluster (Gueguen & Cascales, 2013; Petty et al., 2010). C. rodentium

employs the T6SS as an anti-bacterial tool to deliver effectors to out-compete

neighbouring bacteria in vitro (Gueguen & Cascales, 2013). Further studies are

required however, to identify the effectors responsible and fully understand the role

of this system in infection and its impact on virulence and colonization of the host.

1.2.5 The Type IV secretion system

Although the T4SS shares a similar inner membrane complex to the T2SS it does

not rely on any general secretory pathway and can transport proteins in one step

(Tseng, Tyler & Setubal, 2009). The T4SS is unique due to the fact that it can

translocate and uptake nucleic acids as well as translocate proteins (Tseng, Tyler &

Setubal, 2009; Christie, Whitaker & González-Rivera, 2014). The T4SS is subdivided

into the conjugation system, which is required for the inter-bacteria transfer of DNA,

uptake of DNA and natural competence and the effector translocator system which is

dedicated to the translocation of bacterial effectors to manipulate host cell physiology

Page 33: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

33

(Christie, Whitaker & González-Rivera, 2014). Natural competence and uptake of

DNA will not be covered here and are reviewed elsewhere (Johnsborg, Eldholm &

Havarstein, 2007). The effector translocation system of the T4SS consists of 4 major

components: the type 4 coupling protein (T4CP), the inner-membrane complex

(IMC), the outer-membrane complex (OMC) and the pilus. The T4CP is the ATPase

and effector receptor that couples the substrate to the secretory machinery. Subsets

of effectors contain C-terminal signal sequences for the T4CP receptor while others

can be secreted by the Sec system. The IMC drives translocation across the inner-

membrane and the OMC allows passage through the periplasmic space and

translocation through the outer-membrane (Christie, Whitaker & González-Rivera,

2014). The pilus is essential for cell to cell contact and comprises the needle-like

structure to which the translocon/pore forming protein is attached (Gauthier, Thomas

& Finlay, 2003). A/E pathogens do not encode a T4SS; however they do encode a

structurally similar secretion system known as the T3SS.

1.2.6 The Type III Secretion System

The T3SS, a molecular syringe, which closely resembles the bacterial flagellin

apparatus, enables injection of effectors directly across the bacterial membranes into

the host cell cytoplasm. Once translocated the effectors mimic host cell proteins to

manipulate the host physiology, to aid in bacterial colonization and survival. As many

pathogens encode and rely on a T3SS or multiple T3SS in order to successfully

colonize the host, the T3SS is one of the best characterised and studied secretion

systems of pathogenic bacteria. This is true for many enteric pathogens including

Salmonella spp., EPEC/EHEC, C. rodentium, Yersinia spp. and Shigella spp. where

a functional T3SS is essential for full virulence. A/E pathogens share a highly

homologous T3SS apparatus and effector arsenal which is encoded by the LEE.

Page 34: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

34

i. The Locus of Enterocyte Effacement Pathogenicity Island

The 35 Kb chromosomal pathogenicity island of EPEC E69 and EHEC str. EDL933

encodes genes for the T3SS apparatus (E. coli secretion gene; Esc), chaperones

(Chaperone for E. coli secreted protein; Ces), regulators (Ler), the adhesion Intimin

and its translocated receptor Tir as well as 5 other effectors (E. coli secreted protein;

Esp) EspF, EspZ, MAP, EspH and EspG (Elliott et al., 1998; McDaniel et al., 1995).

The genes are encoded on the major operons termed LEE1-5 (Fig. 1.2).

ii. Biogenesis of the EPEC and the EHEC T3SS

The archetypal T3SS of EHEC/EPEC is composed of a multi-ring structure (basal

apparatus and outer-membrane ring which in is bridged by EscJ) and a needle-like

structure onto which the translocon is assembled (As depicted in Fig. 1.3)

Figure 1 2 The locus of enterocyte effacement. Gene organization, and assigned function, along the locus of enterocyte effacement pathogenicity Island of the attaching and effacing pathogen EHEC.

Page 35: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

35

The basal apparatus is composed of EscD and EscQ-V in the inner membrane while

the outer-membrane ring is composed of EscC which is secreted across the inner-

membrane by the Sec-system (Tree et al., 2009). The T3SS ATPase EscN is

anchored to the inner membrane basal apparatus and supplies the energy for T3S

export and protein translocation. EscF (EprI in EHEC) extends from EscI (the inner

rod) and the basal apparatus across the outer-membrane to form the T3SS needle

on to which EspA translocon extends further to the host cell membrane. EspA is

capped by the pore forming complex which is composed of EspB/D to facilitate

effector translocation (Tree et al., 2009). In order to conserve energy the T3SS is

Figure 1 3 The T3SS translocon of EPEC. An illustrated structure of the EPEC T3SS. EscQ-V and EscD form a multi-ring structure known as the basal apparatus and EscC forms the outer-membrane ring structure, whilst EscJ acts a bridge between the membrane associated rings. Secretion is driven in an ATP-dependent manner requiring the ATPase EscN. EscI (EprI in EHEC) froms the inner needle on to which EscF (EprJ in EHEC) extends a needle-like structure onto which the translocon (EscA) is assembled. EscA extends out towards the host cell, capped by the EspB/D pore, where the effectors are secreted. IM, inner-membrane; OM, outer-membrane; PG, peptidoglycan layer; EM, eukaryotic membrane. Adapted from Pallen and colleagues (Pallen et al., 2005).

Page 36: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

36

tightly regulated at the level of expression and controlled by specific environmental

signals and genetic regulators for a coordinated attack (Tree et al., 2009)

iii. Regulation of the T3SS

While the regulation of the LEE varies between EHEC and EPEC the fundamental

regulatory factors are the same. Both pathotypes encode the major LEE-encoded

regulator (Ler) which responds to certain environmental and stress signals including

bicarbonate, hormones (norepinephrine and epinephrine), pH, DNA damage and

temperature, to up-regulate expression of the LEE (Tree et al., 2009). Ler shares

high homology to a known global regulator (H-NS) which binds to A-T rich regions,

creating loops in the DNA structure, to supress expression. Interestingly Ler directly

counters the activity of H-NS, releasing it from the strand to promote expression of

LEE2-5 (Tree et al., 2009). The ability of the bacteria to be able to communicate to

neighbouring bacteria within its own and surrounding population; meanwhile also

sensing the host, plays a significant role in its decision making at the level of

expression. This is known as quorum sensing.

Many quorum sensing systems that directly regulate the T3SS have been in

characterised in EPEC and EHEC, respectively (Tree et al., 2009). Both EPEC and

EHEC through the quorum sensor QseA, detect host levels of Epinephrine which

triggers the expression of Ler. EPEC also encodes the LuxS quorum sensor, which

through autoinducer-3 (AI-3) and Epinephrine, also activates the expression of Ler

(Sperandio et al., 2003). Futhermore, EHEC through the two component system

QseEF and QseG up-regulates the expression of the T3SS effector EspFu and

controls Tir translocation, respectively (Tree et al., 2009). A host of further regulators

control T3SS expression and secretion including the Plasmid encoded regulator

Page 37: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

37

(Per) and the Integration Host Factor (IHF) (Sircili et al., 2004; Yona-Nadler et al.,

2003)

PerA-C is encoded on the EAF plasmid of atypical EPEC and directly up-regulates

the expression of the adhesion and micro-colony formation factor BFP, as well as

Ler, in response to pH (Sircili et al., 2004; Shin et al., 2001). EHEC does not carry

the EAF plasmid and alternatively encodes seven homologs of Per on OI’s. EHEC

Pers: PchA-E and PchX-Y activate the expression of T3SS effectors found on

OIs/outside the LEE (Tree et al., 2009). As mentioned previously, the T3SS and

flagellum share the same apparatus; therefore, it is hypothesised that expression

and biogenesis of flagella could sterically inhibit the T3SS and secretion (Tree et al.,

2009). It was observed that under laboratory conditions used to induce the

expression of the T3SS, that EPEC becomes non-motile/non-flagellinated (Yona-

Nadler et al., 2003). This was due to IHF which upon activation down regulates the

expression of FlhDC which are positive regulators of the flagellar regulon (Yona-

Nadler et al., 2003). Co-ordinately, Ler activates GrlA which triggers a switch from

flagella to T3SS, reinforcing the level of sophistication of regulation of the T3SS

(Tree et al., 2009).

A further level of regulation that exists but is poorly understood is the switch from

biogenesis to effector secretion. This requires the presence of the pore forming

complex EspB/D. It is thought to be an extra step to conserve energy and to

effectively deliver effectors to the host cytoplasm and not into the extracellular milieu.

Currently, it is hypothesised that this trigger may in fact be a change in ion

concentration due to the exposure to the host cytoplasm upon connection (Tree et

al., 2009). Once secreted the effectors modulate the host cell signalling and

physiology to aid in colonization. The host cell targets and binding partners, as well

Page 38: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

38

as the function of these effectors, have been the centre of many studies over the

past decade.

1.3 Effector proteins and Manipulation of host cell signalling

The best characterized LEE effector, conserved among A/E pathogens, is Tir. Tir is

required for intimate attachment and for pedestal formation, a hallmark of successful

T3SS translocation in vitro (Frankel & Phillips, 2008) (Fig. 1.4A). Translocated Tir

localizes to the host cell surface where it can interact with the bacterial outer-

membrane protein Intimin (Kenny et al., 1997) (Fig. 1.4A). Tir is further

phosphorylated by host tyrosine kinases resulting in the recruitment of Nck

((Gruenheid et al., 2001), neural Wiskott–Aldrich syndrome protein (N- WASP) and

actin-related protein 2/3 (ARP-2/3) which stimulates actin nucleation and

polymerisation (Kalman et al., 1999). This is the driving force behind membrane

protrusion and pedestal formation (Fig. 1.4A-B) (Kenny et al., 1997). EHEC pedestal

formation is similar to that of EPEC with a slight difference due to the fact that Tir is

not phosphorylated. Instead Tir recruits insulin receptor tyrosine kinase substrate

(IRKTS) which forms a complex with the translocated effector Tir cytoskeleton-

coupling protein (TccP). TccP recruits N-WASP resulting in actin polymerisation as

described for EPEC pedestal formation (Garmendia et al., 2004). In addition to the

conserved effectors of the LEE EPEC, EHEC and C. rodentium encode effectors that

have been identified in other pathogenicity islands with many having been

characterised (Wong et al., 2011). To date, 21 conserved effectors have been found

in all A/E pathogens (Iguchi et al., 2009). However, the total number of the non-LEE-

encoded effectors (nle) can vary significantly between EPEC/EHEC strains, altering

the mechanism of infection.

Page 39: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

39

Translocation of A/E pathogen T3SS effectors, have been reported to be responsible

for the modulation of Rho-GTPases (Wong et al., 2012), actin re-organisation and

pedestal formation (as shown in Fig. 1.4 and reviewed by Frankel et al., 2008),

disruption of mitochondrial integrity (Nougayrede & Donnenberg, 2004), regulation of

cell survival and apoptosis (Hemrajani et al., 2010; Dean et al., 2010; Baruch et al.,

2011; Samba-Louaka et al., 2009), inhibition of phagocytosis (Marches et al., 2008)

and modulation of the host inflammatory and immune response (Gao et al., 2013;

Raymond et al., 2013; Baruch et al., 2011; Pearson et al., 2011; Raymond et al.,

2011; Nadler et al., 2010; Newton et al., 2010; Vossenkämper et al., 2010; Gao et

Figure 1 4 EPEC infection is characterised by pedestal and A/E lesion formation in vivo. (A) Pedestal formation is initiated by EPEC through the translocation of Translocated intimin receptor (Tir). Tir is displayed on the eukaryotic cell surface were it binds the bacterial outer-membrane protein Intimin. Tir becomes phosphorylated by the host Tyrosine kinases and in a Nck dependent manner recruits neural Wiskott–Aldrich syndrome protein (N-WASP) and actin-related protein 2/3 (Arp-2/3) leading to actin nucleation and polymerisation; forming the pedestal. EHEC pedestal differs as Tir is not phosphorylated and in an Nck-independentmanner it recruits insulin receptor tyrosine kinase substrate (IRTKS) and through Tircytoskeleton-coupling protein (TccP) then merges with the EPEC pathway by recruiting N-WASP. (B) An electron micrograph of human intestinal cultured mucosa infected with EPEC (Chen et al., 2005). Membrane protrusion and a cup-likestructure can be identified beneath the surface of the bacteria which is a hallmark of actin and cytoskeleton rearrangement and pedestal formation. Microvilli effacementand destruction is also noted which is indicative of A/E lesion formation.

Page 40: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

40

al., 2009). Little is still understood about the biological function of numerous effectors

and how they manipulate the host immune response during infection.

1.4 The Host innate immune response

The Inflammatory response is essential in innate immune signalling and the early

host response to infection (Mogensen, 2009). The innate immune system is the first

line of defence against invading microbes and through pattern recognition receptors

(PRRs) detects distinct conserved pathogen-associated molecular patterns (PAMPs)

(Mogensen, 2009). Recognition of PAMPs through PRRs triggers an intracellular

signalling cascade leading to the expression and secretion of pro-inflammatory

molecules (Mogensen, 2009). The secretion of pro-inflammatory molecules in turn

leads to the recruitment and activation of cells of the innate immune system

including: dendritic cells, neutrophils and phagocytic macrophages. This induces

further inflammatory signalling and the release of pro-inflammatory molecules; thus

allowing priming and activation of the adaptive immune system and the ability to fight

and clear the infection (Mogensen, 2009). Detection of PAMPs and danger

associated molecular patterns (DAMPs) is regulated and controlled through both

intracellular PRRs and extracellular PRRs.

1.4.1 Pattern recognition receptors and host-pathogen detection

The host recognises stimuli through specific PRRs of which the best characterised

are the Toll-like receptors. Each Toll-like receptor responds to a different set of

specific PAMPs/DAMPs, with Toll-like receptor 3 (TLR-3), Toll-like receptor 4 (TLR-

4) and Toll-like receptor 5 (TLR-5) which detect dsDNA, Lipo-polysaccharide (LPS)

and flagellin, respectively; being the most relevant to Gram negative bacterial

infection. The TLRs are able to sample the extracellular milieu and endosomal

compartments for PAMPS, triggering an intracellular signalling cascade leading to

Page 41: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

41

the transcriptional activation of inflammatory cytokines; activating the immune

response.

1.4.2 The Toll-like receptors

TLR-4, an extracellular receptor found on the cell surface of myeloid cells as well as

non- hematopoietic cells, is stimulated by LPS. Binding of LPS through the co-

receptors CD14, LPS-binding protein (LBP) and lymphocyte antigen 96 (Ly-96/MD-2)

triggers oligomerisation of TLR-4 resulting in recruitment of the intracellular adaptor

protein: Myeloid differentiation primary response protein 88 (MyD88). MyD88 in turn

activates IL-1 receptor-associated kinase-4 (IRAK-4) which recruits IRAK-1 (Fig. 1.5)

(Li & Verma, 2002). TNF receptor-associated factor 6 (TRAF6), forms a complex

with IRAK-1 and procaspase-4 (Nickles et al., 2012; Lakshmanan & Porter, 2007; Li

& Verma, 2002) and upon recruitment of the ubiquitin-conjugating enzyme 13

(Ubc13) and ubiquitin-conjugating enzyme E2 variant 1 isoform A (Uev1A) is

modified by K63-linked polyubiquitylation (Lim & Staudt, 2013). Ubiquitlyated TRAF6

is recognised by the ubiquitin binding domain of TGF-β activated and MAP3K7

binding protein 2/3 (TAB2/3) which is found in complex with transforming growth

factor-β-activated kinase 1 (TAK1; Fig. 1.5). At this stage the complex moves to the

cytoplasm where the IĸB Kinase (IĸK) subunit IκKγ (NEMO) is recruited through a

ubiquitin binding domain and phosphorylated by TAK1. In parallel ubiquitlyated

TRAF6 recruits the LUBAC complex which binds and activates NEMO by methionine

linked linear ubiquitin (Emmerich et al., 2013). Downstream, IĸK phosphorylates the

Nuclear factor-Kappa B (NF-κB) inhibitor alpha (IĸBα), signalling it for proteasomal

degradation. Importantly, under resting conditions, IĸBα is in a complex with the NF-

ĸB subunits p65 and p50, restricting both to the cytoplasm (Li & Verma, 2002)).

Upon IĸBα degradation p65 translocates to the nucleus resulting in the expression of

Page 42: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

42

target genes, promoting cell survival and inflammation (Li & Yang, 2011).

Alternatively active TAK1 activates the transcription factors Cyclic-AMP responsive

element-binding protein 1 (CREB1) and Activator Protein 1 (AP-1) via Mitogen-

activated protein kinase (MAPK) kinases 3/6 (MKK3/6) and MAPK kinase 4/7

(MKK4/7), respectively (Fig. 1.5). MKK3/6, via a set of serine threonine kinases

known as p38 MAPKs, promotes the nuclear translocation of CREB1; while MKK4/7

signal via a different branch through phosphorylation of c-Jun N-terminal kinases

(JNKs; Lim & Staudt, 2013). This promotes nuclear translocation of the transcription

factor family AP-1 (Lim & Staudt, 2013; Dunne & O'Neill, 2003). The target genes of

CREB1 are involved in cell differentiation and survival (Impey et al., 2004); whereas,

AP-1 up-regulates the expression genes involved in cell survival and proliferation as

well as inflammation (Li & Yang, 2011). This results in cross talk with the NF-ĸB

pathway and co-combinational propagation of a pro-inflammatory signalling cascade

to transcriptionally re-programme and activate cells of the immune system via

expression of cytokines (Interleukin-1β; IL-1β), chemokines (Interleukin-8; IL-8),

adhesion molecules and metalloproteases (Li & Yang, 2011). At the level of TAK1

activation the intracellular signalling pathway converges with that of the Tumour

necrosis factor receptor (TNFR) and the Interleukin-1 receptor (IL-1R) pathways (Fig.

1.5; Dunne & O'Neill, 2003).

Page 43: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

43

Figure 1 5 The TLR-4 and TNFR1 intracellular signalling cascades. LPS stimulation of TLR-4 drives an intracellular signalling cascade leading to the nuclear translocation of the transcription factors AP-1, CREB1 and NF-κB. Similar TNFα stimulation of TNFR1 activates an intracellular signalling cascade that converges with the TLR-4 pathway at the stage of TAK1 activation. Nuclear translocation of AP-1, CREB1 and NF-κB leads to cell proliferation, cell survival and inflammation. The more information please see text.

Page 44: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

44

TNFα is commonly used as tool to activate and study NF-κB nuclear translocation in

tissue culture assays. Stimulation of the canonical TNFR signalling cascade leads to

TNFR1 aggregation and, through homotypic interaction of their death domains (DD),

the recruitment of the cytoplasmic adaptor Tumour necrosis factor receptor type 1-

associated death domain protein (TRADD) and the serine-threonine kinase Receptor

interacting protein 1 (RIP1; Chaudhary et al., 2000). RIP1 signalling is dependent on

TRAF2/5 where it converges with the TLR-4 pathway to activate TAK1 (Dunne &

O'Neill, 2003; Chaudhary et al., 2000). Caspase-4 has been reported to be essential

for this step (Nickles et al., 2012). Alternatively TNFα stimulates a non-canonical

caspase—8 dependent cascade through the recruitment of the Fas-associated

death-domain protein (FADD) by TRADD. Caspase-8 through an N-terminal DD

binds FADD and in a TRAF2/RIP1 dependent manner activates the serine-threonine

kinase NIK. NIK directly activates NEMO, again converging with the TLR-4 pathway

to activate NF-κB (Chaudhary et al., 2000). In conjunction with the extracellular

PRRs both myeloid and non-myeloid cells utilise a cohort of intracellular PRRs to

detect distinct antigens.

1.5 Intracellular detection via PRRs

The intracellular PRRs are divided into two major classes consisting of the RNA

sensing RIG-I-like helicases (RHLs)/DNA sensors and the Nucleotide-binding

oligomerisation domain-containing (NOD) like proteins (NLRs), as reviewed by

Mogensen (Mogensen, 2009).

1.5.1 RHLs and DNA sensors

Briefly, the two well characterised RHLs include Melanoma associated protein 5

(MDA5) and RIG-I. As intracellular receptors they recognize the 5'-triphosphate

moiety and dsRNA structures of invading pathogens, which is distinctly non-self.

Page 45: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

45

With this in mind they are usually associated with viral infection. Their activation

leads to the initiation of transcription with both receptor signalling pathways

converging to mediate transcription of pro-inflammatory cytokines via NF-ĸB and AP-

1 as well as Type I Interferons (IFNs) via Interferon regulatory factor (IRF) 3 and 7.

The identity of the cytoplasmic dsDNA sensors is less well characterised, however

the best characterised response is via the TBK1 and IRF3 signal cascade, triggering

transcriptional induction of type I IFN genes. Detection of dsDNA, which should

usually be compartmentalised to the nucleus if “self DNA”, activates the sensors:

absent in melanoma 2/ interferon inducible protein 2 (AIM2) and 'DNA-dependent

activator of IRFs (DAI/ZBP1). Activation of DAI converges with the RHLs signalling

cascade stimulating IRF3, IRF7 and NF-ĸB (Yoneyama & Fujita, 2007), whereas

AIM2 is activated to form an inflammasome complex driving a signalling cascade

that is similar to that of the NLRs, which is described below.

1.5.2 The NODs and NOD-like receptors

There are at least 22 genes encoding NLRs identifiable by their central nucleotide-

binding and oligomerisation (NACHT) domain. Whilst the NACHT domain is common

among all NLRs the existence of 3 distinct NACHT domains has separated the NLRs

into 3 groups:

1. NODs (NOD1-2, NOD3/NLRC3, NOD4/NLRC5, NOD5/NLRX4, CIITA (class 2

transactivator)

2. The NOD-like receptor family, pyrin domain containing proteins (NLRPs) 1-14

(also known as NACHT, LRR and PYD domains-containing proteins; NALPs)

3. IPAFs/NLRC4 (Ice protease-activating factor; NLR family CARD domain-

containing protein 4) and neuronal apoptosis inhibitory protein (NAIPs)

Page 46: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

46

These NLRs are further characterised depending on the presence of further

functional domains including: leucine-rich repeat domain (LRR) required for ligand

sensing and the caspase activation and recruitment domain (CARD) and/or the pyrin

domain (PYD). Both CARD and PYD are essential for homotypic protein-protein

interaction for downstream signalling (Mogensen, 2009).

i. NODs: Pathogen Detection and Signalling

The best characterised NODs are NOD1 and NOD2. NOD1 and NOD2 oligomerise

upon stimulation by the bacterial cell wall components meso-diaminopimelic acid

(DAP) and muramyl-dipeptide (MDP), respectively (Schroder & Tschopp, 2010).

Through homotypic CARD-CARD interaction NOD1/2 recruits Receptor-interacting

serine/threonine-protein kinase 2 (RIP2) which through auto-phosphorylation recruits

a signalling complex consisting of TRAF 4, 5 and 6 as well as Ubc13/Uev1A and

Inhibitor of apoptosis protein 1/2 homolog C (cIAP1/2; Schroder & Tschopp, 2010;

Xie, 2013). cIAP1/2 and Ubc13/Uev1A catalyse the K63-Linked poly-ubiquitination of

RIP2 which further recruits IκKγ and the TAK1/TAB2/3 complex (Xie, 2013). NF-ĸB

and MAPK activation follow as described previously. NOD1 and NOD2 can also

stimulate transcription of the type 1 IFNs although through distinct mechanisms.

NOD1 recruits TRAF3 and signals through IFNβ dependent IRF7 alternatively NOD2

acts as a receptor for ssRNA to signal through IRF3 (Xie, 2013). NOD5 and 6 are

less well characterised but may be involved in promoting the production of reactive

oxygen species (ROS); whilst the roles of NOD3/4 are still unknown (Schroder &

Tschopp, 2010). The NOD CIITA was the first NLR to be classified and unlike the

others it has a transcription co-factor activity that regulates the expression of the

Major histocompatibility complex II (MHCII) and MHCI and therefore regulates

Page 47: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

47

antigen presentation (Schroder & Tschopp, 2010). NLRs can be functionally

classified into two distinct groups: those that regulate the formation of a mutli-meric

protein complex known as the inflammasome (AIM2, NLRP1b, NLRP3, NLRP7 and

NLRC4; Rathinam et al., 2012) and those that down-regulate immune signalling

(NLRP10, NLRP12, NLRC3, NLRP6-7 and NLRXC1; Allen, 2014; Wlodarska et al.,

2014; Damm, Lautz & Kufer, 2013; Radian et al., 2013; Anand et al., 2012)

1.6 NLRs and the Inflammasome

The inflammasome is a relatively newly characterised intracellular multi-protein PRR-

complex which forms in response to multiple stimuli and insults including uric acid

crystals, ultraviolet radiation (UV), ATP, DNA/RNA, flagellin, pore forming toxins,

lipoprotein and LPS/bacterial infection to activate the cysteine protease caspase-1

(Vladimer et al., 2013). Active caspase-1 further processes its substrates pro-

interleukin-18 (pro-IL-18) and pro-interleukin-1β (pro-IL-1β) inducing their secretion

independent of the classical Golgi/ER pathway (Mariathasan & Monack, 2007; Scott

& Saleh, 2007). Furthermore, caspase-1 induces a form of programmed

inflammatory cell death known as pyroptosis; releasing DAMPs as a host warning

system in response to infection/ sterile stressors (Miao et al., 2010a). The NLR

responsible for inflammasome formation varies depending on the cell type and the

stimulus.

1.6.1 The NLRC4 inflammasome

NLRC4 is a component of the intracellular receptor that is dedicated to detecting

flagellin and the rod/needle components of the T3SS of enteric pathogens; as well as

the T4SS (Yang et al., 2013; Zhao et al., 2011; Miao et al., 2010b). Detection leads

to the recruitment and activation of caspase-1 in an Asc independent-manner,

leading to IL-1β/IL-18 secretion and pyroptosis of macrophages (Miao et al., 2008).

Page 48: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

48

i. NLRC4 detection of EPEC E69 and C. rodentium ligands

Unlike the other NLRs NLRC4 does not detect the PAMP/DAMP directly and instead

works in concert to associate with the Neuronal apoptosis inhibitory protein (NAIP)

(Lage et al., 2014). Humans only encode one NAIP known as hNAIP; whereas there

have been at least 4 NAIPs in mice that have been identified and characterised for

their bacterial ligands. This includes NAIP1, NAIP2 and NAIP5/6 which bind the

T3SS needle, T3SS rod and flagellin respectively (Lage et al., 2014). The presence

of only 1 NAIP in humans may restrict the spectrum of bacterial ligands that are

detected by NLRC4 compared to mice.

Due to the availability of knockout cell lines, activation of NLRC4 has been well

studied and characterised in the context of Salmonella, EPEC and C. rodentium

infection of immortalised mouse macrophages (iMM). The Salmonella infection of

mouse macrophages elicits an NLRC4 response via NAIP1 detection of the

salmonella needle (PrgI) and NAIP2 2 detection of the inner rod (PrgJ) (Yang et al.,

2013). The EPEC/ C. rodentium homolog to PrgI; EscF, is not recognised by NAIP1,

probably due to their low level of homology (Yang et al., 2013). Alternatively infection

of iMMs with an EPEC ΔescI (the functional homolog of PrgJ) strain abolishes

caspase-1 cleavage in response to infection (Zhao et al., 2011; Miao et al., 2010b).

Furthermore caspase-1 cleavage and NLRC4 activation are dependent on NAIP2

(Zhao et al., 2011). Interestingly the flagellin negative strain (ΔfliC) of EPEC does not

impact on NLRC4 and caspase-1 activation; therefore it can be concluded that

EPEC flagellin does not contribute to inflammasome activation in vitro (Zhao et al.,

2011).

Similar to iMM infection, Salmonella infection of human macrophages activates the

NLRC4-hNAIP-dependent inflammasome. hNAIP has been recently shown to detect

Page 49: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

49

PrgI; therefore, acting as the functional homolog to murine NAIP1 (Yang et al.,

2013). It has been confirmed in a number of cell lines (THP.1, U937, A549) that

EscF is not detected by hNAIP and again does not activate NLRC4 (Yang et al.,

2013; Zhao et al., 2011). In fact 2 amino acids shown to be essential for the

detection of PrgI by hNAIP are not present in EPEC E69 or C. rodentium EscF (Yang

et al., 2013). Although EPEC EscI is required for NAIP2-NLRC4 activation it has not

been tested in human macrophages. Due to the fact that humans only encode one

NAIP and it has been shown to function as NAIP1 it is unlikely EPEC EscI is

detected by hNAIP; however, further studies are required to validate this hypothesis.

It is also important to note that while EscI is required for NLRC4 activation during

infection of iMMs it has not been shown to be directly detected itself by NAIP2;

although this is the case for PrgJ. The deletion of EscI prevents the biogenesis of the

T3SS and subsequently prevents the secretion of T3SS components EscF, EspA,

EspB, EspD and the T3SS effector proteins. The bacterial ligand detected by NAIP2

during EPEC E69 infection could be a number of proteins and remains to be

confirmed. While a role for NLRC4 in EPEC E69 infection remains controversial the

NLR NLRP3 is essential for inflammasome activation in iMMs in response to EHEC

and C. rodentium infection.

1.6.2 NLRP3

NLRP3 differs to other NLRs due to the wide cohort of stimuli to which it responds to

including: alum, ATP, MDP, LPS, nucleic acids, nigericin (bacterial toxin), uric acid

crystals, amyloid-β and UV irradiation to a name a few. This has led to the

hypothesis that NLRP3 itself is activated by a signal common to the above

mentioned stimuli rather than having the capability to recognize such a versatile

range of insults directly (Abderrazak et al., 2015). Interestingly inhibition of ROS or

Page 50: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

50

K+ efflux abrogates the activation of the NLRP3 dependent inflammasome to many

stimuli (Abderrazak et al., 2015). NLRP3 is composed of a tripartite structure

consisting of an N-terminal PYD followed by a NACHT/NBD and finally a LRR

domain, which all play an essential role in its activity and inflammasome assembly as

reviewed by Abderrazak (Abderrazak et al., 2015).

i. The canonical inflammasome

NLRP3 is held in an inactive form by the LRR domain which holds NLRP3 in an

auto-repressive state. Upon stimulation and activation the auto-repressive state is

relaxed exposing the NBD and PYD domain of NLRP3. The exposed PYD recruits

apoptosis associated speck-like protein containing a CARD (Asc) which via its CARD

domain directly recruits pro-caspase-1 to assemble the inflammasome.

Oligomerisation of Asc by self-association further increases procaspase-1

recruitment and size of the multi-meric complex; this is referred to as a “speck” due

to the fact that usually a single strong foci of inflammasome assembly is observed in

a single cell upon activation. Upon recruitment procaspase-1 undergoes self-

proteolytic cleavage and oligomerisation in to the active state; which in turn

processes pro-IL-1β/IL-18 and stimulates pyroptosis. This is referred to as the

canonical inflammasome; alternatively, a non-canonical NLRP3 dependent

inflammasome is activated by bacterial infection and the detection of intracellular

LPS by the PRR caspase-4 in humans and caspase-11 in mice (Shi et al., 2014;

Kayagaki et al., 2011). This in turn activates the assembly of the inflammasome and

caspase-1 activation in human and mouse macrophages (Shi et al., 2014; Sollberger

et al., 2012; Kayagaki et al., 2011).

Page 51: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

51

1.6.3 Caspase-4

Caspase-4 is a cysteine protease and member of the inflammatory caspases and

interleukin-1 beta converting enzyme subfamily (Martinon & Tschopp, 2007). It is

ubiquitously expressed in various tissues with exception of the brain and shares 51%

homology amino acid identity with caspase-1, 75% homology with caspase-5 and

48% homology with caspase-12.

i. Structural activation of caspase-4

Inflammatory caspases are characterised by their long pro-domain containing the

CARD (Sollberger et al., 2012). Caspase-4 contains one long pro-domain (p22) and

two small functionally active domains p20 and p10. Cleavage produces a p30

product which is further processed into the single subunits upon cleavage of the

inter-linker domain (Fig. 1.6; Karki, Dahal & Park, 2007). Caspases can be

characterised by the mechanism in which they become active. Initiator caspases

which include caspases 2, 8, 9 and 10, which contain a long pro-domain, are

induced by adaptor mediated oligomerisation in large protein complexes eg. the

death inducing signalling complex (DISC) or apoptosome (Karki, Dahal & Park,

2007). Activation of a second class of caspases known as the effector caspases is

limited to the proteolysis of the linker region which is carried out by the initiator

caspases. Little is known about the activation of the inflammatory caspases 1, 4 and

5. With their long pro-domain they resemble initiator caspases; however, caspase-4

is activated by dimerization and inter-linker domain cleavage (Karki, Dahal & Park,

2007). Thus, inflammatory caspases show a novel mechanism of activation including

both dimerization and proteolytic cleavage which differs from the well-established

mechanism of activation for initiator and effector caspases.

Page 52: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

52

ii. Caspase-4: Homologs and orthologs

The murine ortholog to human caspase-4 is disputed among the scientific

community. Mice have only 3 inflammatory caspases including caspase-1, -11 (also

known as -4) and -12 (Sollberger et al., 2012). Murine caspase-11 and -12 share

60% and 49% homology with human caspase-4 respectively and both caspase-11

and -12 have been suggested as the functional ortholog to human caspase-4 due to

their role in inflammation and ER-stress induced apoptosis, respectively (Sollberger

et al., 2012; Martinon & Tschopp, 2007). It is proposed that human caspase-4 and

p20 p10 p22 Procaspase-4

Inter-linker domain

Caspase-4 p30 p20 p10

p20

p20

p10

p10 Active caspase-4

Figure 1 6 Structural activation of caspase-4. The zymogen procaspase- 4undergoes proteolytic cleavage upon activation. Cleavage of the p22 CARD domain results in the release of a p30 active domain which undergoes further cleavage and oligomerisation to form the active heterotetramer, consisting of the p20 and p10 subunits

Page 53: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

53

caspase-5 may in fact be a gene duplication of a caspase-11-like ancestral gene,

however this is not unanimously agreed (Martinon et al, 2004; Scott et al, 2007).

Emerging evidence suggests caspase-11 as the functional ortholog to human

caspase-4 due to their functional similarities and ability to detect intracellular LPS

and activate the non-canonical inflammasome (Shi et al., 2014; Sollberger et al.,

2012; Kayagaki et al., 2011).

iii. Caspase-4/-5/-11 and the non-canonical inflammasome

The model of the non-canonical inflammasome is based on the caspase-11

dependent inflammasome response in iMMs. Caspase-11 was first identified to be

essential for caspase-1 activation in response to LPS treatment in the endotoxin

shock model. Deletion of Casp11 completely rescues mortality due to LPS treatment

and abrogates secretion of both IL-1α and IL-1β (Wang et al., 1998). It was not until

15 years later that the term non-canonical inflammasome was to be coined.

Kayagaki and colleagues demonstrated that during infection of iMMs with Gram

negative pathogens, including C. rodentium and EPEC E69, that caspase-11 is

required upstream of activation of caspase-1 and essential for IL-1β/IL-18 and IL-1α

secretion (Fig. 1.7; Kayagaki et al., 2011). Importantly, while Asc and NLRP3 are

essential for IL-18 and IL-1β secretion in response to C. rodentium, suggesting that

caspase-11 acts via the NLRP3 inflammasome (Liu et al., 2012), deletion of NLRP3

or Asc has no effect on IL-1α or pyroptosis (Kayagaki et al., 2011). Non-canonical

inflammasome activation requires two signals. Signal I is a priming signal that

transcriptionally up regulates the components of the inflammasome including

NLRP3, caspase-11 and pro-IL-1β (Fig. 1.7). This is dependent on the Tir domain-

containing adaptor molecule 1 (TRIF) of the TLR-4 pathway, to activate NF-κB and a

type I IFN response in iMM (Fig. 1.7; Gurung et al., 2012; Rathinam et al., 2012).

Page 54: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

54

Alternatively type II IFN has also been shown to be essential for caspase-11

synthesis in vivo in IECs (Oficjalska et al., 2015). Once primed a secondary signal,

signal II, is required to activate caspase-11 and non-canonical inflammasome

activation.

Figure 1 7 The non-canonical inflammasome model. Activation of the non-canonical inflammasome requires two signals. Signal I is a priming signal that through detection of LPS drives the TLR-4/TRIF dependent activation of NF-κB and subsequent Type I Interferon and NF-κB dependent expression of NLRP3, pro-caspase-1, IL-1β and pro-caspase-4. Signal II is the activator, which in this model is LPS. Upon detection of intracellular LPS pro-caspase-4/-11 undergoes self-proteolytic cleavage and oligomerisation. This leads to formation of the inflammasome through recruitment of NLRP3, Asc and pro-caspase-1 in to a macromolecular complex. The exact trigger for this is unknown but requires caspase-4/-11 activation. Pro-caspase-1 undergoes self proteolytic cleavage and oligomerisation and processes pro-IL-1β/IL-18 triggering their secretion.

Page 55: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

55

Although the PAMP/DAMP has not been verified during bacterial infection, the

activator of the non-canonical inflammasome is hypothesised to be intracellular LPS

and more specifically the Lipid A motif (Shi et al., 2014; Kayagaki et al., 2011). The

lipid A motif of LPS directly interacts with a conserved lysine residue at position 19

present in the CARD domain of both caspase-4 and -11, resulting in self-proteolytic

cleavage and oligomerisation in vitro (Fig. 1.7; Shi et al., 2014). In iMM activation of

caspase-11 via cytosolic LPS results in non-canonical inflammasome activation and

the downstream activation of caspase-1 and secretion of IL-1β, IL-18 and IL-1α and

initiation of pyroptosis (Fig. 1.7; Kayagaki et al., 2011; Shi et al., 2014). Little work

has been carried out in human cell lines due to the availability of knock-out mice and

an in vivo model of infection; however, to date, both caspase-4 and caspase-5 have

been implicated in inflammasome activation during enteric pathogen infection and

LPS transfection.

Sollberger and colleagues observed that caspase-4 is essential for caspase-1

activation and IL-1β/IL-18 secretion in the macrophage-like human acute monocytic

leukemia cell line (THP-1) and keratinocytes. This however was in response to

canonical stimuli, acting as a general upstream activator of both the canonical

NLRP3 and AIM2 inflammasome (Sollberger et al., 2012). On the other hand

caspase-4 and caspase-5 can functionally complement Casp11-/- iMM to induce

pyroptosis in response to cytosolic LPS (Shi et al., 2014). Furthermore caspase-4 is

essential for pyroptosis in response to cytosolic LPS in the human cell line THP-1

(Shi et al., 2014). The ability of caspase-4/-5 to activate IL-1β/IL-18 secretion in

human myeloid cell lines, in response to intracellular LPS has not been studied;

therefore, the role of either in activation of the non-canonical inflammasome remains

unclear. Further work is required to understand their function and their role in

Page 56: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

56

inflammasome activation. The fact that humans encode two inflammatory caspases

that can detect LPS may mean that the “non-canonical inflammasome” or the

inflammasome that forms in response to LPS may differ from the murine non-

canonical inflammasome response. In fact a recent discovery of a caspase-4

dependent/ caspase-1 independent inflammasome in human IECs questions the

current hypothesis and non-canonical inflammasome model (Knodler et al., 2014).

iv. The unconventional non-canonical inflammasome in epithelial cells

iMMs have been the primary cell line in which the inflammasome response to enteric

pathogens has been studied in vitro. EPEC E69 and C. rodentium are extracellular

bacterial pathogens that attach to IECs to colonize the host. Therefore IECs may

represent a more physiologically relevant cell line to study the host response and

inflammasome activation during EPEC E69 and C. rodentium infection. Caspase-4

has been implicated to regulate IL-18 secretion in response to Salmonella and EPEC

E69 infection, as well as LPS transfection of polarised IECs (Knodler et al., 2014).

Surprisingly, the secretion of IL-18 and activation of pyroptosis in response to

Salmonella infection of IECs is uniquely caspase-1 independent. Similarly EPEC E69

induced secretion and cleavage of IL-18 is also caspase-1 independent. A caspase-

1 independent inflammasome has not been described previously and whether the

IEC caspase-4 dependent inflammasome requires an NLR or Asc is unknown; but

this pathway significantly differs to the characterised mouse macrophage non-

canonical inflammasome response (Knodler et al., 2014). In support of this the C.

rodentium mouse model of infection has identified that both IECs and myeloid cells

contribute to inflammasome dependent cytokine secretion in vivo (Song-Zhao et al.,

2014; Gurung et al., 2012; Liu et al., 2012; Kayagaki et al., 2011).

Page 57: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

57

1.6.4 The role of the inflammasome in vivo

Many studies have been carried out to investigate the role of the inflammasome

response during C. rodentium infection of C57BL/6 mice. Clearance of C. rodentium

is significantly abrogated day 14 post-infection of Il-18-/-, Il-1β-/-, Nlrp3-/-, Nlrc4-/- and

Casp11/1-/- C57BL/6 mice (Liu et al., 2012) and day 7 post infection in Casp11-/-

C57BL/6 mice (Gurung et al., 2012). The increase in bacterial burden is

accompanied by an increase in crypt hyperplasia, immune cell infiltration and

gastrointestinal inflammation (Liu et al., 2012). This concludes that late in infection

caspase-11, caspase-1, NLRP3, NLRC4 as well as IL-1β/IL-18 secretion, all play a

crucial role in the immune response to C. rodentium infection in vivo. Secretion of IL-

18 at day 8 during C. rodentium infection of bone marrow irradiated Nlrp3-/- and

Nlrc4-/ - C57BL/6 mice is unaffected; while Asc KO abolishes IL-18 secretion

completely (Nordlander, Pott & Maloy, 2014; Song-Zhao et al., 2014). This suggests

that IL-18 maturation and secretion and therefore inflammasome activation, early

during C. rodentium infection, is NLRP3 and NLRC4 independent and occurs

exclusively in the non-hematopoietic cells of the gastrointestinal tract (Song-Zhao et

al., 2014); in an Asc dependent manner. Whether another NLR is required for IEC

dependent IL-18 secretion in vivo or whether caspase-1 is dispensable in vivo

remains unknown. Importantly, the research to date suggests that IL-18 maturation

and secretion in response to infection with enteric pathogens is not exclusively via

the in vitro characterised “conventional” non-canonical inflammasome pathway

described previously. Furthermore these studies conclude that both IL-1β and IL-18

play an essential role in the regulation of the immune response and clearance of C.

rodentium in vivo.

Page 58: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

58

1.6.5 The IL-1 cytokine family

The IL-1 family of cytokines consists of the pro-inflammatory effectors IL-1α and β,

IL-18, IL-33 and IL-36α, β, γ as well as the anti-inflammatory effector IL-37

(Garlanda, Dinarello & Mantovani, 2013). IL-1 was one of the first cytokines to be

discovered to have anti-microbial activity (van der Meer, 1988) and has since been

discovered to be the causative agent of many auto-immune inflammatory diseases

(Garlanda, Dinarello & Mantovani, 2013) including: arthritis and psoriasis.

i. IL-1α and IL-1β share biological properties

IL-1α and IL-1β share the same receptor as agonists for the IL-1 receptor (IL-1R),

which is present on the surface of almost all cell types (Garlanda, Dinarello &

Mantovani, 2013). Whilst this means they are almost functionally redundant the

major difference between IL-1α and IL-1β is that the latter requires processing by

cysteine proteases from its 30 kDa form to a 17 kDa cleavage product to be

biologically active. This is canonically via caspase-1 and inflammasome activation;

however, IL-1β can also be activated extracellularly by neutrophil proteinase-3 or

elastase (Garlanda, Dinarello & Mantovani, 2013). The activity of IL-1α and IL-1β is

further separated by the fact that IL-1β expression is limited to a subset of cell types

and is not expressed in human intestinal epithelial cells (Daig et al., 2000).

Stimulation of IL-1R drives a similar TRAF6/TAK1 dependent NF-κB and MAPK p38

dependent transcriptional activation as seen via the TLR-4 pathway, to prime and

activate cells of the innate and adaptive immune system (Dunne & O'Neill, 2003).

ii. The functional role of IL-1β in vivo

IL-1β plays a significant role in inflammatory signaling in vivo. As mentioned many

cell types express the IL-1R and therefore IL-1β has a broad spectrum of activity and

priming cells including natural killer (NK) cells, monocytes and innate lymphoid cells

Page 59: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

59

(ILC). Through stimulation of NK cells it induces secretion of IFNγ; while activation of

ILC3 drives an IL-17 and IL-22 response (Garlanda, Dinarello & Mantovani, 2013).

While classically behaving as a cytokine and signaling via IL-1R it is also an

essential chemokine in the chemotaxis and recruitment of macrophages (Rider et al.,

2011). IL-1β also has an essential role in activation of the adaptive immune

response. Through signal transduction in endothelial cells and transcriptional

activation of cell surface adhesions and chemokines IL-1β acts to recruit leukocytes

to the site of infection. Stimulation of non-conventional Th1 cells with IL-1β triggers

IFNγ release and subsequent macrophage activation (Garlanda, Dinarello &

Mantovani, 2013). Further to this IL-1β activates an IL-17 response from Th17 and

Tγδ17 T cells resulting in release of IL-8 from endothelial and epithelial cells

(Garlanda, Dinarello & Mantovani, 2013). The response to IL-18 secretion

significantly differs to that of IL-1β; most likely due to the difference in the subset of

cell types that express IL-18R.

iii. IL-18 and IFNγ inducing factor

Regulation of IL-18 differs from that of the other family members due to the fact that

it is constitutively expressed (Puren, Fantuzzi & Dinarello, 1999). How the

expression of IL-18 is manipulated or further regulated is poorly characterized.

Similar to IL-1β, activity of IL-18 is restricted and requires processing of the ~24 kDa

pro-form to the biologically active 18 kDa form to be secreted. IL-18 is not an IL-1R

family receptor agonist and instead binds a distinct receptor aptly named IL-18

receptor (IL-18R). Stimulation of IL-18R activates MAPK p38 and the transcriptional

activators AP-1 and NF-κB in a TRAF6 dependent manner (Dinarello & Fantuzzi,

2003), although not in all cell types (Lee et al., 2004). IL-18 was initially annotated as

IFNγ-inducing factor. Along with IL-12, IL-18 is essential for the expression of IFNγ.

Page 60: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

60

Unlike other pro-inflammatory cytokines IFNγ can be administered as a therapeutic,

without adverse inflammatory side effects; having an essential role in clearance of

bacterial infection and sepsis (Dinarello & Fantuzzi, 2003). The ability of IL-18 to

induce IFNγ gives it a unique role in vivo and function among the IL-1 superfamily.

iv. The functional role of IL-18 in vivo

IL-18 induces the production of IFNγ through NK cells/Th1 cells and ILC1 through

co-stimulation with IL-12 and IL-25, respectively (Garlanda, Dinarello & Mantovani,

2013). As an IFNγ inducer IL-18 stimulates the production of Nitric oxide (NO) to kill

intracellular pathogens, including Salmonella. Furthermore IL-18 has been shown to

facilitate the emigration of neutrophils and leukocytes to the site of infection. This is

by two mechanisms: firstly IL-18 can increase the expression of adhesion molecules

ICAM-1 and VCAM-1 (Dinarello & Fantuzzi, 2003) or alternatively, in an IFNγ

dependent manner, IL-18 triggers a cytokine/chemokine signaling cascade via MIP-

2, MIP-1α, TNFα and finally Leukotriene B4 (LTB4; Verri et al., 2007; Canetti et al.,

2003). LTB4 is a potent neutrophil chemokine. IL-18 dependent recruitment of

neutrophils was studied using intra-peritoneal (IP) injection of mice with rIL-18 or rIL-

15 (Verri et al., 2007; Canetti et al., 2003); therefore, it is not known if IL-18 or the

inflammasome plays an essential role in neutrophil recruitment in the intestinal

mucosa. On the other hand it is established that IL-18 plays a crucial role in the

mouse model of Crohn’s disease and inflammatory bowel disease (IBS; Dinarello &

Fantuzzi, 2003). This has led many studies to target IL-18 as a therapeutic for IBS.

Whether or not IL-18 signaling is targeted by pathogenic E. coli or C. rodentium

during infection has not been studied; although, manipulation of inflammatory

signaling is a key aspect of the A/E pathogen infection strategy.

Page 61: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

61

1.7 Manipulation of the inflammatory response by enteric pathogens

Multiple effectors have been identified to be recruited by EPEC E69 to inhibit pro-

inflammatory signalling via the JNK and NF-κB dependent signalling cascades

(Raymond et al., 2013). The majority of the characterised T3SS anti-inflammatory

effectors are encoded on the PAIs PP4 and IE6 and include NleB1, NleE1, NleC and

NleD.

1.7.1 The EPEC anti-inflammatory effectors NleB1, NleC, NleD and NleE1

inhibit NF-κB and JNK signalling

NleB1, NleE1, NleC and NleD, when ectopically expressed, can function to inhibit

inflammatory signalling via exogenous simulation. Both NleD and NleC are zinc-

dependent metalloendopeptidases which co-operate to inhibit inflammatory

signalling through the direct cleavage of JNK/p38 and p65, respectively (Fig. 1.8;

Baruch et al., 2011; Muhlen, Ruchaud-Sparagano & Kenny, 2011; Pearson et al.,

2011; Sham et al., 2011; Yen et al., 2010). NleB1, but not NleB2, of EPEC functions

as an N-acetyl-D-glucosamine (O-GlcNAc) transferase to disrupt TNFR induced

TRAF-2 dependent NF-κB activation in response to stimulation with TNFα (Fig. 1.8;

Gao et al., 2013; Newton et al., 2010). NleE1 is employed by EPEC to inhibit NF-κB

activation and IL-8 secretion in response to both IL-1β and TNFα stimulation

(Newton et al., 2010). Through a unique S-adenosyl-l-methionine-dependent

methyltransferase activity, NleE1 targets the host ubiquitin sensory complex proteins

TAB2/3 to inhibit IκBα degradation and NF-κB activation (Fig. 1.8; Zhang et al.,

2011). While many of the above mentioned effectors have been characterised to

inhibit inflammatory signalling when ectopically expressed their role in vivo, due to

the complexity of signalling, is less clear.

Page 62: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

62

Figure 1 8 EPEC E69 recruits a plethora of effectors to inhibit inflammatory signalling during infection. EPEC E69 delivers multiple effectors to subvert inflammatory signalling. This includes the metalloendopeptidases NleD and NleC that target and cleave JNK/p38 and p65, respectively. NleE, a methyltransferase targets TAB2/3 to prevent IκBα degradation while NleB is N-acetyl-D-glucosaminetransferase that targets GAPDH to disrupt TRAF2 dependent TNFR signalling.

Page 63: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

63

1.7.2 Inflammatory signalling during EPEC E69 infection

While EPEC E69 secretes a plethora of anti-inflammatory effectors it initially

activates and later inhibits inflammatory signalling and the release of immune

mediators during infection, in a T3SS-dependent manner. Initially, EPEC E69

induces a pro-inflammatory response during in vitro infection of epithelial cells

through activation of MAPK extracellular regulated kinase 1/2 (ERK1/2; Czerucka et

al., 2001; de Grado et al., 2001) and NF-κB (Newton et al., 2010; Savkovic,

Koutsouris & Hecht, 1997). Interestingly, activation is T3SS-dependent and no NF-

κB or ERK1/2 activation is observed during infection with the T3SS mutant’s ΔescN

(Pearson et al., 2011; Newton et al., 2010) or ΔespB (de Grado et al., 2001;

Savkovic, Koutsouris & Hecht, 1997). Furthermore, NF-κB activation is ERK1/2

dependent and the host inflammatory response during infection is TNFR/ IL-1R

independent (Savkovic et al., 2001); suggesting EPEC E69 delivers an unknown

effector to activate pro-inflammatory signalling. Phosphorylation of ERK1/2 signalling

is no longer detectable 1 h post infection (Savkovic et al., 2001) and at later time

points, in a T3SS-dependent manner, EPEC E69 inhibits NF-κB pro-inflammatory

signalling and IL-8 secretion (Pearson et al., 2011; Newton et al., 2010). Furthermore

NF-κB activation and IL-8 secretion, induced by TNFα or IL-1β exogenous

stimulation, is inhibited 4 h post infection in T3SS dependent manner (Pearson et al.,

2011; Newton et al., 2010). Deletion of NleE1 alone significantly increases IL-8

secretion during infection, independent of exogenous stimulation (Pearson et al.,

2011). While deletion of NleC alone does not affect IL-8 secretion; deletion of NleC

in the EPEC ΔnleE1 strain background promotes further IL-8 secretion during

infection (Pearson et al., 2011). Importantly infection with an EPEC E69 strain

harbouring a dual deletion in the PAIs PP4 and IE6 is no longer able to inhibit TNFα

Page 64: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

64

induced NF-κB activation at 4 h post infection and in fact infection with EPEC

ΔPP4/IE6 constitutively activates NF-κB activation and IL-8 secretion (Pearson et al.,

2011; Newton et al., 2010). This is most likely due to an uncharacterised pro-

inflammatory effector and the early ERK1/2 and NF-κB activation; which is no longer

inhibited by the translocation of the anti-inflammatory effectors on islands PP4 and

IE6. Furthermore, complementation of EPEC ΔPP4/IE6 with NleE1 alone is able to

significantly inhibit NF-κB activation and IL-8 secretion during infection to levels of

that seen for the WT (Pearson et al., 2011; Newton et al., 2010). This suggests that

NleE1 is the main inhibitor of NF-κB activation during in vitro infection. Recently,

NleC was identified to inhibit KC, IL-22, MIP-2α and IL-1β expression during in vivo

C. rodentium infection resulting in inhibition of the recruitment of CD11b positive

myeloid cells and a reduction in pathology; confirming an anti-inflammatory role for

effectors in vivo (Hodgson et al., 2015). Therefore the manipulation of the

inflammatory response during EPEC E69 and C. rodentium infection is a complex

network of pro-inflammatory and multiple anti-inflammatory effectors working in

conjunction. While no pro-inflammatory effector(s) has been reported for EPEC E69

the atypical EPEC strain E110019 and C. rodentium encode the characterised pro-

inflammatory effector EspT (Raymond et al., 2011).

1.7.3 EspT is a pro-inflammatory effector of atypical EPEC

EspT of atypical EPEC induces activation of NF-κB and ERK1/2 signalling during

infection, resulting in up-regulation of expression and secretion of the pro-

inflammatory cytokines and chemokines IL-8 and IL-1β (Raymond et al., 2011). EspT

belongs to a family of WxxxE motif proteins with guanine nucleotide exchange factor

(GEF) mimicry and targets Rac1 to activate inflammatory signalling (Raymond et al.,

2011). This activity of EspT is restricted to infection of immune cells and has no pro-

Page 65: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

65

inflammatory effect on epithelial cells. Moreover, EspT is confirmed to contribute to

the expression of the immune mediators TNFα and KC in vivo during C. rodentium

infection (Raymond et al., 2011). While pathogenic E. coli deliver pro-inflammatory

effectors as part of their infection strategy; to date, the pro-inflammatory effector(s) of

EPEC E69 remains unknown.

1.8 NleF of attaching and effacing pathogens

The newly characterised non-LEE encoded effector F (NleF; Echtenkamp et al.,

2008; Tobe et al., 2006) is a core effector which is highly conserved among EHEC

strains and shares 100% identity with homologs of the EPEC strains O127:H6

E2348/69, O26:H11, O55::H7, and O111:H-, as well as sharing 84% amino acid

sequence identity with C. rodentium (Iguchi et al., 2009; Fig. 1.9A). The PAI, OI-71

(EHEC) or PP6 (EPEC), encodes NleA/EspI and also NleF (z6020) in an operon with

NleH2 (Fig. 1.9B). Expression is regulated by the presence of an nle regulated

inverted repeated (NRIR) domain (Garcia-Angulo et al., 2012), which was recently

shown to be a unique promoter for the non-LEE encoded effectors of pathogenic E.

coli (Garcia-Angulo et al., 2012). NleF is thought to have been acquired by horizontal

gene transfer along with NleH1-2 and NleA/EspI. High through-put virulence gene

profiling of typical and atypical EPEC clinical isolates identified the presence of OI-71

(PP6) and nleF associates with serotypes linked to severe outbreaks and diarrhoea

in children (Bugarel et al., 2011). Furthermore Zhang and colleagues identified that

EHEC O157:H7 separated into two dominant lineages, with lineage I strains linked to

disease in humans (Zhang et al., 2007). Interestingly genomic analysis of strains

from lineage I and II showed that both nleF and nleH1-2 are absent from lineage II

strains and therefore may be important for colonisation and disease in humans

(Zhang et al., 2007).

Page 66: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

66

Characterisation of NleFE69 in silico shows no homology to known Homo sapien

proteins or to other characterised/ uncharacterised T3SS effectors of enteric

pathogens or homology with previously described functional domains (Echtenkamp

et al., 2008). Initial functional characterisation of NleF from EHEC O157:H7

(NleFEHEC) identified that when ectopically expressed NleFEHEC localises diffusely

throughout the cell and furthermore, unlike NleA/EspI, it does not manipulate

Figure 1 9 Alignment of NleF of pathogenic E. coli and distribution of PAI Prophage 6 (PP6). (A) ClustalW2 multiple sequence alignment for NleF of following strains: O127:H6 str. E2348/69, O157:H7 str. EDL933/ Sakai/ 85-170, O55:H7 str.CB9615, O111:H- str. B171-1, O26:H11 str. 11368, O103:H2 str. 12009 andCitrobacter rodentium str. ICC169 (B) Schematic of PAI PP6 of EPEC O127:H6 E2348/69. UN, uncharacterised/ hypothetical protein.

Page 67: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

67

intracellular trafficking and/ or secretion (Echtenkamp et al., 2008).Therefore the

functional role of NleF and the host target binding partners remain unknown.

1.9 Aims and outline

The aim of this study was characterise the functional role of NleF in infection using in

vitro and in vivo models.

Page 68: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

68

Chapter 2: Materials and Methods

Page 69: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

69

2.1 Bacterial strains, reagents, growth conditions and plasmids

Strains used in this study are listed in Table 2.1. E. coli and C. rodentium were

cultured in Miller Luria Broth (LB) at 37 C, at 200 rpm with the appropriate

antibiotics: 50 µg/ml kanamycin, 100 µg/ml ampicillin and 30 μg/ml chloramphenicol.

The plasmids constructed and primers used for cloning in this study can be found in

Table 2.2 and cDNA templates in Table 2.3. nleFE69 was amplified from E. coli

O127:H6 E2348/69 and nleFCR was amplified from C. rodentium ICC169 genomic

DNA by PCR using KOD Hot Start Polymerase (Novagen). Human CASP4 and

CASP5 were amplified from pICC1651 and pICC1653, respectively. Mouse Casp11

was amplified from pICC1650. GGCT, HBXIP, DHFR and DHRL1 were amplified

from plasmids outlined in Table 2.3. All cloning was carried using restriction site

digestion (NEB) and T4 DNA Ligase (NEB) unless specified otherwise. Site-directed

mutagenesis was carried out by inverse PCR using KOD Hot Start polymerase and

mismatch primers (See Table 2.4). All constructs were confirmed by sequencing

(GATC). All reagents used in this study were purchased from Sigma-Aldrich, unless

stated otherwise.

2.2 Y2H screening and YDH hybrids

A Yeast-2-Hybrid screen was conducted using pGKBT7-nleFE69 using the HeLa cell

cDNA Library following the manufacturer’s instructions (Clontech). The inserts of

positive clones were amplified using the primer pair AD-LDF/R using Green Taq 2x

Master mix (NEB) and sent for sequencing. A Yeast Direct Hybrid was carried out

following the manufacturer’s instructions (Clontech). AH109 were co-transformed

with pGBT9-bait and pGADT7-prey (See Table 2.2) and plated onto Difco Yeast

Nitrogen Base without amino acids (SD) agar supplemented with 2% glucose, 20

mg/L adenine hemisulfate (Ade), 20 mg/L arginine HCl, 20 mg/L histidine HCl

Page 70: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

70

monohydrate (His), 30 mg/L isoleucine, 30 mg/L lysine HCl, 20 mg/L methionine, 50

mg/L phenylalanine, 200 mg/L threonine, 30 mg/L tyrosine, 20/L mg uracil, 150

mg/ml valine and lacking tryptophan (Trp) and Leucine (Leu) (Double Drop-out;

DDO) for selection of transformed clones. Clones positive for both plasmids were re-

streaked on to SD DDO and SD QDO /-His/-Ade supplemented with 40 mg/L x-α-gal (SD QDO) for selection of positive interactions. Plates were incubated at 30 ºC for 3-

10 days.

2.3 Tissue culture

The human epithelial colorectal adenocarcinoma cells (Caco-2) clones TC7

(Chantret et al., 1994)) as well as the human embryonic kidney (HEK) 293E cells

(ATCC) were grown in Dulbecco’s Modified Eagle's Medium (DMEM) – high glucose

(4500 mg/L) supplemented with 15 % (v/v) heat inactivated Fetal calf serum (FCS;

Gibco), 2 mM Glutamine (Invitrogen), 0.1 mM Non-essential amino acids (NEAA)

and the antibiotics Penicillin and Streptomycin (P/S) at 100 U/ml and 100 μg/ml,

respectively. Cells were kept at 37 0C in a 5 % CO2 atmosphere and routinely

screened for Mycoplasma. C57BL/6 immortalised mouse macrophages (iMM) were

grown in DMEM – high glucose (4500 mg/L) supplemented with 10 % (v/v) heat

inactivated FCS, 2 mM Glutamine and 0.1 mM NEAA at 37 0C in a 5 % CO2

atmosphere. The human epithelial cervical adenocarcinoma cells (HeLa) were grown

in DMEM – low glucose (1000 mg/L) supplemented with 10 % (v/v) heat inactivated

FCS, 2 mM Glutamine and 0.1 mM NEAA at 37 0C in a 5 % CO2 atmosphere. All cell

lines were passaged routinely by treatment in 0.02% EDTA solution supplemented

0.1% trypsin (Gibco) for 5 min (30 min for Caco-2). The Human acute monocytic

leukemia cell line (THP-1; ATCC) were grown in suspension in Roswell Park

Memorial Institute (RMPI) medium supplemented with 10% FCS at 37°C in a 5%

Page 71: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

71

CO2 atmosphere. THP-1s were were seeded in 24-well plates at 7.5x105 and

differentiated in 75 nM phorbol myristate acetate (PMA) for 4 days and starved in

serum free RPMI with or without 200 ng/ml LPS before extraction for Western

blotting as described below.

2.4 Transfection assays

2.4.1 Plasmid transfection of HeLa cells

HeLa cells were seeded and grown to a confluence of ~40% in either 24 or 6 well

plates (BD Falcon). The media was replaced with serum free DMEM and transfected

with the eukaryotic expression vectors described in Table 2.2, using the transfection

reagent GeneJuice (Novagen), according to the manufacturers specifications. As a

control cells were left unstimulated or stimulated with 20 ng/ml TNFα for 0.5 h or 16 h

where specified. For immunofluorescence staining transfected cells were washed 3

times in warm phosphate buffered saline (PBS) at indicated time points and fixed for

20 min in 3.4% paraformaldehyde (PFA; Agar Scientific). Fixed cells were washed 3

times in warm PBS and stored at 4ºC for further analysis. Alternatively, for RNA

extraction or Western blotting, transfected cells were washed 3 times in warm PBS

and stored at -80ºC for further analysis.

2.4.2 Transfection of short inhibitory RNA

HeLa cells were seeded to a confluence of ~40% in a T25 flask (BD Falcon) and

transfected with 20 nM ON-Targetplus SMARTpool (Dharmacon) small interfering

RNA (siRNA) for CASP4 (L-004404-00-0005), CASP8 (L-003466-00-0005) or

CASP9 (L-003309-00-0005) or with non-targeting scrambled siRNA (D-001810-10-

05; Dharmcon) as a control, using the siRNA transfection reagent Hiperfect

(Qiagen), according to the manufacturer’s specifications. For dual knockdown, HeLa

Page 72: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

72

cells were transfected with 20 nM of CASP4 and CASP8 smartpool siRNA or 40 nM

of the non-targeting control siRNA. Thirty six hours post-transfection cells were

washed and frozen for analysis by RT-qPCR or Western blotting as described below.

Alternatively 24 h post-transfection cells were treated in 0.02% EDTA/ 0.1% trypsin

for 5 min and seeded into 24 well (BD Falcon) plates. Twelve hours later siRNA-

treated cells at a confluence of ~40% were transfected with the mammalian

expression vectors (see Table 2.2) using GeneJuice, as described above. Twenty

four hours post-transfection cells were washed 3 times in warm PBS and stored at -

80ºC for further analysis.

2.5 Isolation of RNA and RT-qPCR

RNA was extracted and isolated following the manufacturer’s instructions (Qiagen;

RNeasy mini kit). Samples were pre-treated with RQ1 DNase (Promega) at 37 C for

10 min followed by DNase denaturation at 72ºC for 15 min. Reverse transcription

(RT) was carried following the manufacturer’s instructions (Promega) with the

following adaptations: RNasin (Promega) was added to the reaction buffer at 2-4

U/µl. RT Quantitative-PCR (RT-qPCR) was carried out using the 7300 Real Time

PCR System (Applied Biosystems) using the standard cycle conditions and primer

pairs IL-8F/R, CASP4F/R, CASP8F/R, CASP9F/R and GAPDHF/R (see Table 2.4).

Changes in gene expression levels were analysed relative to GFP (where stated),

using GADPH as a standard and using the ΔΔCT method.

2.6 Immunofluorescence staining and antibodies

Fixed cells were initially quenched in 50 mM NH4Cl followed by permeabilisation in

0.2% Triton X-100. Fixed cells were then washed 3 times in PBS blocked in 1%

Bovine serum albumin (BSA) diluted in PBS (BSA-PBS). All antibodies, unless

stated otherwise, were diluted directly in 1% BSA-PBS. Primary antibodies were

Page 73: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

73

incubated for 45 min followed by 3 washes in PBS and second round of blocking for

15 min. Secondary antibodies and/or reagents were incubated for a further 30 min.

Cover-slips were then washed 3 times in PBS and mounted onto microslides using

Prolong Gold anti-fade reagent (Invitrogen) before visualising on the Zeiss

Axioimager immunofluorescence microscope using the Zeiss Axiovision Rel 4.5

software. Primary antibodies: the mouse monoclonal antibodies anti-Myc clone 4A6

(05-724; Millipore), anti-TOMM22 (ab57523; Abcam), the rabbit polyclonal antibody

anti-N-terminal NF-κB p65 Clone A (sc-109; Santa Cruz) and the chicken polyclonal

antibody anti-EaeM β (full-length intimin β of Porcine EPEC) (Girard et al., 2005).

Secondary antibodies: DyLight488-conjugated donkey anti-mouse IgG (H+L)

(catalog no. 715-485-150; Jackson immunoresearch), Donkey anti-Rabbit IgG (H+L)

(catalog no. 711-295-152; Jackson immunoresearch) and goat anti-chicken IgY

(H+L) (catalog no. 103-605-155; Jackson immunoresearch). Stains: Phalloidin-

Alexa-647 (A-22287; Invitrogen), 4,6-Diamidino-2-phenylindole (DAPI) (D3571

;Invitrogen) and Phalloidin-Alexa-555 (P1951).

2.7 Bacterial priming, cell growth analysis and attachment assays

EPEC E69 strains were grown for 16 h ON as described above and primed by sub-

culturing 1:100 in DMEM low glucose with no additives supplemented with

appropriate antibiotics for 3 h, with no agitation in an atmosphere of 5% CO2. To

analyse cell growth an aliquot of each bacterial culture was extracted at 45 min

intervals and diluted by serial dilution in sterile warm PBS. Diluted samples were

plated at a range of 1 x103-1 x105/10 µl on LB-agar. Plates were stored at 37ºC for

16 h and colony forming units (CFU) were counted the following day.

Page 74: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

74

2.8 Generation of an EPEC E69 nleF deletion strain

An EPEC E2348/69 strain harboring a deletion in the gene nleF was generated using

the Lambda Red system (Datsenko & Wanner, 2000). The nleF gene along with 500

bp 5’ and 400 bp 3’ were amplified from Escherichia coli O127:H6 E2348/69

genomic DNA and cloned into TOPO Blunt II (Invitrogen) using the primers specified

in Table 2.2 using blunt end ligation. The kanamycin cassette was amplified from

pKD4 using the primer pair specified in Table 2.2. Using inverse PCR nleF was

removed and replaced with the kanamycin cassette using blunt end ligation to

generate pICC1674. The linear product was amplified from pICC1674 using primer

pair 1F/R. Mutagenesis was confirmed by sequencing using the primer pair

2.9 Infection of assays

2.9.1 Infection of the HeLa cell line

HeLa cells were seeded 16 h prior to infection for a confluence of 40 %. HeLa cells

were washed 3 times in warm PBS prior to infection and the medium was replaced

with serum free DMEM low glucose. Bacterial strains were grown and primed as

described above and expression of NleF in the EPEC ΔnleF pnleFE69 and NleE1 in

the EPEC ΔPP4/IE6 pNleE1 complemented strains was induced with the addition of

1 mM Arabinose or 0.5 mM IPTG at 0.5 h before infection, respectively. At 3 h post

serum-starving the HeLa cell medium was removed and replaced with 0.5 ml of a 1:3

dilution of primed bacterial culture. At 1 h post infection HeLa cells were washed 3

times in warm PBS and incubated for a further 0.5 h or 3 h. For the 4 h time point

where indicated cells were stimulated with 20 ng/ml TNFα at 3.5 h post infection for

0.5 h. At 1.5 h and 4 h cells were washed 3 times in warm PBS and fixed in 3.4 %

PFA for immunofluorescence or stored at -20 ºC for analysis by Western blotting.

Page 75: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

75

2.9.2 Infection of iMM

iMM were seeded at 1.5 x106/ml and infected the next day. Three hours prior to

infection the medium was replaced with DMEM high glucose with FCS and

supplements with or without 200 ng/ml LPS (O111:B4) in Hanks balanced salt

solution (HBSS; Gibco). EPEC E69 bacterial cultures were primed as above and

primed or un-primed iMM were infected at an MOI of 25 for 1 h. Alternatively iMM

were treated with STBL2 (Clontech) E. coli grown over-night in LB at 37ºC at 200

rpm at an MOI of 25. At 1 h post-treatment cells were washed 5 times in warm PBS

and the medium was replaced with DMEM high glucose with FCS/ supplements and

further supplemented with 200 µg/ml gentamicin for 20 h. At 19 h post treatment

primed iMM were treated with 5 mM ATP for 1 h. Supernatants were taken at 21 h

post treatment/infection and centrifuged at 13000 rpm at 4 ºC for 10 min. Cleared

supernatants were analysed by ELISA or stored at -20ºC for further analysis.

Alternatively the supernatant was extracted for analysis of LDH release; while

monolayers were analysed for cell death by PI uptake as described below.

2.9.3 Infection of the Caco-2 cell line

Caco-2 TC7 were seeded at 3.5 x104 and upon reaching a confluent layer the media

was changed every day for 7 days. On day 6 post confluence antibiotics were

removed from the media. On day 7 post confluence cells were starved for 3 h in

DMEM high glucose with no FCS or antibiotics. Bacterial cultures were primed as

above and serum-starved Caco-2 TC7 were infected at an MOI of 10. After 3 h the

cell layer was washed twice in warm PBS and the medium was replaced with DMEM

high glucose no FCS supplemented with Penicillin and Streptomycin (Sigma Aldrich)

at 100 U/ml and 100 μg/ml, respectively. Alternatively cell layers were treated with

1% Triton X-100 in PBS for 10 min and plated for enumeration of attachment. After 1

Page 76: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

76

h the wash in PBS was repeated and the cells were left a further 16 h in DMEM high

glucose no FCS supplemented with 200 µg/ml gentamicin. Supernatants were

cleared by centrifugation at 13000 rpm at 4ºC for 10 min. Supernatants were

precipitated for Western blotting with the addition of 10 % (v/v) trichloroacetic acid at

4 ºC for 17 h. Precipitated samples were centrifuged at 13000 rpm for 45 min at 4 ºC.

Pellets were washed once in ice cold methanol and re-suspended in 2 x LDS. Cell

monolayers were stored at -20 ºC for RNA extraction or Western blotting.

Alternatively the supernatant was extracted for analysis of LDH release; while

monolayers were analysed for cell death by PI uptake as described below.

2.10 Western blotting and coomassie staining

At indicated time points cells were lysed in loading dye (LDS; 250 mM Tris pH 6.8,

40% glycerol, 6 % SDS, 1 % bromophenol blue and 0.8% β-mecaptoethanol) and

boiled for 10 min, separated by SDS-PAGE gel and then either transferred to a

PVDF membrane (GE Healthcare) or incubated for 10 min in Instant Blue

(Expedion). The PVDF membranes were blocked in Tris buffer saline pH 7.4 (10 mM

Tris, 150 mM NaCl)/0.1% Tween-20 (TBS-T) containing 5% semi-skimmed milk

(blocking buffer) for 45 min before incubation with primary antibody in blocking buffer

for 16 h at 4 C. Membranes were washed in TBS-T and the secondary antibody was

incubated for 2 h in blocking buffer. Western blots were visualized by the addition of

EZ-ECL solution (GE Healthcare) and developed using the LAS-3000 imager (Fuji).

Primary antibodies: mouse monoclonal antibodies anti-caspase-4 clone 4B9 (sc-

56056; Santa Cruz), anti-α-Tubulin clone DM1A (T6199), anti-NLRP3 clone Cryo-2

(AG-20B-0014-C100; Adipogen), the mouse polyclonal antibody anti-caspase-11

p20 clone A-2 (sc-374615; Santa cruz) and anti-pro-IL-18 (CPTC-IL18-1; DSHB), the

rabbit monoclonal anti-IL-18 (PM014; MBL), anti-caspase-5 (4429; Cell signalling)

Page 77: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

77

and the rabbit polyclonal antibodies anti-IκBα (29442; Cell Signaling), anti-GFP

(Ab290; Abcam) and anti-caspase-1 (2225; Cell signaling). Secondary antibodies:

Horse radish peroxidase (HRP)-conjugated goat anti-rabbit IgG (Fc fragement;

catalog no.111-035-008; Jackson immunoresearch) and HRP-conjugated goat anit-

mouse IgG (Fc fragement; catalog no, 115-035-008; Jackson immunoresearch).

2.11 Cytokine secretion analysis and cell death assays

Supernatants post-treatment or post-infection were removed and stored at -20ºC.

Supernatants were thawed and diluted in reagent buffer and enzyme-linked

immunosorbent assays (ELISAs) were carried out following the manufacturer’s

instructions. Mouse KC and IL-18 were purchased from eBioscience and mouse/

human IL-1β and mouse IL-1α were purchased from R&D systems. Supernatants

from infection were taken and analysed for Lactose dehydrogenase (LDH) release

using the Cytotox 96 non-radioactive cytotoxicity assay (Promega), following the

manufacturer’s specifications. As a control for total LDH, cell lysis buffer (1 % Triton-

X100/ PBS) was added for 30 min at 37ºC directly to the medium and cell layer.

Absorbance was measured at 490 nm using the FluoStar Omega plate reader and

results are displayed as percentage of total release. Alternatively the media was

removed and cell layers were incubated in 3.3 µg/ml propidium iodide (Invitrogen) in

warm PBS (PI/PBS) for 15 min and fluorescence was measured at an excitation of

510 nm and emission of 610 nm using the FluoStar Omega plate reader. As a

control PI/PBS alone was measured or cell lysis buffer (0.05 % Triton X-100/PBS)

supplemented with 3.3 µg/ml propidium iodide was added for 15 min at 37ºC.

Results are displayed as a percentage of total PI uptake.

Page 78: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

78

2.12 Affinity –tagged purification and size exclusion

BL21 Star were cultured for 16 h in LB at 37 C at 200 rpm. Bacteria were sub-

cultured at 1:100 into 1 L LB supplemented with 50 µg/ml kanamycin and incubated

at 37 C at 200 rpm until OD600 of 0.4-0.6. Cultures were then induced with 0.5 mM

IPTG for 18 h at 18 C. Cells were harvested by centrifugation at 10000 rpm for 20

min and re-suspended in 30 ml ice cold His-lysis buffer (20 mM Tris, 0.5 M NaCl,

and 5 mM Imidazole pH 7.9). The cells were lysed by Emulsiflex following the

manufacturer’s instructions (Emulsiflex-B15; Avestin) and centrifuged at 14000 rpm

for a further 30 min at 4 C. Supernatant was removed and applied to 5 ml His resin

(Novagen) pre-charged in 5 mM NiSO4 and pre-equilibrated in His-lysis buffer and

rocked at 4 C for 1.5 h. Samples were applied to a Poly-Prep Chromatography

column (Qiagen) and flow-through was collected. The column was washed twice with

20 ml His-lysis buffer and once in 20 ml wash buffer (0.5 M NaCl, 20 mM Tris pH 7.9

and 60 mM Imidazole). His-tagged fusion proteins were eluted with 10 x 1 ml elution

buffer (His-lysis buffer supplemented with 1 M Imidazole). Fractions containing His-

purified NleF were checked by SDS-PAGE gel electrophoresis and further purified by

size exclusion (Akta prime) with a Superdex75 column (GE Healthcare; 10/300GL).

2.13 Co-expression and purification of the caspase-4/NleFE69 complex

BL21 Star co-transformed with pACYC-DUET-1-CASP4 His-p20/p10 and pMal-c2x-

nleFE69 were cultured for 16 h in LB at 37 C at 200 rpm. Bacteria were sub-cultured

at 1:100 into 1 L LB supplemented with 50 µg/ml ampicillin and 15 µg/ml

chloramphenicol and incubated at 37 C at 200 rpm until OD600 of 0.4-0.6. Cultures

were then induced with 1 mM IPTG for 18 h at 20 C. Bacterial pellets were re-

suspended in 20mM Tris-HCl pH 7.4, 250mM NaCl and lysed by sonication and

purified by amylose and talon (Kotik-Kogan et al., 2008) affinity chromatography.

Page 79: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

79

Complex formation was analysed by size exclusion (Akta prime) with a Superdex200

column (GE Healthcare) using the Gel Filtration Markers Kit for Protein Molecular

Weights 12,000-200,000 Da to determine complex size. Size exclusion fractions

were verified by SDS PAGE gel. For the MBP pull-down assays cell lysates of BL21

co-transformed with pMAL-c2x/pACYC-DUET-1 His-p20 C258S/p10, pMAL-c2x-

nleFE69/ pACYC-DUET-1 His-p20 C258S/p10 and pMAL-c2x-nleFE69/ pACYC-DUET-

1 His-p20 C258S were incubated with amylose resin for 1.5 h at 4 �C and then

washed with 50 ml wash buffer (20mM Tris-HCl pH 8.0, 250mM NaCl). The proteins

bound to the resin were eluted with 10 mM maltose and analysed by SDS-PAGE.

2.14 Caspase activity assays

BL21 star transformed with pET28a-empty, pET28a-CASP4, pET28a-CASP4 C258S

or pET28a-Casp11 were grown, induced and lysed as described above. Soluble

lysates at 200 µg.ml were incubated with or without 50 µM Ac-LEVD-AFC in 20 mM

PIPES, 100 mM NaCl, 10 mM DTT, 1 mM EDTA, 0.1% CHAPS, 10% sucrose pH 7.2

or 20 mM Tris, 250 mM NaCl pH 7.4 for caspase-11 and caspase-4, respectively.

Purified recombinant His-NleF and homologs were added at varying concentrations

from 50 nM to 1 pM. Fluorescence was measured in 5 min intervals at 37 C using

an excitation of 410 nm and emission of 520 nm using FLUOstar Omega plate

reader (BMG Labtech). Results are displayed as percentage of activity (RFU/min)

relative to the positive control: caspase-4 lysate with the addition of Ac-LEVD-CHO

only.

2.15 shRNA construction and retroviral transductions

HEK293E cells (ATCC) were seeded in to 6 well plates (BD Falcon) and transfected

with the packaging plasmids pCMV-VSV-G, pCMV-MMLV-pack (Shenoy et al., 2012)

Page 80: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

80

and pMXs-IRES-puro (Table 2.2) containing the gene of interest to construct a

Vesicular Stomatis pseudo-typed retrovirus for transduction, following the

manufactures’ instructions (Clontech) and using Lipofectamine 2000 (Invitrogen).

After 48 h the supernatant was collected, filtered (0.45 µM) and added directly to the

medium of pre-seeded iMM. After 24-48 h transduced cells were selected by

puromycin (Gibco Invitrogen) at 5 µg/ml. Transduction was confirmed by

immunofluorescence and or Western blot, as described below. For stable

knockdown, short hairpin (sh)-RNA (Table 2.2) was constructed and cloned into

pMX-CMV-YFPc1 using one-step sequence and ligation independent cloning (SLIC)

(Jeong et al., 2012) following the miR30 based (Chang et al., 2013) and optimised

miR-E (Fellmann et al., 2013) shRNA vector design. Primer pairs were annealed,

filled in by PCR and amplified by shRNAF/R (Table 4). PCR amplicons were cloned

into pMX-CMV-YFPc1 using SLIC and XhoI digestion. Vectors were constructed to

target CASP4 (CGACTGTCCATGACAAGAT and lacZ (TRCN0000072226) as a

non-targeting shRNA control. Caco-2 TC7 were transduced as above and selected

for with 10 µg.ml puromycin. Transduction and knockdown were confirmed, as

described above.

2.16 Generation of C. rodentium nleF knockout mutant

C. rodentium strain ICC169 ΔnleF was generated using a modified version of the

lambda red-based mutagenesis system (Datsenko and Wanner, 2000). The nleF

gene and its flanking regions were PCR-amplified from WT C. rodentium ICC169

genomic DNA using the primer pair 4F/R and cloned into pGEMT vector (Promega).

The nleF gene was then excised using inverse-PCR (primer pair 5F/R) and the

resulting linear product was BamHI digested, allowing insertion of the non-polar

aphT (Galan, Ginocchio & Costeas, 1992) cassette. After verifying for correct

Page 81: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

81

orientation of the kanamycin cassette, the insert was PCR-amplified using primer

pair 4F/R. The presence of the mutation was confirmed by PCR and DNA

sequencing amongst the kanamycin resistant clones (primer pair 6F/R).

2.17 C. rodentium in vivo mouse model of infection

Pathogen-free female 18-20 grams C57BL/6 mice or male/female Casp1/11-/-

C57BL/6 mice were infected with the C. rodentium strains found in Table 2.1, as

described previously (Raymond et al., 2011). Mice were inoculated with WT C.

rodentium, C. rodentium ΔnleF or the complemented C. rodentium ΔnleF

pACYC184-nleFCR strain grown over-night in LB and re-suspended in PBS at ~5 x

109 CFU. Stool samples were collected on the days indicated and CFU/g of stool

was enumerated the next day retrospectively, by plating on LB agar with antibiotics.

On the day indicated mice were culled and segments of the colon were extracted for

analysis as described below. A minimum of 5 C57BL/6 mice per group were used

per independent repeat. For C57BL/6 Casp1/11-/- a minmum of 2 mice per group

were used per independent repeat with a final n= 8 and n=6 for WT ICC169 and C.

rodentium ∆nleF infections, respectively. Procedures were approved by the local

Ethical Review Committee and were performed in accordance with the Animals

Scientific Procedures Act (1986).

2.18 Sample collection and FACs analysis

Four cm segments of the terminal colons were cut, opened longitudinally and rinsed

in sterile PBS and placed in 4ml of RPMI-1640 supplemented with 10% FBS, 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). First, the tissue was homogenized using ‘intestine’

Page 82: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

82

setting followed by incubation at 37°C, 5% CO2 for 30 min in a shaking incubator

and then a final dissociation step was performed using ‘Lung 2’ setting on the

gentleMACS dissociator. The digested preparation was disrupted to a single cell

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

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

0.5-1x106 cells/ml.

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

BSA 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 eBio-science. Data

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

analysed with the FlowJO (Tress star) analysis programme. Data is shown as a

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

determined by their position on the forward and 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.

2.19 Ex-vivo ex-plants

After 4 cm of a colon was cut for flow cytometry analysis, the next 1cm from the

same mouse was cut for ex vivo organ culture. The colon section was washed with

PBS, weighed (the average weight of this particular section of the colon was not

significantly different throughout experimental groups) and incubated in 400 μl of

Page 83: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

83

RPMI containing P/S, gentamicin and FBS at 37°C for 24 h. Concentrations of IL-18

(eBioscience) and KC (CXCL1, R&D systems) were measured as described above.

2.20 Statistical analysis

All in vitro data was analysed by using the non-parametric Kruskal-Wallis test and

post-hoc Dunns multiple comparison test of all columns using the GraphPad Prism 5

software and represented as the mean +/- standard deviation, unless stated

otherwise. A P value less than 0.05 (P<0.05) was considered significant. All in vivo

data was analysed using the non-parametric Mann Whitney test using the GraphPad

Prism 5 software and represented as the average +/- standard error, unless stated

otherwise. A P value less than 0.05 (P<0.05) was considered significant.

Page 84: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

84

Kanr, Kanamycin resistance 50 µg.ml; Cmr, Chloramphenicol resistance 10 µg.ml; Nalr, Nalidixic acid.

Table 2 1 Bacterial strains used in this study.

Strains Description Source ICC481 EPEC O127:H6 strain E2348/69 (Levine et al., 1978) ICC1082 EPEC O127:H6 strain E2348/69 ΔnleF, Kanr This study ICC192 EPEC O127:H6 strain E2348/69 ΔescN, Kanr (Garmendia et al., 2004) ICC1063 EPEC O127:H6 strain E2348/69 ΔPP4/IE6, Cmr, Kanr (Pearson et al., 2011) - BL21 Star Invitrogen - Y187, Saccharomyces cerevisiae Clontech - AH109, Saccharomyces cerevisiae Clontech ICC169 Citrobacter rodentium, Nalr (Schauer & Falkow, 1993) - Citrobacter rodentium, Nalr, Kanr This study - STBL2 Invitrogen

Page 85: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

85

Table 2 2 Plasmids and Primers used for cloning in this study.

Plasmids Description Source pACYC184 E. coli plasmid cloning vector containing the p15A origin of replication, Cmr NEB pACYC184-derivative

Expressed protein

Primers 5’�3’ RS -

- NleFE69 TTTGATATCAAGAAGGAGATATACCTACGTAATGTTACCAACAAGTGGTTCTTCAGC TGCAGGATCCTCATCCACATTGTAAAGATCCTTTGTT

EcoRV BamHI

This study

- NleFCR TTTGATATCAAGAAGGAGATATACCTACGTAATGTTTGGTATGTTACCAGCTAGTAG TGCAGGATCCCTATCCAATTTGCAAAGATCCTTTGT

EcoRV BamHI

This study

pBAD22 pBAD-Arabinose inducible bacterial expression vector; Ampr (Guzman et al., 1995)

pBAD22-derivative

Expressed protein

Primers 5’�3’ RS

pICC1667 NleFE69 CCGCCGGGAATTCATGTTACCAACAAGTGGTTCTTCAGA TGCAGGATCCTCATCCACATTGTAAAGATCCTTTGTT

EcoRI PstI

This study

pKD4 Plasmid backbone containing Kanr template (Datsenko & Wanner, 2000)

pET28a(+) Bacterial expression vector with T7 lac promoter; N-terminal 6x His tag with thrombin cleavage site or C-terminal 6x His tag; Kanr

Novagen

pET28a(+)-derivative

Expressed protein

Primers 5’�3’ RS -

pICC1659 His-NleFE69 CGCGGGATCCATGTTACCAACAAGTGGTTCTTCAGC AAATATGCGGCCGCTCATCCACATTGTAAAGATCCTTTGTT

BamHI NotI

This study

pICC1672 His-NleFE69

CGCCATATGTTACCAACAAGTGGTTCTTCAGC ATCTCGAGTTAAGATCCTTTGTTGTAAGTAAGAT

NdeI XhoI

This study

pICC1660 His-NleFE69

1-185 CGCGGGATCCATGTTACCAACAAGTGGTTCTTCAGC ATTGCGGCCGCTTAAGATCCTTTGTTGTAAGTAAGAT

BamHI NotI

This study

- His-NleFCR CGCGGGATCCATGTTTGGTATGTTACCAGCTAGTAG AAATATGCGGCCGCCTATCCAATTTGCAAAGATCCTTTGT

BamHI NotI

This study

- His-NleFCR

1-185

CGCGGGATCCATGTTTGGTATGTTACCAGCTAGTAG ATTGCGGCCGCTTAAGATCCTTTGTTGTAAGTAAGAT

BamHI NotI

This study

- His-Caspase-4

CGCGCGGATCCATGGCAGAAGGCAACCACAGAAA AAATATGCGGCCGCTCAATTGCCAGGAAAGAGGTAGAAAT

BamHI NotI

This study

- His-Caspase-11

CGCGCGGATCCATGGCTGAAAACAAACACCCTGAC AAATATGCGGCCGCCTCAGTTGCCAGGAAAGAGGTAG

BamHI NotI

This study

- His-Caspase-4 C258S

catcattgtccaggccTCCagaggtgcaaacc ggtttgcacctctGGAggcctggacaatgatg

- This study

- His-Caspase-4 p20

CGCGGGATCCCAAATATCCCCCAATAAAAA AAATATGCGGCCGCCTAGTCTCTGACCCACAGTTCC

BamHI NotI

This study

- His-Caspase-4 p10

CGCGGGATCCGCTGTTTACAAGACCCACGT AAATATGCGGCCGCTCAATTGCCAGGAAAGAGGT

BamHI NotI

This study

pGADT7 Plasmid for expression of proteins in yeast with a GAL4 activation domain fusion; N-terminal HA tag; Ampr

Clontech

pGADT7-derivative

Expressed protein

Primers 5’�3’ RS -

- DHFR CGCCATATGATGGTTGGTTCGCTAAACTGCATCG CGCGGGATCCTTAATCATTCTTCTCATATACTTCA

NdeI BamHI

This study

- DHFRL1 GCCATATGATGTTTCTTTTGCTAAACTGCATCG CGCGGGATCCTTAATCATCCTTCTCACATACTTCA

NdeI BamHI

This study

- TMP21 N/A N/A Schroeder, G. - GGCT ATATCCCGGGTTATGGCCAACTCGGGCTGCAAGGACGTC

CCTAAAGAGTTTGTGTTTCCCCCTTTTTG XmaI XhoI

This study

- HBXIP GCCGGGAATTCATGGAGCCAGGTGCAGGTCACCT CGCGGGATCCTCAAGAGGCCATTTTGTGCACTG

EcoRI BamHI

This study

Page 86: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

86

- HBXIP Y2H

GCCGGGAATTCATGGCCTGTGGTGGAGAAGGA CGCGGGATCCTCAAGAGGCCATTTTGTGCACTG

EcoRI BamHI

This study

- HBXIP short isoform

GCCGGGAATTCATGGAGGCGACCTTGGAGCA CGCGGGATCCTCAAGAGGCCATTTTGTGCACTG

EcoRI BamHI

This study

- Caspase-5 CGCGCGGATCCATATGTTCAAAGGTATCCTTCAGAGT ATCTCGAGTCAATTGCCAGGAAAGAGGTAGAAA

BamHI XhoI

This study

- Caspase-11

GCCGGGAATTCATGGCTGAAAACAAACACCCTGAC CGCGGGATCCCTCAGTTGCCAGGAAAGAGGTAG

EcoRI BamHI

This study

- Caspase-12

GCCGGGAATTCATGGCGGCCAGGAGGACACATGAAAG CGCGGGATCCTGCTAATTCCCGGGAAAAAGGTAGAA

EcoRI BamHI

This study

- Caspase-4 ATATCCCGGGTATGGCAGAAGGCAACCACAGAAA CGCGGGATCCTCAATTGCCAGGAAAGAGGTAGAAATATC

XmaI BamHI

This study

- Caspase-4 P30

ATATCCCGGGTCAAATATCCCCAATAAAAA CGCGGGATCCTCAATTGCCAGGAAAGAGGTAGAAATATC

XmaI BamHI

This study

- Caspase-4 P20

ATATCCCGGGTCAAATATCCCCAATAAAAA CGCGGGATCCCTAGTCTCTGACCCACAGTTCC

XmaI BamHI

This study

- Caspase-4 P10

ATATCCCGGGT GCTGTTTACAAGACCCACGT CGCGGGATCCTCAATTGCCAGGAAAGAGGTAGAAATATC

XmaI BamHI

This study

pGADT7-caspase-4 P30 derivative

Expressed protein

Primers 5’�3’ RS This study

- Caspase-4 P30 R159A

GAATGGAGCTGACTTTGACATCA gcCGGAGGCAGATGGTCAAACTC

N/A This study

- Caspase-4 P30 H237A

TGGCATCCTGGAGGGAATCTGCG gcAGACATGAGTACCAAGAATG

N/A This study

- Caspase-4 P30 W313A

GAGAGACAGCACAATGGGCTCT gcGGACACGTTGTGTGGCGTTGA

N/A This study

- Caspase-4 P30 R314A

AGACAGCACAATGGGCTCTATC gcCCAGGACACGTTGTGTGGCG

N/A This study

pGBT9 Plasmid for expression of proteins in yeast with a GAL4 binding domain fusion; Ampr Clontech pGBT9-derivative Expressed

protein Primers 5’�3’ RS

pICC1664 NleFE69 CGCGGATCCCGATGTTACCAACAAGTGGTTCTTCAGCAA AAAACTGCAGTCATCCACATTGTAAAGATCCTTTGTT

BamHI PstI

This study

pICC1666 NleFE69 1-

185

CGCGGATCCCGATGTTACCAACAAGTGGTTCTTCAGCAA CTGCACTGCAGTTAAGATCCTTTGTTGTAAGTAAGAT

BamHI PstI

This study

pICC1669 NleFCR CGCGGATCCCGATGTTTGGTATGTTACCAGCTAGTAG AAAACTGCAGCTATCCAATTTGCAAAGATCCTTTGT

BamHI PstI

This study

pGKBT7 Plasmid for expression of proteins in yeast with a GAL4 activation domain fusion; N-terminal Myc tag; Ampr; Kanr

Clontech

pGKBT7-derivative

Expressed protein

Primers 5’�3’ RS

pICC1665 NleFE69 CGCGGATCCCGATGTTACCAACAAGTGGTTCTTCAGCAA AAAACTGCAGTCATCCACATTGTAAAGATCCTTTGTT

BamHI PstI

This study

pKD46 Codes for lambda-red recombinase; Ampr (Datsenko & Wanner, 2000)

pSA10 pKK177-3 derivative containing lacI, IPTG inducible; Ampr (Schlosser-Silverman et al., 2000.)

pSA10-derivative Expressed protein

Primers 5’�3’ RS

pSA10-nleE NleE1E69 N/A N/A (Vossenkämper et al., 2010)

pRK5:myc Vector for the expression of proteins with N-terminal Myc tag in mammalian cells Clontech pRK5:myc-derivative

Expressed protein

Primers 5’�3’ RS -

pICC1661 Myc-NleFE69

CGCGCGGATCCATGTTACCAACAAGTGGTTCTTCAG TGCACTGCAGATCATCCACATTGTAAAGATCCTTTG

BamHI PstI

This study

pGEM-T High copy cloning plasmid for with insertional inactivation of β-galactosidase; Ampr Promega pGEM-T Insert Primers 5’�3’ RS

Page 87: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

87

derivative - nleFCR

flanks with Kan cassette

CGCGGATCCGCTACTAGCTGGTAACATACC CGCGGATCCTCGTTGAAGGATGCAGTGATG

BamHI BamHI

This study

pICC1662 Myc-NleFE69 1-

185

CGCGCGGATCCATGTTACCAACAAGTGGTTCTTCAG CTGCACTGCAGTTAAGATCCTTTGTTGTAAGTAAGAT

BamHI PstI

This study

pICC563 Myc-GFP N/A N/A (Clements et al., 2011)

Myc-NleFCR

CGCGGGATCCATGTTTGGTATGTTACCAGCTAGTAG TGCACTGCAGTTAAGATCCTTTGTTGTAAGTAAGAT

BamHI PstI

This study

pRK5:ha Vector for the expression of proteins with N-terminal HA tag in mammalian cells under the CMV promoter

Clontech

pRK5:ha-derivative

Expressed protein

Primers 5’�3’ RS

- HA-TMP21 N/A N/A Schroeder, G. - HA-Cherry N/A N/A Frankel TOPO Blunt II Parental plasmid for TOPO cloning of blunt-end PCR products; Kanr Invitrogen TOPO Blunt II derivative

Insert Primers 5’�3’

- nleFE69 with flanks

GATATGGAAGATACAACAAAAGTGTT ACAGAAGAGGCAGGCCAGCA

This study

pICC1674 nleFE69 flanks with Kan cassette

TTAGAGGCCTGTGAGCCTGT ATCAAAACCCCCTTAACAAATAAATC TTAGAGGCCTGTGAGCCTGT ATCAAAACCCCCTTAACAAATAAATC

This study

pACYC-Duet-1 IPTG inducible bi-directional bacterial expression vector, Cmr Novagen pACYC-DUET-1 derivative

Expressed protein

Primers 5’�3’ RS

- Caspase-4 P10

CCGGATATCGGCTGTTTACAAGACCCACGTGGAG ATCTCGAGTCAATTGCCAGGAAAGAGGTAGAAA

EcoRV XhoI

This study

- Caspase-4 His-P20 C258S /P10

CGCGGGATCCGCAAATATCCCCCAATAAAAA AAATATGCGGCCGCCTAGTCTCTGACCCACAGTTCC

BamHI NotI

This study

pMAL-c2x Bacterial expression vector for expression and purification of desired proteins with an N-terminal maltose binding protein (MBP) tag, Ampr

NEB

pMAL-c2x-derivative

Expressed protein

Primers 5’�3’ RS -

MBP-NleFE69

CCGCCGGGAATTCATGTTACCAACAAGTGGTTCTTCAGC AAAACTGCAGTCATCCACATTGTAAAGATCCTTTGTT

EcoRI PstI

This study

MBP-NleFE69 1-

185

CCGCCGGGAATTCATGTTACCAACAAGTGGTTCTTCAGC CTGCACTGCAGTTAAGATCCTTTGTTGTAAGTAAGAT

EcoRI PstI

This study

pMXs-IRES-Puro

Retroviral plasmid for viral package signal, transcription and processing, puromycin selectable, Ampr

Invitrogen

pMXs-IRES-Puro-derivative

Expressed protein

Primers 5’�3’ RS

pICC1646 Caspase-4 CGCGCGGATCCATGGCAGAAGGCAACCACAGAAA ATCTCGAGTCAATTGCCAGGAAAGAGGTAGAAA

BamHI XhoI

This study

- Caspase-5 CGCGCGGATCCATGTTCAAAGGTATCCTTCAGAGT ATCTCGAGTCAATTGCCAGGAAAGAGGTAGAAA

BamHI XhoI

This study

- Caspase-11

CGCGCGGATCCATGGCTGAAACAAACACCCTGAC ATCTCGAGTCAGTTGCCAGGAAAGAGGTAG

BamHI XhoI

This study

- Myc-NleFE69

CGCGCAATTGGCCGCCACCATGGAGCAGAAGCTG TGCAGGATCCTCATCCACATTGTAAAGATCCTTTGTT

MfeI BamHI

This study

- Myc-NleFE69 1-

185

CGCGCAATTGGCCGCCACCATGGAGCAGAAGCTG TGCAGGATCCTTAAGATCCTTTGTTGTAAGTAAGAT

MfeI BamHI

This study

- YFP N/A N/A Shenoy, A. pMXs-CMV-yfp derivative

Expressed protein

Primers 5’�3’ RS Shenoy, A.

- YFP miR30

ctgaaacataaaatgaatgctcgAGAAGGTATATTGCTGTTGACAGTGAGCGCcgactgtccatgacaagatattagtgaagccacagat

MfeI

This study

Page 88: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

88

CASP4 aagatattagtgaagccacagatgtaatatcttgtcatggacagtcgTTGCCTACTGCCTCGGACTTCAAGaattgttgttgttaacttgcaa

- YFP miR30 lacZ non-targeting control

AATTGTTGTTGTTAACTTGCtcgagaaggtatattgctgttgacagtgagcgACGTCGTATTACAACGTCGTGAtagtgaagccacagat TTATTTGTAACCATTATAActtgaagtccgaggcagtaggcaACGTCGTATTACAACGTCGTGAtacatctgtggcttcactaTCACGAC

PsiI This study

Page 89: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

89

Table 2 3 cDNA and templates used in this study.

pICC Plasmid backbone and cDNA template Source pICC1653 pMD-CASP5 Open Biosystems pICC1651 pDNR-LIB-CASP4 Open Biosystems pICC1650 pDNR-LIB-Casp11 Open Biosystems - pENTR221-DHFR Parle-McDermott, A. - pDEST17-DHFRL1 Parle-McDermott, A. - pOBT7-GGCT Open Biosystems - pHBXIP Garcia-Saez, I. pICC893 pKD4-Kanamycin cassette (Datsenko & Wanner, 2000)

Page 90: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

90

Primer Sequence Assay/amplicon 1F 1R

GATATGGAAGATACAACAAAAGTGTT ACAGAAGAGGCAGGCCAGCA

Mutagenesis amplicon Mutagenesis amplicon

pKD4F pKD4R

TGTGTAGGCTGGAGCTGCTTC CATATGAATATCCTCCTTAGTTCC

Kanamycin cassette Kanamycin cassette

3F 3R

GTAAAGGTGGTAATGCTGTCGT GAAGGAATACAAAGCCGTCAC

Mutagenesis screening Mutagenesis screening

AD-LD-F AD-LD-R

CTATTCGATGATGAAGATACCCCACCAAACCC GTGAACTTGCGGGGTTTTTCAGTATCTACGATT

Y2H screening primers

shRNAF TGTACAAGTCCGGACTCAGATGATCGACTTCTTAACCCAACAGAAGGCTCGAGAAGGTATATTGCTG

shRNA amplicon

shRNAR GCGGCCGCGGCGCGCCGGCCGAATTCTAGCCCCTTGAAGTCCGAGGCAGTAGG

shRNA amplicon

IL-8F IL-8R

AGAAACCACCGGAAGGAACCATCT AGAGCTGCAGAAATCAGGAAGGCT

RT-qPCR RT-qPCR

GAPDHF GAPDHR

TCGACAGTCAGCCGCATCTTCTTT ACCAAATCCGTTGACTCCGACCTT

RT-qPCR RT-qPCR

CASP4F CASP4R

GCAGACTCTATGCAAGAGAAG TGGTCCAGCCTCCATATT

RT-qPCR RT-qPCR

CASP8F CASP8R

GGATGGCCACTGTGAATAA CAGTCAGGATGGTGAGAATATC

RT-qPCR RT-qPCR

CASP9F CASP9R

GTGCTCTTGAGAGTTTGAGG ACAGCCGTGAGAGAGAAT

RT-qPCR RT-qPCR

4F TTCAGTCATTACCAACACAGG Mutagenesis amplicon 4R TTCCTATGTGGTGAATTGCG Mutagenesis amplicon 5F 6F

CGCGGATCCGCTACTAGCTGGTAACATACC CGCGGATCCTCGTTGAAGGATGCAGTGATG

Inverse PCR Inverse PCR

7F ATATATGATGCCAATGGCGAT Mutagenesis screening 7R GACTAGCATTAACACTCATGC Mutagenesis screening

Table 2 4 Primers used for mutagenesis and RT-qPCR in this study.

Page 91: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

91

Results: Chapter 3 - Identification and confirmation

of host interacting partners

Page 92: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

92

3.1 A Y2H screen identifies caspase-4 as an interacting partner of NleFE69

In order to determine the function of EPEC E2349/69 NleF (NleFE69) a yeast-2-hybrid

(Y2H) screen was carried out to identify putative host binding partners. A Y2H

screen utilises the GAL4 promoter to identify interacting proteins of a chosen bait

protein from a HeLa cell cDNA prey library. The bait protein is expressed in pGKBT7

or pGBT9 along with an N-terminal GAL4 binding domain in Saccharomyces

cerevisiae AH109; this is then mated with Saccharomyces cerevisiae Y187 pre-

transformed with pGADT7 containing the library of prey proteins with a GAL4

activation domain. Interaction of the bait with a prey protein, through diploid

formation (AH109 meeting with Y187), will result in the GAL4 DNA binding domain

and activation domain being brought into close proximity. This in turn activates

transcription of four reporter genes (ADE2, HIS3, MEL1, and lacZ). Transcription of

the reporter genes ADE2 and HIS3 results in the expression of the essential amino

acids adenine (Ade) and histidine (His), respectively. Transcription of MEL1 and lacZ

results in the expression of α-galactosidase and β-galactosidase, respectively. The

bait plasmids pGKBT7/pGBT9 and prey plasmid pGADT7 further confer selectivity

through the expression of tryptophan (Trp) and leucine (Leu), respectively. This

allows for the screening of clones expressing proteins that interact by plating on the

selective yeast media SD SDO (-Trp), DDO (-Leu and –Trp), TDO (-Ade, -His,- Leu)

and QDO (-Trp, -Leu, -Ade and –His), while supplementation of the media with X-α-

Gal and activation of the reporter gene MEL1 results in the secretion of α-

galactosidase which in turn reacts with the chromogenic substrate, causing a blue

colouration of a positive colony.

Page 93: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

93

Expression of NleFE69 in AH109 had no effect on cell growth kinetics and no auto-

activity (ability to grow on DDO/TDO/QDO; data not shown). Screening of NleFE69,

with a mating efficiency of 52.1%, resulted in 320 positive clones. Multiple re-

streaking of positive clones on selective QDO X-α-Gal media reduced the number of

positive clones to 149. All colonies were checked by colony PCR to verify the

presence of pGADT7 and an insert by using the primers AD-LD-F/R shown in Table

2.4 and sent for sequencing. The Y2H screen identified 18 unique hits of which there

were multiple clones for COP9 constitutive photomorphogenic homolog subunit 5

(COPS5) and 21 hits for caspase-4 (Table 3.1).

Table 3 1: A Y2H screen for NleFE69 identifies caspase-4 as a putative host

target. FP; false positive, UT; untested Hit Gene ID No. Interaction by YDH

COP9 constitutive photomorphogenic homolog subunit 5 (COPS5) 10987 COPS5 35 FP

CASP4 837 CASP4 21 Yes

Microtubule associated protein 4 4134 MAP4 2 UT

Cell Division cyclin kinase-7 8317 CDC7 1 UT

Dynactin subunit 2 10671 DCTN1 1 UT

Cytokine induced apoptosis inhibitor 1 57019 CIAPIN1 1 UT

Vesicle transport protein USE1 55850 USE1 1 UT

ATPase family AAA domain containing 5 79915 ATAD5 1 UT

Metal response element binding transcription factor 2 22823 MTF2 1 UT

WW domain binding protein 11 51729 WBP11 1 UT

Vacuole membrane protein 1 81671 VMP1 1 UT

CG1-130 51020 HDDC2 1 UT

Methylmalone aciduria and homocystinuria type D protein 27249 MMADHC 1 UT

Hepatitis B virus X-interacting protein 457120 HBXIP 1 FP

Transforming growth factor B 84897 TBRG1 1 UT

Cleavage and Polyadenylation specific factor 2, 100 KDa 53981 CPSF2 1 UT

Gamma-glutamylcyclotransferase 79017 GGCT 1 FP

IMPACT (Imprinted and ancient gene protein homolog) 55364 IMPACT 1 UT

Page 94: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

94

COPS5 is the mostly highly cited false positive for the Clontech Y2H screen and was

therefore dismissed. To verify caspase-4 as a true hit the full length cDNA of CASP4

isoform gamma was cloned into pGADT7. Co-transformation of AH109 with an

empty plasmid as a negative control or with pGADT7-CASP4 and plating on DDO or

the selective QDO media confirmed caspase-4 as a true interacting partner of

NleFE69 in the yeast-direct-hybrid (YDH) system (Fig. 3.1A)

3.2 NleFE69 specifically interacts with caspase-4

Caspase-4 is an inflammatory caspase that controls ER stress induced apoptosis

and may be required for activation of the non-canonical inflammasome (Shi et al.,

2014; Hitomi et al., 2004). Caspase-4 shares significant amino acid sequence

similarity with the inflammatory caspase caspase-5; which are both putative

functional orthologs of murine caspase-11 (Fig. 3.2A). Due to the lack of commercial

CASP5 cDNA the templates for CASP5 were amplified directly from HeLa and THP-

1 cDNA pools. The caspase-5 transcript present in both is a previously described

isoform of caspase-5 that is annotated as an N-terminally truncated form of the

Figure 3 1: NleFE6 interacts with full length caspase-4. (A) A Yeast-direct-hybrid assay co-transforming AH109 with full length NleFE69 (pGBT9-nleFE69) and caspase-4 isoform-γ (pGADT7-CASP4). Results are representative of at least three biological repeats.

Page 95: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

95

canonical isoform alpha (Fig. 3.2B) (Munday et al., 1995). This HeLa isoform was

used throughout further studies. AH109 were co-transformed with pGBT9::nleFE69

and pGADT7::CASP5 or pGADT7::CASP4 and plated on DDO and QDO to

determine if NleFE69 specifically interacts with caspase-4. NleFE69 specifically

interacted with caspase-4 and not caspase-5 in the YDH system (Fig. 3.2C).

Page 96: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

96

Figure 3 2 Caspase-5 shares significant homology with caspase-4 but does not interact with NleFE69. (A) Clustal W2 multi-alignment of murine caspase-11, caspase-4γ and caspase-5α/f. (B) Alignment of CASP5α and CASP5 amplified from HeLa cDNA. (C) A Yeast-direct-Hybrid assay co-transforming AH109 with full length NleFE69(pGBT9-nleFE69) and caspase-4 isoform-γ (pGADT7-CASP4) or caspase-5 from HeLacDNA (pGADT7-CASP5). Results are representative of three biological repeats.

Page 97: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

97

3.3 NleFE69 interacts with active caspase-4 dependent on its’ C-terminal motif

LQCG

Caspase-4, as a zymogen, contains a p22 CARD domain which is subsequently

cleaved to release its active (p30) form (Fig 3.3A). NleFE69 was determined to

interact with both full length and the p30 domain of caspase-4 by YDH, suggesting it

may inhibit substrate binding/protease activity directly (Fig. 3.3B). The p20 domain

was auto-active and therefore interaction of p20 and NleFE69 alone could not be

confirmed, although p10 alone was not sufficient to confer binding (Fig. 3.3B).

Blasche and colleagues previously showed that NleFE69 interacts with active

caspase-9 through direct interaction of the caspase-9 substrate domain and the four

C-terminal amino acids (LQCG) of NleFE69 (Blasche et al., 2013). To investigate if

interaction of NleFE69 and caspase-4 is dependent on the C-terminal of NleFE69 the

YDH was repeated using an NleFE69 mutant with the four C-terminal amino acids

deleted (NleF1-185 E69). Deletion of the four C-terminal amino acids, or mutation of the

amino acids of the putative substrate binding domain W313, R314 or R152 of

caspase-4, abolished interaction in the YDH system (Fig. 3.3B-C). Alternatively

mutation of the putative substrate binding domain amino acid H237 had no effect on

binding (Fig. 3.3C). The putative substrate binding domain and active site of

caspase-4 is formed of both the p20 and p10 subunits. Single mutations in either the

p10 subunit (W313A or R314A) or the p20 subunit (R152A) suggested that the

interaction of caspase-4 and NleFE69 required both the p20 and p10 subunits (Fig.

3.3C). NleF of C. rodentium (NleFCR) was auto-active in the YDH system and

therefore could not be screened using this assay.

Page 98: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

98

Figure 3 3: The C-terminal motif LQCG of NleFE69 interacts with active caspase-4. (A) Schematic of caspase-4 structural activation through domain cleavage and oligomerisation. A Yeast-direct-hybrid assay to access and map interaction of NleFE69 and NleF1-185 E69 with (B) pro-caspase-4 and caspase-4 p30 and (C) caspase-4 p30 with single amino acid mutations of the putative caspase-4 substrate domain: R152A, H237A, W313A and R314A. Empty pGADT7 and pGBT9 were used as negative controls. Results are representative of at least three biological repeats. p30; caspase-4 p20 and P10 subunits and the inter-linker domain.

Page 99: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

99

3.4 NleFE69 binds and inhibits the catalytic activity of caspase-4/11

Through the YDH assay NleFE69 was shown to bind to the p30 domain of caspase-4

suggesting it may inhibit protease activity. To determine if NleFE69 binding to

caspase-4 inhibits its proteolytic activity; first, an activity assay was developed.

Recently it was shown that upon binding to LPS full length caspase-4, and the

murine ortholog caspase-11, undergo self-proteolytic processing, oligomerisation

and activation (Shi et al., 2014). Activation of each caspase was determined using a

fluorogenic caspase substrate that upon cleavage can be detected by fluorescence

emission at a determined wave length. The principle of self-activation in the

presence of LPS was employed to design an in house caspase activity assay for

both caspase-4 and -11.

3.4.1 Developing a caspase-4/-11 activity assay

Full length caspase-4 and caspase-11 were cloned into the IPTG inducible bacterial

expression plasmid pET28a(+) and expressed in BL21 Star and analysed by

immunoblotting. Expression and bacterial lysis was sufficient to induce self-

proteolytic cleavage of both caspase-4 and caspase-11, as evidenced by the

presence of the p20 cleavage product (Fig. 3.4A-B). As a control a protease dead

caspase-4 mutant (C258S) was expressed in BL21 Star. Mutation of the active site

cysteine prevented cleavage of caspase-4 suggesting that the cleavage products

observed were not a result of degradation or bacterial proteases. The carboxy-

peptidase substrate LEVD is a known substrate and inhibitor of the in vitro activity of

caspase-4. Caspase-4/-11 bacterial lysates were incubated with LEVD with a

fluorogenic 7-amino-4-trifluoromethylcoumarin tag (LEVD-AFC). Enzymatic cleavage

of the fluorogenic substrate releases AFC switching from an emission wavelength of

400 nm to 495-505 nm. This can be used to detect enzymatic and caspase activity.

Page 100: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

100

Addition of LEVD-AFC to both caspase-4 and caspase-11 bacterial lysates resulted

in specific cleavage of LEVD-AFC as observed by the increase in relative fluorescent

units (Fig. 3.4B). No LEVD specific proteolytic activity was observed for BL21 over-

expressing caspase-4 C258S or with BL21 lysate alone (Fig. 3.4A).

Figure 3 4 Caspase-4/-11 undergoes self-proteolytic cleavage and oligomerisation to cleave the fluorescent substrate LEVD-AFC. An activity assay using the caspase-4 substrate reporter Ac-LEVD-AFC was constructed using lysate of BL21 expressing (A) pro-caspase-4 or (B) pro-caspase-11. Caspase-4 C258S and BL21 lysate were used as negative controls. Self proteolytic activation was determined by western blot. Results are an average of three independent experiments in duplicate. RFU; relative fluorescent units

Page 101: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

101

3.4.2 Purification of NleFE69 for in vitro activity assays

To determine if NleFE69 functions to inhibit the catalytic activity of caspase-4/-11;

NleFE69, NleF1-185 E69 and NleFCR were cloned into the IPTG inducible bacterial

expression plasmid pET28a(+) and expressed with an N-terminal His-tag in BL21

Star. Initially expression of NleF for purification was optimised for BL21 star over-

expressing NleFE69. Expression of NleFE69 was induced at a range of temperatures

at 0.5 mM IPTG and analysed by SDS-PAGE gel and immunoblotting. Induction of

protein expression was optimum at 18 ºC as indicated by the band present in the

induced lanes for both the SDS-PAGE gel and Western blot at ~ 21 kDa (Fig 3.5A-

B).

In all further experiments expression of NleFE69 and its homologs were carried out

under these conditions. His-tagged proteins were purified by affinity tagged

purification and further purified by gel filtration. Solubility and purity were analysed by

SDS-PAGE gel (Fig. 3.6 A-C). His-tagged NleFE69, NleF1-185 E69 and NleFCR were

Figure 3 5 Optimisation of IPTG induced expression of NleFE69 in BL21 Star. E. coli BL21 Star transformed with pET28a-nleFE69 were sub-cultured at 37 ºC until OD600 of 0.6 before inducing with 0.5 mM IPTG at either 37 ºC or 30 ºC for 3 h or 18 ºC for 16 h. Bacterial pellets were re-suspended in LDS and analysed by SDS-PAGE gel or Western blot using anti-His-HRP as indicated in Material and methods. L, Ladder

Page 102: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

102

highly soluble and stable upon lysis and purification (Fig. 3.6). Only the purist

fractions from gel filtration were collected. These were concentrated and stored at -

80 ºC for use in further assays.

Figure 3 6 Purification of NleF homologs of A/E pathogens. Induced E. coli BL21Star transformed with (A) pET28a-nleFE69, (B) pET28a-nleF1-185 or (C) pET28a-nleFCRwere lysed by Emulsiflex and purified by His-tagged affinity purification and gel filtration. Fractions were analysed by SDS-PAGE gel. UV peaks from gel filtration were run to test presence and purity of NleF ~22-23 kDa. L, Ladder; -, un-induced; +, induced; I, insoluble; S, soluble; FT, flow-through; W, wash; E, elution; F, Gel filtration fraction.

Page 103: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

103

3.4.3 NleF of A/E pathogens inhibit the activity of caspase-4/-11

Caspase-4/-11 activity assays were repeated as above with the addition of purified

NleFE69, NleF1-185 E69 and NleFCR at a concentration range of 1 pM to 50 nM. Addition

of recombinant NleFE69 efficiently inhibited the rate of activity of caspase-4 in a dose-

dependent manner with an IC50 of 5 nM; NleF1-185 E69 required a significantly higher

inhibitory concentration with an IC50 of 25.2 nM (Fig. 3.7A).

Figure 3 7 NleF inhibits caspase-4 and -11 activity in vitro. (A) Dose dependent inhibition of caspase-4 activity with recombinant purified NleFE69 or NleF1-185 E69 (B) Comparison of caspase-4 activity upon the addition of NleFE69 or NleFCR at 1 nM. (C) Dose dependent inhibition of caspase-11 activity with recombinant purified NleFCR. (D) Comparison of caspase-11 activity upon the addition of NleFE69 or NleFCR at 50 nM.. Purity was analysed by SDS-PAGE gel and fluorescence was measured at 30 min. Representative of at least three biological repeats in duplicate.

Page 104: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

104

This confirmed that NleFE69 directly interacts with the active form of caspase-4 via its

C-terminal motif LQCG to inhibit in vitro caspase protease activity. Infection of

immortalised mouse macrophages (iMM) with the mouse restricted A/E pathogen C.

rodentium activates caspase-11, the putative functional ortholog to human caspase-

4, leading to activation of the non-canonical inflammasome (Kayagaki et al., 2011).

Therefore we tested the ability of NleFCR, which shares 84% amino acid sequence

identity with NleFE69, to inhibit the protease activity of caspase-11. Addition of

recombinant NleFCR successfully inhibited the activity of caspase-11 and cleavage of

LEVD-AFC in a dose-dependent manner with an IC50 of 13 nM (Fig. 3.7C).

Interestingly NleFE69 inhibited caspase-4 activity more efficiently that NleFCR and vice

versa for caspase-11 (Fig. 3.7 B and D), suggesting a host specific adaption. While

this demonstrated a conserved function of the NleF homologs to inhibit the activity of

caspase-4/-11, the mechanism of inhibition remains unknown.

3.5 Preliminary data suggests NleFE69 forms a complex with the active

caspase-4 hetero-tetramer

OspC3, a T3SS effector of the bacterial pathogen Shigella spp., is delivered during

infection of epithelial cells to inhibit the activity of caspase-4 (Kobayashi et al., 2013).

OspC3 binds to the p20 subunit of caspase-4 to prevent oligomerisation and

formation of the heterotetramer and subsequent activation of the caspase

(Kobayashi et al., 2013). The YDH data suggests that NleFE69 binding to caspase-4

is dependent on residues present in both the p20 and p10 subunits. Due to the

insolubility of the recombinant caspase-4 p10 subunit the binding of the individual

domains and NleFE69 could not be tested directly. To determine if NleFE69 forms a

Page 105: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

105

complex with the heterotetramer to prevent activity/ oligomerisation, recombinant

NleFE69 was co-expressed and co-purified with caspase-4 p20 and p10.

3.5.1 Co-purification of active caspase-4 and NleFE69

To investigate if NleFE69 forms a complex with the active form of caspase-4 the

expression vectors pMAL-c2x-nleFE69 and pACYC-DUET-1-CASP4 His-p20/p10

were co-transformed into BL21 star and an MBP pull-down assay was carried out.

The pMal-c2x empty and pACYC-DUET-1-CASP4 His-p20 plasmids were used as

negative controls. The lysates were purified by amylose affinity chromatography and

analysed by SDS-PAGE gel. Pull-down of His-p20 was dependent on the expression

of both the p10 subunit and MBP-NleFE69 and did not co-purify with MBP or MBP-

NleFE69 alone (Fig. 3.8A). Although p10 was not detectable by SDS-PAGE gel (Fig.

3.8A), this suggested that NleFE69 interaction requires both p20 and p10 subunits. To

establish if NleFE69 forms a complex with the active form of caspase-4 the MB-

NleFE69/His-p20/p10 elution was further purified by Talon purification. Following

tandem purification the elution was separated by size exclusion and the fractions

were analysed by SDS-PAGE gel (Fig. 3.8B). Three chromatographic peaks were

observed, which corresponded to free MBP-NleF69 (67 kDa), free His-p20 (31 kDa),

and a complex containing NleFE69 and caspase-4 His-p20/p10 subunits (230 kDa),

respectively. While no p10 was visible the molecular weight of the complex,

predicted at 192 kDa, indicated that MBP-NleFE69 forms a complex with the hetero-

tetramer form of caspase-4. MBP is a large molecular weight tag that would interfere

with any future structural determination work. For this reason the MBP tag was

cleaved and size exclusion was repeated to remove cleaved MBP. Under these

conditions a complex was formed that consisted of NleFE69, caspase-4 His-p20 and

p10 as determined by SDS-PAGE gel and mass spectrometry (Fig. 3.8C). Due to

Page 106: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

106

incomplete cleavage the complex varied from 100-200 kDa depending on whether

none, two or one of the MBP subunits had been removed. While this promisingly

suggested that NleFE69 forms a complex with the active form of caspase-4, which

requires both p20 and p10, the final yield under these conditions was too low to

pursue further structural studies and requires optimisation.

Figure 3 8 Recombinant NleFE69 forms a complex with the active caspase-4 complex. (A) BL21 star co-transformed with either pMal-c2x-nleFE69 or the empty vector along with pACYC-DUET-1-CASP4 His-p20 or pACYC-DUET-1-CASP4 His-p20/p10 were induced as described in materials and methods and purified by affinity chromatography. Elution for each sample was analysed by SDS-PAGE gel. (B) Amylose affinity chromatography elution was further purified by Talon affinity chromatography and size exclusion. Size exclusion fractions were analysed by SDS-PAGE gel. Peak sizes were determined using a Gel filtration molecular weight markers kit as described in materials and methods (C) Following size exclusion the co elute was Factor Xa treated and samples were analysed by size exclusion.

Page 107: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

107

3.6 Discussion

Through a Y2H screen caspase-4 was identified as a binding partner of NleFE69. This

interaction was further confirmed by, and mapped to the putative substrate binding

domain of caspase-4 and the C-terminal four amino acids of NleFE69. However, Y2H

and YDH assays have their limitations. Each protein is tagged with a large N-

terminal domain and interaction/ activation of reporter genes requires nuclear

subcellular localisation. For this reason some interactions, which may not be

optimum in the conditions of the nuclear compartment, cannot be detected via this

method or may be false. Caspase-4 along with caspase-8 and caspase-9 were

identified to bind NleFE69 through the same C-terminal dependent interaction by

Blasche and colleagues. Interaction was confirmed by luciferase pull-down assay

further validating that NleFE69 binds caspase-4 (Blasche et al., 2013). While blasche

and colleagues also identified both caspase-8 and -9, NleFE69 was not observed to

play a significant role in cell death inhibtion during infection (Blasche et al., 2013).

Therefore we pursued caspase-4 as a putative partner.

Through the construction of a caspase activity assay NleFE69 was identified to inhibit

the proteolytic activity of caspase-4. Furthermore the recombinant C. rodentium

homolog of NleF was observed to inhibit the activity of the murine ortholog, caspase-

11, in a dose dependent-manner. This observation suggested that NleF homologs

have a conserved function; to bind and inhibit inflammatory caspase activity. NleFE69

shares significantly high homology with NleF of C. rodentium, sharing a highly

conserved C-terminal domain. Deletion of the C-terminal motif LQCG significantly

abrogated the ability of NleFE69 to inhibit the in vitro activity of caspase-4.

Interestingly, the four C-terminal amino acid domain of NleFCR differs to that of

NleFE69 by a single substitution (C188I). This substitution may be a selective and

Page 108: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

108

adaptive advantage for the host specific inhibition of caspase-4 or caspase-11 by

NleFE69 and NleFCR, respectively. In support of this NleFE69 was unable to inhibit

caspase-11 activity in vitro as efficiently as NleFCR and vice versa for caspase-4.The

four C-terminal amino acids of NleFE69 LQCG and the four C-terminal amino acids of

NleFCR LQIG may represent novel peptides and candidates for the synthetic peptide

fluoromethylketone caspase inhibitors. Although whether the four amino acid

peptides alone are sufficient to inhibit caspase activity requires testing.

Enteric pathogens have been previously reported to inhibit caspase activity in a

T3SS-dependent manner; including caspase-4. Although, this is the first

EPEC/EHEC effector identified to do so. The Shigella spp. effector OspC3 binds the

p20 subunit of caspase-4 to inhibit oligomerisation and functional/structural activation

(Kobayashi et al., 2013). Alternatively, Yersinia spp. can inhibit inflammasome

activation by injecting YopK to mask T3SS needle detection, indirectly inhibiting

caspase activation (Brodsky et al., 2010). Blasche and colleagues identified

caspase-4, 8 and -9 as binding partners of EHEC NleF; where they solved the NleF-

EHEC-caspase-9 complex by crystallography. In our study interaction of caspase-4

and NleFE69 was identified by YDH to require residues present in both the p20 and

p10 subunits, indicating a different mechanism of inflammasome and caspase-4

inhibition in comparison to Ospc3. This was confirmed by co-purification of the

caspase-4 heterotetramer complex along with NleFE69. Here we propose a model of

inhibition that reflects that of the caspase-9/NleF complex and crystal structure

solved by Blasche and colleagues – where the C-terminal of NleFE69 associates with

the caspase-4 substrate binding domain (Blasche et al., 2013).

To confirm this model the crystal structure of caspase-4 would need to be solved.

However the final yield of the caspase-4/NleFE69 complex was too low to pursue

Page 109: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

109

further structural studies. The yield was limited by the insolubility of the caspase-4

p10 subunit. This is common among the inflammatory caspases including caspase-1

and caspase-5 (Roschitzki-Voser et al., 2012), which share high homology with

caspase-4. Using the crystal structure of caspase-1 p10 as a model it is possible to

identify putative caspase-4 p10 subunit hydrophobic residues exposed on the

surface. Through a series of site directed mutagenesis substitutions, to reduce

surface entropy and improve solubility, it may be possible to provide more favourable

conditions for initial crystallisation (Dale et al., 1994). This does have its

disadvantages, especially the fact that the primary sequence of caspase-4 may need

to be considerably altered before the p10 subunit is soluble enough to pursue

structural studies. Mutation of certain residues required for solubilisation may also

interfere with caspase-4/NleFE69 complex formation. An alternative method, albeit

more costly, is to purify active caspase-4 by LEVD affinity chromatography. Cell lysis

of BL21 expressing full length caspase-4 stimulates caspase-4 self-proteolytic

cleavage and oligomerisation. The active complex could be purified from the

bacterial cell lysate through affinity chromatography using the known caspase-4

substrate LEVD.

Although the data above convincingly identifies caspase-4 as a binding partner of

NleFE69 further studies are on-going to confirm caspase-4 and NleFE69 interaction.

This includes co-immunoprecipitation in Caco-2 cells lines transduced with pMXs-IP-

nleFE69 or during EPEC E69 infection. To study the biological relevance of this

interaction the cellular function of caspase-4 and the impact of NleF-caspase-4

interaction during infection was investigated.

Page 110: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

110

Results: Chapter 4 – Caspase-4, the functional ortholog of murine

caspase-11 is target by EPEC NleF

Page 111: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

111

4.1 Caspase-4 is the functional ortholog of murine caspase-11

Caspase-4 and caspase-11 have recently been characterised as intracellular innate

immune receptors that detect the Lipid A motif of LPS in vitro, resulting in their

activation (Shi et al., 2014). LPS transfection or A/E pathogen bacterial infection of

iMM triggers caspase-11 activation, resulting in formation of the non-canonical

inflammasome - leading to pyroptosis and IL-1 family cytokine maturation and

secretion (Kayagaki et al., 2011). However the role of human caspase-4 and

caspase-5 is less clear.

4.1.1 Caspase-4 can functionally complement caspase-11 to activate the

non-canonical inflammasome in response to EPEC infection

To determine if caspase-4 or caspase-5 can activate the non-canonical

inflammasome in response to EPEC E69 infection Cas11-/- iMM were

complemented with either CASP4 or CASP5 by retroviral transduction. Cas11-/- iMM

were also complemented with Casp11 or yfp as controls. Infection of iMM was

carried out using the T3SS mutant (ΔescN) of EPEC E69 as a model of EPEC E69

non-canonical inflammasome activation. This is due to the fact that WT EPEC E69

infection of iMM is known to trigger both the non-canonical inflammasome and the

NLRC4-NAIP2 inflammasome (Zhao et al., 2011). This is dependent on the

expression of the EPEC E69 inner rod protein EscI. Therefore, to test whether

caspase-4 or -5 can activate the non-canonical inflammasome in response to EPEC

E69 in iMM, a T3SS mutant must be used to prevent NLRC4 activation. Initially, to

verify that the ΔescN strain does not activate the NLRC4-dependent inflammasome,

WT and Nlrc4-/- iMM were infected with WT EPEC E69 or the ΔescN strain.

Supernatants post-infection were analysed by ELISA for the secretion of the

inflammasome dependent cytokine IL-1β. WT EPEC E69 infection of iMM triggered a

Page 112: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

112

robust NLRC4-dependent secretion of IL-1β (Fig 4.1A-B). Alternatively infection with

the ΔescN strain activated an NLRC4-independent inflammasome leading to

secretion of IL-1β (Fig. 4.1A-B)

Infection with the ΔescN strain was repeated using the CASP4, CASP5 and CASP11

transduced Casp11-/- iMM cell lines (Fig. 3.10A). A laboratory strain of E. coli

(STBL2) and LPS alone were used as controls. Secretion of IL-1β and IL-1α in

response to EPEC ΔescN infection was, as reported, dependent on caspase-11

expression and abolished in Casp11-/- iMM (Fig. 4.2B-C). Extracellular LPS alone did

not activate the non-canonical inflammasome while STBL2 activated a robust non-

canonical inflammasome response and IL-1β/IL-1α secretion in caspase-11

complemented Casp11-/- iMM (Fig. 4.2B-C). Caspase-4 fully complemented Casp11-

/- iMM to activate the secretion of both IL-1β and IL-1α in response to EPEC ΔescN

and STBL2 (Fig. 4.2B-C). On the other hand caspase-5 complemented Casp11-/-

iMM were unable to function to activate the murine non-canonical inflammasome in

response to EPEC ΔescN or STBL2 infection (Fig. 4.2B-C). This suggested that

caspase-4 but not caspase-5 is able to function as an ortholog of the murine non-

Figure 4 1 EPEC activates the NLRC4 inflammasome in a T3SS dependent manner. WT and Nlrc4-/- iMM were infected at an MOI of 25 with DMEM sub-cultured WT EPEC E69 or the ΔescN strain for 18 h. (A) Supernatants post infection were analysed by ELISA. Data analysed by Mann-Whitney test. *, P<0.05 (B) Enumeration of CFUs for each bacterial culture. CFU; colony forming units, iMM; immortalised mouse macrophages. Results are an average of two biological repeats carried out in triplicate. UI, uninfected.

Page 113: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

113

canonical inflammasome activator caspase-11. Furthermore it indicated that EPEC

E69 can trigger the non-canonical inflammasome in a T3SS independent-manner.

Figure 4 2 Caspase-4 can functionally complement caspase-11 to trigger the murine non-canonical inflammasome. (A) Casp11-/- iMM transduction with pMXs-IP-Casp11, pMXs-IP-CASP4 and pMXs-IP-CASP5, or pMXs-IP-yfp as a negative control, was confirmed by Western blot. (B-C) Transduced iMM were infected with STBL2 or EPEC ΔescN as described in material methods. Cells were left untreated or treated with 200 ng/ml LPS as controls. Supernatants post infection were analysed for IL-1β and IL-1α secretion by ELISA. Representative of two biological repeats carried out in duplicate. UT; un-transduced. *, P<0.05 for indicated columns vs LPS treated. Data was analysed by Two way ANOVA and bonferroni compare all columns.

Page 114: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

114

4.1 Generation of an nleF deletion EPEC strain

To investigate if NleFE69 functions to inhibit caspase-4 in infection; initially, an nleF

deletion mutant was generated. To generate an nleF KO strain a kanamycin cassette

flanked with the genomic DNA 5’ and 3’ to nleF of EPEC E69 was generated in the

plasmid TOPO-II-Blunt. This was constructed by PCR amplification of the genomic

DNA of EPEC E69 with primers to amplify nleF along with 500 bp 5’ and 400 bp 3’

(Fig. 4.3A). The nleF gene was removed by inverse PCR and replaced with a

kanamycin cassette. This newly generated construct was amplified by PCR and

transformed into WT EPEC E69 harbouring the pKD46 temperature sensitive

plasmid encoding the lambda red recombinase. Successful recombination events

were selected for by plating the transformation mix on LB agar with the addition of

kanamycin. Positive clones were confirmed by colony PCR. WT EPEC E69 genomic

DNA or the TOPO-II plasmids generated for EPEC E69 and EHEC nleF

mutagenesis were used as controls. Positive clones were further validated by

sequencing (Fig. 4.3A). Initially the fitness of the deletion strain was tested. Deletion

of nleF had no effect on bacterial growth kinetics in the minimal medium DMEM in

comparison to WT EPEC E69 (Fig. 4.3B). Furthermore complementation of the

ΔnleF strain with either the constitutive expression plasmid pACYC184-nleFE69 or the

arabinose inducible plasmid pBAD22-nleFE69 had no effect on growth kinetics (Fig.

4.3B).

Page 115: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

115

Figure 4 3 Deletion of nleF does not affect EPEC E69 growth kinetics. EPEC E69 pKD46 transformed with pTOPO-II-blunt-Kn induced with 0.5 mM IPTG were plated on LB-Kn to select for positive clones. (A) Positive clones 1-3 were screened by colony PCR using primers. The plasmid pTOPO-II-blunt-Kn for EPEC and EHEC ΔnleF and EPEC E69 genomic DNA (WT) were used as controls. A schematic for the amplicons of positive and negative clones is included. (B) EPEC E69 WT, the ΔnleF strain (clone 1) and the ΔnleF strain complemented with either pACYC-nleF or BAD22-nleF were sub-cultured from LB into DMEM and growth kinetics were measured by CFU every 45 min over 3 h. Average of two biological repeats carried out in triplicate.

Page 116: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

116

4.2 NleFE69 inhibits activation and secretion of caspase-4 during infection of

polarised intestinal epithelial cells

To determine if NleFE69 targets caspase-4 during infection Caco-2 TC7 were infected

with the WT EPEC E69, the EPEC nleF deletion mutant (ΔnleF) or the

complemented ΔnleF pnleFE69 strain and supernatants were analysed for secreted

caspase-4 and IL-18. Importantly, deletion of nleFE69 alone significantly increased

the secretion of active caspase-4 during infection of IECs in comparison to the WT

EPEC E69 strain (Fig. 4.4A). Furthermore, complementation of the ΔnleF strain with

pnleFE69 or the addition of the caspase-4 inhibitor Ac-LEVD-CHO successfully

prevented caspase-4 activation and secretion during EPEC E69 infection (Fig. 4.4A-

D). Caspase-4 can functionally complement the role of caspase-11 in Casp11-/- iMM,

to activate the non-canonical inflammasome in response to EPEC E69 infection and

emerging data fundamentally supports a role for caspase-4 in the activation of the

human non-canonical inflammasome (Knodler et al., 2014; Shi et al., 2014).

However, little is known about the human non-canonical inflammasome, especially in

non-hematopoietic cells. Interestingly, no IL-18 cleavage was detected in response

to WT EPEC E69 infection of Caco-2 TC7 (Fig. 4.4). Importantly, infection with the

EPEC ΔnleF strain stimulated the secretion of the active p18 peptide of IL-18,

correlating with caspase-4 activation during infection (Fig. 4.4). Expression levels of

IL-18 were unaffected by infection and all EPEC strains adhered to the Caco-2 TC7

cells at similar levels (Fig. 4.4B-C); which suggested that NleFE69 is not acting at the

transcriptional level.

Page 117: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

117

The loss of inhibition of inflammasome activity through deletion of nleFE69 was

complemented by constitutive expression of NleFE69 alone, inhibiting both caspase-4

and IL-18 processing and secretion (Fig. 4.4D). This suggested that EPEC E69

Figure 4 4 NleFE69 inhibits secretion of active caspase-4 and IL-18 duringinfection of IECs. (A) Caco-2 TC7 were infected with the WT EPEC E69, ΔnleF orΔnleF pnleF strains for 19 h and supernatants and lysates were analysed for IL-18and caspase-4 by Western blot as described in materials and methods. (B) Lysateswere also analysed 4 h post-infection as control for IL-18 expression. (C) Three hourspost-infection cell layers were lysed and plated to determine bacterial CFU to enumerate EPEC E69 attachment for each strain. (D) Infection was repeated with or without the addition of 20 µM or 100 µM Ac-LEVD-CHO. Results are representative of at least two biological repeats. UI, uninfected; SN, supernatant.

Page 118: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

118

secretes the effector NleFE69 during infection to target and inhibit caspase-4

activation and the secretion of biologically active IL-18.

Activation of caspase-4 and the non-canonical inflammasome in response to

intracellular LPS is reported to control pyroptotic cell death (Knodler et al., 2014).

While it was observed that WT EPEC E69 infection inhibited caspase-4 activation in

an NleFE69 dependent manner, deletion of nleF did not increase cell cytotoxicity

during infection (Fig. 4.5A).

As with NleFE69, addition of Ac-LEVD-CHO did not significantly reduce cell death in

Figure 4 5 Deletion of nleF alone does not increase cell death during infection of IECs. Caco-2 TC7 were infected with the WT EPEC E69, ΔnleF or ΔnleF pnleF strains for 19 h with or without the addition of 100 µM Ac-LEVD-CHO, as described in materials and methods. (A) Cell cytotoxicity was analysed by LDH release and displayed as a percentage of total LDH. Representative of at least two biological repeats carried out in triplicate. UI, uninfected; LEVD, Ac-LEVD-CHO.

Page 119: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

119

comparison to infection with the WT EPEC E69 or ΔnleF strain (Fig. 4.5A). This

indicated that while NleFE69 and inhibition of caspase-4 activation may control IL-18

secretion, cell death during EPEC E69 infection is caspase-4/NleFE69 independent.

4.3 Activation of caspase-4 in response to EPEC E69 infection triggers IL-18

processing and secretion

To validate that the processing and secretion of IL-18 observed during infection with

the ΔnleF strain is caspase-4-dependent Caco-2 TC7 were transduced with CASP4

specific short-hairpin RNA (shRNA). Caco-2 TC7 were transduced with either YFP-

mIR30E shRNA targeting CASP4 or lacZ as a non-targeting (NT) negative control.

CASP4 specific shRNA-mediated knockdown successfully and specifically abolished

expression of caspase-4 in comparison to WT Caco-2 caspase-4 levels (Fig. 4.6A).

Moreover, no effect on caspase-4 protein levels were observed with the NT control

transduced cell line. Similarly no knockdown of the highly homologous protein

caspase-5 was observed in any condition, showing that the shRNA has high

specificity for caspase-4 (Fig. 4.6A). Following confirmation of successful knockdown

the transduced Caco-2 TC7 cell lines were infected with the WT EPEC E69 or ΔnleF

strains. Supernatants were analysed 19 h post-infection for the secretion of active IL-

18. As described previously, infection of the NT control cell line with the EPEC ΔnleF

mutant significantly increased the processing and secretion of IL-18 in comparison to

infection with the WT EPEC E69 strain (Fig. 4.6B). However, no secreted processed

IL-18 was detectable in response to infection of the caspase-4 knockdown cell line

with either strain (Fig. 4.6B).

Page 120: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

120

This suggested that caspase-4 is essential for IL-18 processing and secretion during

infection. Following knockdown, to further investigate the role of caspase-4 in cell

cytotoxicity, both LDH release and PI uptake were analysed post-infection. While

knockdown of caspase-4 successfully inhibited IL-18 processing no effect on cell

cytotoxicity during infection with the WT EPEC E69 or ΔnleF strain was observed

Figure 4 6 Caspase-4 is essential for IL-18 processing and secretion during EPEC E69 infection of IECs. Caco-2 TC7 were transduced with the shRNA retroviral plasmid pMXs-IP-yfp-mIR30E CASP4 (C4) and pMXs-IP-yfp-mIR30E lacZ (NT) as a non-targeting negative control. (A) Caspase-4 knockdown, shRNA efficiency and specificity was confirmed by Western blot. (B) Transduced Caco-2 TC7 were infected with the WT EPEC E69 and ΔnleF strains. At 19 h post-infection supernatants were analysed for IL-18 secretion and cleavage by Western blot. Representative of two biological repeats (C-D) Cell cytotoxicity was analysed by LDH release and PI uptake. Results are displayed as a percentage of total LDH or total PI uptake, respectively. Results are an average of two biological repeats carried out in quadruplicate.

Page 121: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

121

(Fig. 4.6C-D). This confirmed, unlike IL-18 secretion, cell death induced by EPEC

E69 infection is caspase-4-independent. This indicated that cell death and IL-18

cytokine secretion during EPEC E69 IEC infection are mutually exclusive.

Recently it was reported that caspase-1 is dispensable for the Caco-2 TC7 caspase-

4-dependent inflammasome (Knodler et al., 2014). This would suggest that the IEC

non-canonical inflammasome significantly differs to that of the macrophage non-

canonical inflammasome. Within in this study, using LPS primed THP-1 lysate as a

control, no expression of full length caspase-1 (45 kDa) could be detected in the

lysates of the Caco-2 clones TC7 or BBE1 (Fig. 4.7A). Alternatively two bands at ~23

kDa were detected using a p20 anti-caspase-1 antibody (Fig. 4.7A). The lower band

was also present as a cleavage product in the THP-1 lysate (Fig. 4.7A).

Figure 4 7 Full length caspase-1 is not detectable in the lysates of the Caco-2clones TC7 or BBE1. Caco-2 TC7 and BBE1 were polarised and differentiated as described in material and methods and lysates were analysed by Western blot for caspase-1 and IL-18 expression. As a control THP-1 cells were differentiated by PMA and either left unstimulated or stimulated with 200 ng/ml LPS for 3 h

Page 122: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

122

A p20 caspase-1 band is associated with activation of caspase-1 and the

inflammasome. While this band may represent active caspase-1 no cleavage of IL-

18 was detectable by Western blot (Fig. 4.7). Furthermore we have shown above

that under normal conditions in uninfected IECs IL-18 is neither secreted nor

processed. Whether full length caspase-1 is expressed by Caco-2 or not, and

whether its activity is required for IL-18 secretion requires further investigation.

The results above identify that NleFE69, upon translocation, targets and inhibits

caspase-4 activity. As predicted, caspase-4 functions similarly to the murine

caspase-11 to trigger IL-18 cytokine processing and secretion in response to Gram

negative bacterial infection. However, we report that cell death during EPEC E69

infection of IECs is caspase-4 and therefore NleFE69 independent. Here the caspase-

4 inflammasome significantly differs from that of the characterised murine non-

canonical inflammasome of macrophages - where cell death and IL-18 secretion are

directly linked.

4.4 Discussion

Little is known about the role of caspase-4 in human epithelial cells. Emerging

evidence proposes a role for caspase-4 upstream to IL-18 processing and secretion

in response to Gram negative bacterial infection (Knodler et al., 2014). Both

caspase-4 and caspase-5 share significant sequence homology to the murine

caspase-11 and are both implicated as the human functional ortholog. Caspase-11

functions upstream to caspase-1 and inflammasome activation in response to

intracellular LPS (Shi et al., 2014; Kayagaki et al., 2011). To verify that caspase-4

and not caspase-5 is the functional ortholog of caspase-11 Casp11-/- iMM were

transduced to express caspase-4 or caspase-5. Here we report that caspase-4

Page 123: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

123

functionally complemented caspase-11 in response to Gram negative bacterial

infection to activate the non-canonical inflammasome and cytokine secretion.

Although cytokine maturation and secretion had not been studied using this model

previously, it has been reported that caspase-5 can regulate non-canonical

inflammasome-dependent pyroptotic cell death in response to intracellular LPS. It is

therefore proposed that caspase-5 may play a role in non-canonical inflammasome

cytokine maturation (Shi et al., 2014). We did not test the capability of either caspase

to induce pyroptosis, however the failure of caspase-5 to complement Casp11-/- iMM

may be due to the caspase-5 isoform we used. We were unable to detect/amplify the

caspase-5a/f isoforms used by Shi and colleagues from HeLa or THP-1 cDNA pools

and instead amplified the caspase-5 isoform first described by Munday and

colleagues (Munday et al., 1995). In support of our findings a very recent publication

sheds light on the role of caspase-4 and caspase-5 in the human macrophage non-

canonical inflammasome. Casson and colleagues observed that caspase-4 plays the

conserved role of caspase-11 to activate the non-canonical inflammasome in human

primary macrophages, in response to Gram negative bacterial infection (Casson et

al., 2015). Specifically, caspase-4 was shown to regulate IL-1α secretion

independent to caspase-1 during both Legionella pneumophila and S. typhimurium

infection. Caspase-4 knockdown had no effect on IL-1β secretion during infection;

although, both of these pathogens simultaneously activate the NLRC4-dependent

inflammasome. In this case caspase-4 activity is most likely redundant. Caspase-5

on the other hand was shown to be dispensable for secretion of both IL-1α and IL-

1β. In response to sterile activation through LPS transfection it was further shown by

Casson and colleagues that caspase-4 is essential for both IL-1β and IL-1α secretion

(Casson et al., 2015). Having confirmed a putative function for caspase-4, which has

Page 124: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

124

now been further confirmed in a human macrophage model of the non-canonical

inflammasome, the role of NleFE69 during infection was explored.

The activation of caspase-4 during EPEC E69 infection has not been previously

studied. In response to WT EPEC E69 infection it was observed that caspase-4

undergoes processing and is secreted. Activation during infection was significantly

increased upon infection with the EPEC E69 ΔnleF mutant. This identified a novel

function for the previously uncharacterised NleFE69 effector as an inhibitor of

caspase-4. To determine if caspase-4 functions to activate the non-canonical

inflammasome in IECS the secretion of IL-18 upon infection was analysed. IL-18

processing and secretion was only detectable during infection upon deletion of nleF.

This was further validated to be caspase-4 dependent by knockdown. This

established that caspase-4 activation and the IEC non-canonical inflammasome

response is dampened during infection in a T3SS and an NleFE69 dependent

manner.

While this identifies that caspase-4 is essential for IL-18 secretion and processing in

response to EPEC E69 infection, Knodler and colleagues reported that caspase-1 is

dispensable for the IEC caspase-4 dependent inflammasome. Caspase-4 can

directly cleave IL-1 family cytokines, albeit with a lower efficiency than caspase-1;

therefore, caspase-1 may not be essential (Sollberger et al., 2012). In our study we

did not observe any full length caspase-1 in either Caco-2 clones BBE1 or TC7 nor

any NLRP3 by Western blot (data not shown). Without NLRP3 or caspase-1 a

caspase-4 dependent pathway would supersede. Whether caspase-1 is expressed

in IECs in vivo requires further investigation.

Page 125: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

125

Pyroptosis is a form of inflammatory programmed cell death that differs significantly

from that of apoptosis. Caspase-1 is the master controller of pyroptosis, inducing

plasma membrane pore formation, osmotic lysis and the release of pro-inflammatory

cellular cytoplasmic contents (Bergsbaken, Fink & Cookson, 2009). Caspase-11 is

essential for pyroptosis triggered by Gram negative bacterial infection of murine

macrophages including C. rodentium and E. coli (Kayagaki et al., 2011). We did not

observe any correlation between caspase-4 activity and cell death during EPEC E69

infection of IECs. Neither caspase-4 knockdown nor LEVD treatment affected the

level of LDH release or PI uptake. Although caspase-11 has been shown to be

essential for pyroptosis this may be due to downstream caspase-1 effects. This

would explain why in IECs where little or no caspase-1 is expressed, that caspase-4

does not control pyroptosis. This may also implicate caspase-4 in a novel or

unconventional non-canonical inflammasome that does not induce pyroptosis.

Cell death during infection with either WT EPEC E69 or the ΔnleF strain, whilst

significant, was not considerably high in comparison to the uninfected control. This

supports previous findings that EPEC E69 successfully inhibits cell death in epithelial

cells as a mechanism to prevent cell turnover; to aid in bacterial colonisation of the

host (Wong Fok Lung et al., 2014; Hemrajani et al., 2010). It may be suggested that

in order to see a difference between the WT EPEC E69 and ΔnleF strain a higher

MOI may be required. To test this we doubled or halved the MOI and repeated the

infections as carried out previously. Just by doubling the MOI cell death was at 100%

after 3 h of infection (Data not shown). This indicated that there is a fine line between

total inhibition and complete cell death during EPEC E69 infection of epithelial cells.

We conclude that NleFE69 inhibits caspase-4 activity and IL-18 processing and

secretion during epithelial cell infection; however, as observed previously (Blasche et

Page 126: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

126

al., 2013; Black et al., 2010), NleFE69 does not play a role in programmed cell death.

Many characterised EPEC E69 effectors have been identified to target and interact

with multiple partners. Our collaborators, through the use of an NF-κB luciferase

reporter assay in HeLa cells identified putative T3SS effectors, that when ectopically

expressed, activated NF-κB. One of these putative pro-inflammatory effectors was

NleFE69.

Page 127: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

127

Results: Chapter 5 – NleFE69 activates NF-κB signalling

during EPEC infection

Page 128: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

128

1.10 NleFE69 localises to the mitochondria and activates NF-κB nuclear

translocation

To explore whether NleFE69 activates the nuclear translocation of the inflammatory

transcription factor NF-κB, HeLa cells were transfected with the mammalian

expression plasmids pRK5-myc-nleFE69 (HeLa-NleF), pRK5-myc-nleF1-185 E69 (HeLa-

NleF1-185) or pRK5-myc-gfp (HeLa-GFP) as a negative control, and analysed by

immunofluorescence staining.

1.10.1 NleFE69 localises to the mitochondria dependent on its C-terminal

motif LQCG

At 24 h post-transfection it was observed that staining of NleFE69 was either diffusely

localised throughout the cytoplasm and nucleus of the cell, or in a punctate peri-

nuclear localisation (Fig. 5.1A). Localisation post 24 h was not studied due to the fact

that prolonged expression of NleFE69 was cytotoxic (data not shown). Co-staining of

the major organelles of the cell and Myc-NleFE69 was carried out to determine the

identity of the punctate staining observed during ectopic expression of NleFE69. No

co-localisation was observed for the Golgi apparatus (GM130) or endoplasmic

reticulum ER (calnexin) in HeLa-NleF (Fig. 5.1A); however, the punctate peri-nuclear

staining of NleFE69 co-localised with the antibody staining for TOMM22. TOMM22 is

a mitochondrial outer membrane protein, suggesting that NleFE69 localises to the

mitochondria when ectopically expressed (Fig. 5.1A). No co-localisation with

TOMM22 or punctate staining was observed for HeLa-GFP (Fig. 5.1A). Enumeration

of HeLa-NleF exhibiting mitochondrial staining or diffuse staining showed that neither

localisation phenotype was dominant (Fig. 5.1C). Previously it has been reported

that the C-terminal motif of NleFE69, LQCG, is essential for its function to bind and

inhibit caspases (Blasche et al., 2013). Localisation of NleFE69 to the mitochondria

Page 129: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

129

may be dependent on interaction with host target proteins; therefore, the localisation

of NleFE69 was compared between HeLa-NleF and HeLa-NleF1-185. Interestingly

deletion of the 4 C-terminal amino acids of NleFE69 abolished localisation to the

mitochondria, resulting in a diffuse staining in all transfected cells (Fig. 5.1B) This

suggested that the mitochondrial localisation of NleFE69 may be caspase dependent.

Page 130: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

130

Page 131: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

131

1.10.2 Ectopically expressed NleFE69 activates nuclear translocation of NF-

κB

To determine if ectopic expression of NleFE69 is sufficient to induce nuclear

translocation of NF-κB the subcellular localisation of the NF-κB subunit p65 was

determined in HeLa-NleF and HeLa-GFP, by immunofluorescence. NF-κB, under

resting conditions, is sequestered in the cytoplasm in complex with IκBα. Upon

activation and degradation of IκBα p65 is released and translocates into the nucleus

where it transcriptionally up-regulates pro-survival and pro-inflammatory genes under

Figure 5 1 Ectopically expressed NleFE69 localises to the mitochondria dependent on its C-terminal motif LQCG. Representative immunofluorescence images of NleFE69 subcellular localisation in HeLa 24 h post-transfection with pRK5-myc-nleF, pRK5-myc-nleF1-185 E69 or pRK5-myc-gfp as a control. (A-B) Coverslips were co-stained for the major organelles including the ER (anti-calnexin), the golgi (anti-GM130) and the mitochondria (anti-TOMM22). The nucleus and Myc were stained using DAPI and anti-Myc, respectively. Bars, 10 µM. Representative of at least two biological repeats carried out in triplicate (C) Cells exhibiting mitochondrial localised or diffuse staining for NleFE69 were enumerated. 100 cells per condition were counted. Average of three biological repeats carried out in triplicate.

Page 132: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

132

its control (Li & Verma, 2002). Cells treated with TNFα, or cells transfected with the

TAB2/3 inhibitor NleE1 (HeLa-NleE) or HeLa GFP were used as controls. As

expected stimulation with TNFα significantly increased the number of cells exhibiting

nuclear localised p65 and following previous publications this could be inhibited by

expression of NleE1 (Newton et al., 2010) (Fig. 5.2A-B). Interestingly, expression of

NleFE69 alone (HeLa-NleF) stimulated the nuclear translocation of p65. The number

cells with nuclear p65 for HeLa-NleF were significantly higher than that for HeLa-

GFP or the un-transfected control (Fig. 5.2A-B). The localisation of NleFE69 and

ability to bind and inhibit the host targets caspase-4 and caspase-8 is dependent on

the 4 C-terminal amino acids of NleFE69 (Blasche et al., 2013). To test whether the

ability of NleFE69 to activate NF-κB p65 translocation is C-terminal dependent the

number of cells positive for nuclear p65 were compared between HeLa-GFP, HeLa-

NleF and HeLa-NleF1-185. Deletion of the 4 C-terminal amino acids for NleFE69

significantly abrogated its functional activity to induce nuclear p65 translocation (Fig.

5.2A-B). Importantly this suggested that NleFE69 can activate the nuclear

translocation of the transcription factor NF-κB p65 and may act via interaction with

either caspase-4 or caspase-8. While the ability of NleFE69 to induce nuclear

translocation of p65 was dependent on its C-terminal domain (Fig. 5.2A-B) there was

no observed difference in p65 nuclear translocation between HeLa-NleF exhibiting

either diffuse staining or mitochondrial staining for NleFE69 (Fig. 5.3A-B).

Page 133: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

133

Figure 5 2 Ectopically expressed NleF activates nuclear translocation of p65. (A)Representative immunofluorescence images of p65 localisation using anti-N-terminalp65 in HeLa cells 24 h post transfection with pRK5-myc-nleF, pRK5-myc-gfp, pRK5-myc-nleE1 or pRK5-myc-nleF1-185 E69. The nucleus and Myc were stained using DAPI and anti-Myc, respectively. Cells were stimulated with 20 ng.ml TNFα for 0.5 h as a positive control. Bars, 10 µM (B) Cells exhibiting nuclear p65 staining were enumerated. 100 cells per condition were counted. Average of three biological repeats carried out in triplicate. *, P<0.05. Arrows indicate nuclear p65.

Page 134: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

134

Figure 5 3 NleFE69 activity is independent of subcellular localization. Representative immunofluorescence images of NleF/GFP localisation in HeLa cells 24 h post transfection with pRK5-myc-nleF or pRK5-myc-gfp using anti-Myc. Bars, 10 µM. The nucleus and NF-κB p65 were stained using DAPI andante-N-terminal p65, respectively. (B) Cells with mitochondrial or diffuse NleFE69 staining exhibiting nuclear p65 were enumerated. 100 cells per condition were counted. Average of three biological repeats carried out in triplicate.

Page 135: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

135

1.11 NleFE69 up-regulates the expression of the chemokine IL-8

As a transcription factor the translocation of p65 to the nucleus results in the up-

regulation of a subset of genes under the control of the NF-κB promoter. To

investigate if NleFE69 induces the up-regulation of NF-κB dependent genes

expression of IL-8 mRNA of HeLa-GFP, HeLa-NleF and HeLa-NleF1-185 was

analysed 24 h post transfection by quantitative reverse-transcription PCR (qRT-

PCR). The house keeping gene GAPDH was used as a loading control while TNFα

stimulated cells were used as a positive control for up-regulation of IL-8 expression.

IL-8 expression levels were significantly increased in cells treated with TNFα (11-

fold) and in HeLa-NleF (5.8-fold) in comparison to HeLa-GFP (Fig. 5.4A). As with the

ability of NleFE69 to activate the nuclear translocation of NF-κB p65, the up-regulation

of the expression of IL-8 was dependent on the C-terminal domain of NleFE69 (Fig.

5.4A). Up-regulation of IL-8 expression was not observed for HeLa-NleF1-185 in

comparison to HeLa-GFP.

Figure 5 4 Ectopic expression of NleFE69 induces up-regulation the NF-κB promoter dependent chemokine IL-8. (A) IL-8 mRNA levels 24 h post-transfection for HeLa transfected with pRK5-myc-nleFE69, pRK5-myc-nleF1-185 E69 or pRK5-myc-gfp were analysed by qRT-PCR. TNFα at 20 ng/ml was added for 16 h as a positive control. *, P<0.05 for GFP vs Un + TNFα and GFP vs NleFE69, Mann Whitney test. Results are an average of three biological repeats. (B) Protein levels of Myc-GFP, Myc-NleFE69 and Myc-NleF1-185 relative to tubulin were analysed in whole cell extracts by Western blot using anti-Myc and αnti-tubulin, respectively. Results are representative of two biological repeats. Un; un-transfected.

Page 136: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

136

Transfected cell lysates were analysed by immunoblotting to determine that

expression levels of NleFE69 and NleF1-185 E69 were similar and stable during

transfection. NleFE69 and NleF1-185 E69 were expressed at similar levels during

transfection and no difference in transfection efficiency was observed (Fig. 5.4B;

data not shown). This determined that ectopically expressed NleFE69 induces the

nuclear translocation of p65 and the up-regulation of the pro-inflammatory NF-κB

dependent chemokine IL-8. While the mechanism of activation is unknown the fact

that the activity of NleFE69 is dependent on its C-terminal domain suggests it may

function via the inflammatory caspase-4 or caspase-8.

1.11.1 NleFE69 induced NF-κB activation is independent of interaction with

caspase-4, -8 or -9.

Both caspase-4 and caspase-8, in response to multiple stimuli function to activate

NF-κB via a number of intracellular signalling cascades (Nickles et al., 2012;

Lakshmanan & Porter, 2007; Chaudhary et al., 2000). To explore if the mechanism

of NleFE69-dependent activation of NF-κB was through interaction with either

caspase-4 or caspase-8 HeLa-GFP and HeLa-NleF were transfected with caspase

specific short inhibitory RNA (siRNA). Non-targeting siRNA was transfected as a

negative control and knockdown of mRNA in HeLa of the known interacting partner’s

caspase-4, -8 and -9 was confirmed by qRT-PCR and Western blot, where stated.

Transfection of siRNA for caspase-4, -8 and -9 successfully depleted cellular levels

of each caspase to 11%, 18% and 10% of WT levels in comparison to the non-

targeting siRNA transfected control (Fig 5.5A-D). Thirty six hours post siRNA

transfection HeLa were transfected with pRK5-myc-gfp or pRK5-myc-nleFE69. As

before, mRNA was extracted and IL-8 expression levels were analysed by qRT-

PCR. Knock down of caspase-4, -8 or -9 had no observed effect on the NleFE69-

Page 137: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

137

dependent up-regulation of IL-8 expression in comparison to the non-targeting

siRNA transfected control or GFP (Fig. 5.5E).

Caspase-4 and caspase-8 function independently to activate NF-κB nuclear

translocation; therefore, NleFE69 may activate either caspase, or both, to stimulate

p65 translocation and up-regulation of IL-8 expression. To investigate this as a

Figure 5 5 NleFE69 functions independently of caspase-4, -8 or -9 to activate IL-8 expression. HeLa cells were transfected with siRNA for (A) CASP4, (B) CASP8 or (C) CASP9. mRNA levels in comparison to the non-targeting control were analysed 36 h post-transfection by qRT-PCR. Results are an average of three biological repeats. (D) Caspase-4 levels relative to tubulin in HeLa whole cell extracts of siRNA transfected cells were analysed by Western blot 36 h post-transfection. Results are an average of two biological repeats. (E) Thirty-six hours post-transfection with siRNA HeLa were transfected with pRK5-myc-nleFE69 or pRK5-myc-gfp and IL-8 mRNA analysed by RTq-PCR. Results are representative of those of two biological repeats carried out in triplicate

Page 138: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

138

possible mechanism HeLa were dual transfected with either both caspase-4 and -8

siRNA or the non-targeting contro siRNAl. Dual knockdown successfully reduced the

expression of both caspase-4 and -8 (Fig. 5.6A). HeLa transfected with both

caspase-4 and -8 siRNA or the non-targeting control were then transfected with

either pRK5-myc-nleFE69 or pRK5-myc-gfp. Dual knockdown of caspase-4 and -8

had no effect on the ability of NleFE69 to induce up-regulation of IL-8 expression as

determined by qRT-PCR (Fig. 5.6B). This determined that the mechanism of

activation is not through stimulation of either caspase-4 or caspase-8.

Figure 5 6 Knock-down of caspase-4 and -8 has no effect on the pro-inflammatory activity of NleFE69. (A-B) HeLa cells were co-transfected with siRNA for caspase-4 and -8 (20 nM) or scrambled control siRNA (40 nM) alone. mRNA levels were analysed 36 h post transfection by qRT-PCR. (C) Thirty six hours post-transfection with respective siRNA, HeLa cells were transfected with either pRK5-myc-gfp or pRK5-myc-nleFE69. IL-8 mRNA levels were analysed by qRT-PCR. Resultsare representative of one experiment carried out in triplicate.

Page 139: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

139

1.12 NleFE69 activates the nuclear translocation of NF-κB early during EPEC

E69 infection

EPEC E69 activates pro-inflammatory signalling, in a T3SS-dependent manner,

early during in vitro tissue culture epithelial cell (Newton et al., 2010; de Grado et al.,

2001; Savkovic et al., 2001; Savkovic, Koutsouris & Hecht, 1997). Interestingly,

Savkovic and colleagues observed that early during infection EPEC E69 actives NF-

κB activation. Moreover, this activation is dependent on a functional T3SS (Savkovic

et al., 2001). Newton and colleagues further observed that in the absence of the

pathogenicity islands PP4 and IE6, which encode the characterised NF-κB inhibitory

effectors including NleC, NleB1 and NleE1, EPEC E69 activates NF-κB p65

translocation and IL-8 secretion in a T3SS dependent manner . To determine if

NleFE69 functions during infection to activate NF-κB p65 nuclear translocation HeLa

cells were infected for 1.5 h with WT EPEC E69, ΔescN, ΔnleF or the complemented

ΔnleF pnleFE69 strain and analysed by immunofluorescence staining (Fig. 5.7A). No

difference in adherence or pedestal formation (excluding the ΔescN strain) was

observed between the different strains (Fig. 5.7A). As previously reported, WT EPEC

E69 (Savkovic, Koutsouris & Hecht, 1997) robustly activated NF-κB p65 nuclear

translocation 1.5 h post-infection (Fig. 5.7A-B); this was dependent on a functional

T3SS as the ΔescN strain was unable to promote p65 translocation (Fig. 5.7A-B).

This re-confirmed that at 1.5 h post-infection NF-κB p65 nuclear translocation is

T3SS (Newton et al., 2010) dependent. Deletion of nleF abolished NF-κB p65

nuclear translocation during EPEC E69 infection to that of the level seen for the

ΔescN strain (Fig. 5.7A-B). This was partially restored through complementation of

the ΔnleF strain (Fig. 5.7A-B). This suggested that NleFE69 is the T3SS effector

responsible for activation of p65 early during infection.

Page 140: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

140

Figure 5 7 NleFE69 activates p65 nuclear translocation early during infection. (A) Representative immunofluorescence images of NF-κB p65 localisation using anti-N-terminal p65, 1.5 h post-infection with WT EPEC E69, ΔescN, ΔnleF or the complemented ΔnleF pnleFE69 strain. Actin and the nucleus were stained using phalloidin-Cy5 and DAPI, respectively. EPEC were visualised using anti-eae. Bars, 10 µM. (B) The number of cells exhibiting nuclear p65 were counted for 8 separate fields of view per condition. Average of three biological repeats carried out in triplicate. (C) IL-8 mRNA levels 1.5 h post-infection were analysed by qRT-PCR. *, P<0.05 for UI vs WT and UI vs ΔnleF pnleFE69. UI; uninfected.

Page 141: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

141

To determine if NleFE69 dependent activation of NF-κB p65 nuclear translocation

during infection leads to up-regulation of genes under the control of the NF-κB

promoter the infection was repeated and mRNA was extracted and analysed by

qRT-PCR. As with NF-κB p65 nuclear translocation infection with the WT EPEC E69

strain induced a 28.5 fold increase in IL-8 mRNA levels in comparison to the

uninfected (Fig. 5.8C). Alternatively infection with the ΔescN strain only induced a

2.3 fold increase in comparison (Fig. 5.8C). Interestingly, deletion of nleF alone

reduced the level of IL-8 expression to that seen for the ΔescN strain; again this was

partially restored upon complementation with pnleFE69 (Fig. 5.8C). This suggested

that NleFE69 stimulates the expression of IL-8 early during EPEC E69 infection.

At later time points post-infection EPEC E69 delivers a plethora of T3SS effectors to

inhibit NF-κB signalling (Gao et al., 2013; Raymond et al., 2013; Baruch et al., 2011;

Pearson et al., 2011; Raymond et al., 2011; Nadler et al., 2010; Newton et al., 2010;

Vossenkämper et al., 2010; Gao et al., 2009). This includes the T3SS effector and

TAB2/3 inhibitor NleE1 (Newton et al., 2010). To determine if NleFE69 functions to

activate NF-κB at later time points post-infection HeLa cells were infected for 4 h with

the WT EPEC E69, ΔescN or the ΔnleF strain and p65 localisation was analysed by

immunofluorescence staining. As a control for activation HeLa cells were also

infected with the ΔPP4/IE6 strain and the ΔPP4/IE6 strain complemented with

pnleE1. At 3.5 h post-infection, as a control for inhibition of NF-κB p65 nuclear

translocation, WT EPEC E69 infected cells or uninfected cells were treated with 20

ng/ml TNFα. No difference in NF-κB sub-cellular localisation between the

uninfected/unstimulated or infection with the WT EPEC E69, ΔescN or the ΔnleF

strain was observed at 4 h post-infection (Fig. 5.8A-B). Furthermore, stimulation of

WT EPEC E69 infected cells with TNFα did not stimulate p65 nuclear translocation;

Page 142: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

142

while stimulation of uninfected cells with TNFα resulted in p65 nuclear translocation

in almost 100% of cells (Fig. 5.8A-B). This suggested at 4 h post-infection EPEC E69

inhibits NF-κB activation in a T3SS-dependent manner.

Page 143: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

143

Alternatively, as described previously, infection with EPEC ΔPP4/IE6 resulted in

activation of p65 nuclear translocation in infected cells (Fig. 5.8A-B). This could be

complemented by NleE1 alone, suggesting T3SS-dependent NF-κB activation at 4 h

post-infection is TAB2/3 dependent (Fig. 5.8A-B). This determined that at 1.5 h post-

infection, in a T3SS-dependent and NleFE69-dependent manner, WT EPEC E69

activates NF-κB p65 nuclear translocation; while at later time points, through the

secretion of NleE1 alone, EPEC is capable of inhibiting NF-κB nuclear translocation

activated either in a T3SS-dependent manner or through exogenous stimuli.

Figure 5 8 EPEC inhibits exogenous stimulation and T3SS-dependent activation of NF-κB at 4 h post-infection. Representative immunofluorescence images of NF-κB p65 localisation using anti-N-terminal p65 4 h post-infection with WT EPEC E69, ΔescN, ΔnleF, ΔPP4/IE6 or the complemented ΔPP4/IE6 pnleE strain. EPEC and the nucleus were stained using anti-eae and DAPI, respectively. Uninfected and WT infected cells were treated with 20 ng/ml TNFα for 30 min where stated. Bars, 10 µM. (B) The number of cells exhibiting nuclear p65 were counted for 8 separate fields of view per condition. Average of three biological repeats carried out in triplicate. *, P<0.05 for UI vs UI + TNFα and UI vs ΔPP4/IE6. UI; uninfected

Page 144: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

144

1.13 NleFE69 dependent activation of NF-κB is up stream to IκBα

degradation

Proteasomal degradation of IκBα is a crucial step in canonical NF-κB signalling and

activation and is downstream to TAB2/3 recruitment (Li & Verma, 2002). HeLa cells

were infected with WT EPEC E69, ΔnleF or the complemented ΔnleF pnleFE69 strain

to determine if NleFE69 acts at the stage of IκBα degradation in the NF-κB signalling

cascade during infection. At 1.5 h post-infection the level of IκBα relative to tubulin in

whole cell lysates was analysed by Western blot. Infection with the WT EPEC E69

strain resulted in degradation of IκBα relative to the uninfected condition (Fig. 5.9A).

On the other hand there was no difference in the level of IκBα between the

uninfected and the ΔnleF infected condition (Fig. 5.9A). Infection with the ΔnleF

pnleFE69 strain partially rescued the ability of EPEC E69 to induce IκBα degradation

(Fig. 5.9A). This suggested that the NleFE69 dependent activation of NF-κB during

EPEC E69 infection acts through and upstream to the degradation of IκBα, although

the host binding partner remains unknown.

Figure 5 9 EPEC E69 induces IκBα degradation in an NleFE69 dependentmanner during infection. (A) HeLa cells were infected for 4 h with the WT EPEC E69, ΔnleF or ΔnleF pnleFE69 strain and IκBα (anti-IκBα) levels relative to tubulin (anti-tubulin) were analysed by Western blot. Densitometry was carried out using imageJ and is an average of two biological repeats.

Page 145: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

145

1.14 NleFE69 interacts with human DHFR

Although caspase-4, -8, and -9 knockdown did not inhibit the pro-inflammatory

activity of NleFE69, alternative putative binding partners have been identified through

Y2H screens carried out by other groups. Blasche and colleagues identified

dihydrofolate reductase (DHFR) as a possible interacting partner (Blasche, 2013).

DHFR is an essential enzyme required for the de novo synthesis of nucleic and

amino acids. Recently the previously annotated pseudogene DHFRP4 was reported

to encode the fully functional mitochondrial localised DHFR homolog DHFRL1

(McEntee et al., 2011). To investigate if NleFE69 interacts with either DHFR or

DHFRL1 AH109 were co-transformed with pGBT9-nleFE69/nleF1-185 E69 and pGADT7-

DHFR/DHFRL1 and plated on selective media. Co-transformation with pGBT9-

nleFE69 and pGADT7-CASP4 was used as a positive control. As reported by Blasche

and colleagues NleFE69 interacted with DHFR in the YDH system (Fig. 5.10A)

(Blasche, 2013). This interaction, unlike caspase binding, was not dependent on the

C-terminal motif LQCG (Fig. 5.10A). Strikingly, although DHFR and DHFRL1 share

93 % amino acid identity NleFE69 did not interact with DHFRL1 in this system (Fig.

5.10A). Olsen and colleagues further identified TMP21, a transmembrane protein

involved in vesicular transport, as an interacting partner of NleFE69 through a Y2H

screen (Olsen et al., 2013). Using the Clontech YDH system we were unable to

reproduce this interaction (Fig. 5.10B). Whether NleFE69 activates NF-κB through

inhibition of DHFR requires further research.

Page 146: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

146

The in vitro infection data and biochemical interaction assays used identify two

possible functions of NleFE69 during EPEC E69 infection. Firstly, that NleFE69 is

recruited to inhibit caspase-4 activity and inflammasome activation. Thus preventing

IL-18 activation and secretion and therefore acting as an anti-inflammatory effector.

Secondly, we identified that NleFE69 is essential for the early activation of NF-κB p65

nuclear translocation during epithelial cell infection. Subsequently, NleFE69 stimulates

the NF-κB dependent expression of pro-inflammatory immune modulators. To further

validate these putative functional roles of NleFE69 we sought to characterise NleFCR

in vivo using the C. rodentium mouse model of A/E pathogen infection.

Figure 5 10 NleFE69 interacts with DHFR but not DHFRL1. A Yeast-direct-Hybrid assay co-transforming AH109 with full length NleFE69 (pGBT9-nleFE69) or the C-terminal truncated NleFE69 (pGBT9-nleF1-185 E69) with (A) DHFR (pGADT7-DHFR), DHFRL1 (pGADT7-DHFRL1) or caspase-4 isoform-γ (pGADT7-CASP4) as a positive control. (B) A Yeast-direct-Hybrid assay co-transforming AH109 with full length NleFE69 (pGBT9-nleFE69) with TMP21 (pGADT7-TMP21) or caspase-4 isoform-γ (pGADT7-CASP4) as a positive control. Results are representative of two biological repeats

Page 147: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

147

1.15 Discussion

An NF-κB luciferase reporter assay screen, carried out by our collaborators, against

ectopically expressed EPEC E69 T3SS effectors previously detected NleFE69 as a

putative activator of the pro-inflammatory and pro-survival transcription factor NF-κB.

This study further confirmed that ectopic expression of NleFE69 in the HeLa cell line

promotes the nuclear translocation of the NF-κB p65 subunit; leading to the up-

regulation of the expression of the pro-inflammatory chemokine IL-8. The C-terminal

motif of NleFE69, as described above, is required for optimal binding of the interacting

partner’s caspase-4, -8 and -9. Ectopic expression of an NleFE69 mutant lacking the

four C-terminal amino acids LQCG abolished the pro-inflammatory activity of NleFE69.

This suggested that NleFE69 induced NF-κB activation may be caspase-dependent.

Furthermore this domain was essential for the mitochondrial subcellular localisation

observed for ectopically expressed NleFE69; although, it is important to note that the

ability of NleFE69 to activate NF-κB was not dependent on its subcellular localisation.

Early during infection EPEC E69 activates pro-inflammatory signalling in a T3SS

dependent-manner. Initially, EPEC E69 induces a pro-inflammatory response

through activation of ERK1/2 (Czerucka et al., 2001; de Grado et al., 2001; Savkovic

et al., 2001) and NF-κB (Newton et al., 2010; Savkovic, Koutsouris & Hecht, 1997).

Although pro-inflammatory effectors have been discovered in other A/E

pathogens/pathotypes including EspT (Raymond et al., 2011), the effector

responsible during EPEC E69 infection was previously unknown. In our study we

observed that NleFE69 was essential for activation of NF-κB and IL-8 expression

during EPEC E69 infection. Furthermore it was shown that the pro-inflammatory

activity of NleFE69 was inhibited by the TAB2/3 inhibitor NleE1. This along with the

evidence that NleFE69 stimulates the degradation of IκBα proposes a model where

Page 148: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

148

NleFE69 triggers NF-κB activation upstream to or at TAK1 phosphorylation (Fig. 5.11).

Whether ERK1/2, JNK or other MAPKs are activated in an NleFE69-dependent

manner was not investigated in this study and warrant further research. Although

NleFE69 was identified to be essential for the early activation of NF-κB during

infection the host target protein and mechanism of activation remain unknown.

1.15.1 Mechanism of NF-κB activation

Both caspase-4 and caspase-8 have previously been suggested to be integral for

canonical and non-canonical activation of NF-κB in response to TNFR/TLR ligand

binding (Nickles et al., 2012; Lakshmanan & Porter, 2007; Chaudhary et al., 2000).

Figure 5 11 EPEC E69 initially activates NF-κB then later inhibits inflammatory signalling in a T3SS dependent manner. EPEC E69 infection triggers an early NleFE69 dependent activation of the transcription factor NF-κB, acting up stream to TAB2/3 activation. At later time points post infection the T3SS effectors NleE1, NleB, NleC and NleD inhibit pro-inflammatory signalling.

Page 149: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

149

NleFE69 shares no structural homology to any known/characterised proteins and is

not predicted to have any enzymatic activity. With this in mind the current model

suggests that NleFE69 binds to and inhibits caspases through association with the

substrate binding domain in a C-terminal dependent-manner (Blasche et al., 2013).

Subsequently this prevents substrate binding and caspase proteolytic activity,

preventing downstream signalling. If knockdown of caspase-4/-8 successfully

prevented the pro-inflammatory activity of NleFE69 it would suggest that NleFE69

interacts with either caspase to directly promote activation of the intrinsic NF-κB

signalling cascade. However the pro-inflammatory activity of NleFE69 was determined

to be independent on its interaction with the known interacting partner’s caspase-4, -

8 or -9. Many viral effector proteins have been identified to bind to caspase-8 both to

the DED domain of the full length zymogen and to the active form (Mocarski, Upton

& Kaiser, 2011). While those that bind the active caspase form are restricted to

interaction with this structural form of caspase-8 (Mocarski, Upton & Kaiser, 2011),

NleFE69 can bind both the full length and active caspase-8 (Blasche et al., 2013).

Many of these viral effectors similarly bind caspase-8 to prevent extrinsic apoptosis

including Cowpox CrmA, Baculovirus p35 (Chaudhary et al., 2000), Human

papillomavirus Type 16 (HPV-16) E6/E7 (Vandermark et al., 2012) and Herpes

simplex virus 1 (HSV-1) R1/UL39 (Dufour et al., 2011); although, none have been

shown to subsequently activate NF-κB. Although this may not be a caspase-8

dependent mechanism, viral caspase-8 inhibitors have been reported to inhibit NF-

κB activation including HPV-16 E6/E7 (Vandermark et al., 2012). Therefore it is

unlikely that the pro-inflammatory activity of NleFE69 is a consequence of caspase-8

inhibition.

Page 150: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

150

The host target of NleFE69 responsible for NF-κB activation may be a yet

uncharacterised or a previously identified but untested host binding partner. Two

reported putative host binding partners include the transmembrane protein TMP21

(Olsen et al., 2013) and DHFR (Blasche, 2013) an essential enzyme in the supply of

biologically active folate for nucleic acid and glycine biosynthesis. Interaction with

TMP21 through the YDH system could not be reproduced in our hands. DHFR is an

essential enzyme in the production of thymine and is required for rapid cell division.

It is therefore an ideal drug target for not only chemotherapy (Methotrexate) but

through host specific anti-folates it is also targeted in the treatment of a spectrum of

microbial and parasitic infections including the bacteriostatic trimethoprim and the

anti-malarial’s pyrimethamine and cycloguanil (Rastelli et al., 2000; Schweitzer,

Dicker & Bertino, 1990). Thymidylate synthase (TS) is an essential enzyme for the

conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine

monophosphate (dTMP) downstream to DHFR. Interestingly, knockout of TS in a

Human colon carcinoma cell line induces thymineless stress and activation of NF-κB

and AP-1 (Harwood et al., 2000). Moreover the previously described DHFR

pseudogene DHFRP4, also known as DHFRL1, was recently shown to be expressed

and functional. DHRFL1, unlike the cytoplasmic/nuclear DHFR, is localised to the

mitochondria (McEntee et al., 2011). NleFE69 was confirmed to bind DHFR using the

YDH system, although DHFR and DHFRL1 share 93 % amino acid sequence

identity NleFE69 specifically interacted with DHFR only. This may be due to the

limitation of the YDH assay requiring the proteins to be localised to the nucleus and

alternative methods need to be used to rule out binding to DHFRL1. More research

is required to determine if NleFE69 targets DHFR/DHFRL1 during infection and

whether this interaction is key to NleFE69-dependent activation of NF-κB.

Page 151: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

151

To further elucidate the role of NleF and to determine the consequence of either NF-

κB activation or inhibition of caspase-4/-11, during in vivo infection, C57BL/6 mice

were infected with the EPEC/EHEC-like A/E pathogen C. rodentium.

Page 152: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

152

Results: Chapter 6 - NleFCR inhibits IL-18 secretion and neutrophil

recruitment in vivo

Page 153: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

153

6.1 NleFCR inhibits IL-18 secretion and neutrophil recruitment in vivo

To determine whether NleFE69 manipulates immune signalling during infection and to

confirm the function of NleFE69, the C. rodentium infection of C57BL/6 mice was used

as a model of in vivo A/E pathogen infection.

6.1.1 NleFCR is not required for normal colonisation in vivo

C57BL/6 mice were infected with the WT ICC169 C. rodentium, C. rodentium ΔnleF

or the complemented C. rodentium ΔnleF pACYC184-nleFCR (pnleFCR) strain by o.g.

Initially colonisation fitness of each strain was tested. Faeces were collected on the

days indicated and CFU were enumerated. The prototypic colonisation pattern of

C57BL/6 mice was demonstrated by the WT C. rodentium strain (Fig. 6.1A).

Following inoculation there was a dip in bacterial load which then picked back up at

day 4 and reached a peak at ~day 8 (Fig. 6.1A). Following day 8 WT C. rodentium

was slowly cleared and colonisation decreased. No difference in colonisation was

observed during infection with the C. rodentium ΔnleF or C. rodentium ΔnleF pnleFCR

strains in comparison to WT C. rodentium (Fig. 6.1A).

Figure 6 1 NleFCR is not required for C. rodentium colonisation. C57BL/6 mice were infected by o.g. with 1x108/ml WT C. rodentium or the ΔnleF C. rodentium and ΔnleF C. rodentium pnleFCR strains. Infection was followed from day 0 to day 14 and where indicated faeces were collected and plated. A minimum of five mice per group were used. Results are representative of two biological repeats

Page 154: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

154

6.1.2 C. rodentium delivers NleFCR to inhibit IL-18 secretion

Both Inflammasome activation and NF-κB are early innate immune responses to

bacterial infection, occurring at the epithelial cell barrier of the gastrointestinal tract.

To determine if NleFCR manipulates immune signalling during infection colon ex-

plants were cultured on day 4 and 8 post-infection and IL-18 cytokine secretion was

measured by ELISA. The inflammasome independent cytokine KC (CXCL1) was

measured as a control.

Infection with the C. rodentium ΔnleF mutant significantly increased IL-18 secretion

at day 4 post-infection in comparison to WT C. rodentium infection and the PBS

control (Fig. 6.2A). Importantly, no difference in the secretion of the NF-κB

dependent cytokine KC was observed during infection with WT C. rodentium or C.

rodentium ΔnleF in comparison to the PBS control (Fig. 6.2B). Complementation of

the C. rodentium ΔnleF strain with the plasmid encoded NleFCR reduced the

Figure 6 2 NleFCR inhibits early secretion of IL-18 from colonic tissue in vivo. C57BL/6 mice were infected by oral gavage with 1x108/ml WT C. rodentium or the ΔnleF C. rodentium and ΔnleF C. rodentium pnleFCR strains or inoculated with PBS as a control. On day 4 and 8 post-infection 1cm of mouse colons were isolated and cultured ex vivo for 24h. (A and C) IL-18 and (B) KC secretion were measured by ELISA. Results for day 4 p.i are an average of two biological repeats with 5-6 mice

Page 155: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

155

secretion of IL-18 to similar levels observed during infection with WT C. rodentium

(Fig. 6.2A). Conversely we observed an increase in KC secretion (Fig. 6.2B). The

latter result is consistent with our finding that NleFE69 activates NF-κB during

infection. NleFCR-dependent IL-18 secretion inhibitory effects were only observed

early during infection, as no difference in IL-18 secretion was observed on day 8 p.i.

(Fig. 6.2C). IL-18 facilitates the emigration of neutrophils and leukocytes to the site of

infection, although this has not been tested in the gastrointestinal tract (Dinarello &

Fantuzzi, 2003; Verri et al., 2007; Canetti et al., 2003).

6.2 Deletion of nleF increases colonic neutrophil recruitment during C.

rodentium infection

Colonic tissue from infected mice was extracted on day 4 post-infection with either

the WT C. rodentium, the C. rodentium ΔnleF or the C. rodentium ΔnleF pnleFCR

strain. The terminal colon was homogenised and the immune cell population was

evaluated by flow cytometry. A significant increase in neutrophil recruitment was

observed upon infection with the C. rodentium ΔnleF strain in comparison to the WT

C. rodentium strain (Fig. 6.3A). This increase in neutrophil recruitment was

sequestered upon complementation of the ΔnleF strain with the constitutive

expression plasmid pACYC184-nleFCR (Fig. 6.3A and Fig. 6.4A). The change in

neutrophil recruitment was specific and no change in macrophage, dendrocyte or

lymphocyte recruitment was observed upon infection with the ΔnleF strain in

comparison to the WT C. rodentium strain (Fig. 6.3A-B). Furthermore no difference

was observed for neutrophil recruitment between all strains at later time points of

infection at day 8 or 14 (Fig. 6.3C). This suggested that NleFCR specifically inhibits

the recruitment of neutrophils early during infection. To determine if neutrophil

Page 156: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

156

recruitment during C. rodentium ΔnleF infection is inflammasome dependent, the

infection was repeated in Casp1/11-/- mice.

Figure 6 3 NleFCR inhibits early colonic recruitment of neutrophils. C57BL/6 mice were infected by o.g. with WT, ΔnleF or ΔnleF C. rodentium pnleF strains or inoculated with PBS as a control. Immune cell recruitment into the colon was evaluated by flow cytometry as described in materials and methods (A) Neutrophils (CD11b+Ly6G+), macrophages (CD11b+Ly6G-F4/80+) and dendrocytes (CD11c+CD11b-) are presented as percentage of myeloid cells or absolute numbers (B) B cells (B220+CD3-), cytotoxic T cells (CD8+CD4-) and T helper cells (CD4+CD8-) are presented as a percentage of lymphocytes. Results are an average of two biological repeats with 4-6 mice per group. (C) Recruitment of neutrophils was further analysed on day 8 and 14 p.i. Results are an average of one biological repeat. *P<0.05, WT vs ΔnleF and ΔnleF vs ΔnleF pnleF, Mann-Whitney test. Results are expressed as the mean +/- the standard error of the mean. PI, post-infection.

Page 157: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

157

6.3 Recruitment of neutrophils and IL-18 secretion early during C. rodentium

infection is caspase-1/11 dependent.

Casp1/11-/- mice were infected with the WT C. rodentium or ΔnleF C. rodentium

strain and as before, at day 4 post infection, IL-18 secretion and immune cell

infiltrates were analysed by ELISA and FACs. Secretion of IL-18, as expected, was

completely abolished during C. rodentium infection of Casp1/11-/- mice (Fig. 6.4A).

As a control KC levels were also tested. No difference in KC was observed between

infections with either the WT C. rodentium or C. rodentium ΔnleF strain (Fig. 6.4A).

Furthermore the levels of KC were similar to that seen during WT C. rodentium

infection of WT C57BL/6 mice. This confirmed that at day 4 post infection, IL-18

secretion, and the increase observed upon deletion of nleF, is dependent on a

functional caspase-1/11 inflammasome.

Figure 6 4 NleFCR dependent inhibition of neutrophil recruitment is viacaspase-1/11. C57BL/6 Casp1/11-/- mice were infected by oral gavage with WT C.rodentium or the ΔnleF C. rodentium strains. (A) On day 4 post-infection 1cm ofmouse colons were isolated and cultured ex vivo for 24 h. KC and IL-18 secretionwere measured by ELISA. (B) Immune cell recruitment into the colon was evaluated by flow cytometry. Forward and side scatter plots were used to identify the myeloid cell and lymphocyte populations using FlowJo software. Doublets found within this population were excluded using FSC-W/FSC-A and SSC-W/SSC-A. Neutrophils (CD11b+Ly6G+) are presented as percentage of myeloid cells or absolute number.Results are an average of two biological repeats and expressed as the mean +/- thestandard error of the mean.

Page 158: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

158

The colonic immune cell population at day 4 post infection of Casp1/11-/- mice was

evaluated by flow cytometry. In the absence of caspase-1 and caspase-11, infection

with the C. rodentium ΔnleF mutant no longer induced increased neutrophil influx

(Fig. 6.5B). Neutrophil recruitment in mice infected with the ΔnleF strain was reduced

to that observed during WT C. rodentium infection (Fig. 6.5B). As seen previously,

no difference in the macrophage or leukocyte cell populations was observed during

infection with either the WT C. rodentium or C. rodentium ΔnleF strain (Data not

shown). This suggested that caspase-1 and caspase-11, and therefore the

inflammasome, is required for neutrophil recruitment early during C. rodentium

infection.

6.4 Discussion

C. rodentium NleF shares 84 % amino acid sequence identity with NleF of both

EPEC E69 and EHEC EDL933. Although little in vitro work with NleFCR has been

carried out in this study it was identified that NleFCR can inhibit the murine ortholog to

human caspase-4: caspase-11. We therefore proposed that NleF homologs may

have a conserved function during A/E pathogen infection.

As with many non-LEE encoded effectors NleFCR was not essential for colonisation

of C57BL/6 mice. In order to investigate if NleFE69 manipulates the host immune

response during infection the colons of infected mice were cultured ex-vivo on day 4

post-infection. Analysis of the supernatants revealed that C. rodentium inhibited

caspase-1/11-dependent, and therefore inflammasome-dependent, secretion of the

pro-inflammatory cytokine IL-18. Importantly, no difference in KC secretion of mice

infected with either the WT C. rodentium or C. rodentium ΔnleF strain was observed.

This demonstrated that C. rodentium successfully targets and inhibits the host

caspase-1/11 dependent inflammasome response during infection in an NleFCR

Page 159: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

159

dependent manner. At later time points post infection no difference in IL-18 secretion

was observed.

Early during C. rodentium infection IL-18 secretion is epithelial cell dependent and

NLRP3/NLRC4 independent (Nordlander, Pott & Maloy, 2014; Song-Zhao et al.,

2014). Alternatively at day 14 post infection both NLRC4 and caspase-11/NLRP3 are

essential for IL-18 secretion (Gurung et al., 2012; Liu et al., 2012). This is most likely

due to the influx of myeloid cells and the activation of the classical murine

macrophage non-canonical inflammasome (Kayagaki et al., 2011). Following this

model of inflammasome activation during C. rodentium infection we propose that

NleFCR is delivered into enterocytes to inhibit early activation of the caspase-4/11-

dependent non-canonical inflammasome. At later time points due to immune cell

infiltration the caspase-1 NLRC4/NLRP3-dependent inflammasomes takes

precedence. The NLRC4 inflammasome is not caspase-11 dependent and is

therefore not predicted to be manipulated by NleFCR. Moreover, C. rodentium and

EPEC E69 are extracellular pathogens and although many in vitro studies have

identified T3SS dependent mechanisms to prevent myeloid cell specific functions

including phagocytosis (Tahoun et al., 2011; Marches et al., 2008), whether NleFCR

is translocated into macrophages or not during C. rodentium infection is unknown. It

is therefore predicted that NleFCR is unable to prevent IL-18 secretion at later times

due to cell specificity.

The infection of Il-18-/- C57BL/6 mice with C. rodentium results in delayed clearance

and an increase in bacterial burden (Liu et al., 2012). IL-18 is therefore essential for

bacterial clearance and the host mounted immune response to C. rodentium

infection. IL-18 has been implicated to facilitate the emigration of neutrophils and

leukocytes to the site of insult (Verri et al., 2007; Canetti et al., 2003; Dinarello &

Page 160: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

160

Fantuzzi, 2003); although, a role for IL-18 in immune cell recruitment in the gastro-

intestinal tract in response to Gram negative bacterial infection has not been studied

previously. In order to characterise the recruitment of immune cells to the colon

during infection with either the WT C. rodentium or C. rodentium ΔnleF strain the

colonic immune cell population was analyzed at day 4 post infection by flow

cytometry. The increase in IL-18 secretion observed upon deletion of nleF correlated

with an increase in the infiltration of neutrophils. No change in recruitment of other

myeloid cell types or leukocytes was observed. Moreover as with the secretion of IL-

18 no change in immune cell recruitment was observed at day 8 post infection. The

increase in neutrophil recruitment in response to ΔnleF infection was abolished upon

infection of Casp1/11-/- mice. This suggested that neutrophil recruitment to the colon

early during infection is in fact inflammasome-dependent.

It is previously reported that neutrophil recruitment during C. rodentium infection is

CXCL2R dependent and essential for bacterial clearance and protection against

progression to diarrheagenic associated disease (Spehlmann et al., 2009). CXCL2

also commonly known as MIP2α, is the predominant CXCL2 ligand expressed early

during C. rodentium infection (Spehlmann et al., 2009). Further, it is essential for IL-

18-dependent up-regulation of the potent neutrophil chemokine LTB4 (Verri et al.,

2007; Canetti et al., 2003). Whether IL-18 is responsible for the CXCL2R-dependent

recruitment of neutrophils during C. rodentium infection requires further study.

Alternatively, the caspase-4/-11 dependent cytokine IL-1α (Casson et al., 2015;

Kayagaki et al., 2011) is also a potent neutrophil chemokine (Rider et al., 2011). With

the current evidence it is not possible to delineate which inflammasome dependent

cytokine is responsible for neutrophil recruitment observed upon deletion of nleF. To

Page 161: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

161

further investigate this, infections must be carried out in the C57BL/6 Il-18-/- and Il-1α-

/- backgrounds or in the presence of IL-18 binding protein, a known IL-18 inhibitor.

Recent studies have underlined the importance and contribution of non-

hematopoietic cell non-inflammasome and inflammasome- forming NLRs in intestinal

homeostasis and regulation of the intestinal immune response to enteric infection

(Elinav, Henao-Mejia & Flavell, 2013). Whilst non-hematopoietic and IEC expressed

NLRP6, NLRP12 and NLRC4 play integral roles in intestinal homeostasis and the

host mediated clearance of C. rodentium, the mechanisms by which they are

identified do so are independent of IL-1 family cytokine secretion (Allen, 2014;

Nordlander, Pott & Maloy, 2014; Wlodarska et al., 2014; Anand et al., 2012). Here

we propose the caspase-4 dependent IEC inflammasome is crucial for IL-18 cytokine

maturation and the early innate immune response to C. rodentium.

In conclusion we identified a novel in vivo role for the inhibition of IL-18 secretion and

inflammasome dependent neutrophil recruitment by the T3SS effector NleFCR during

C. rodentium infection. On the other hand our in vitro data also suggested a role for

NleFE69 in the T3SS dependent activation of the transcription factor NF-κB.

6.4.1 NleFCR as a pro-inflammatory effector of C. rodentium

C. rodentium employs a plethora of anti-inflammatory and immuno-modulatory

effectors to dampen the host immune response during infection. The extent to which

C. rodentium has evolved to prevent activation of this pathway with a multi-effector

and highly redundant approach indicates that the suppression of the inflammatory

response is critical to its infection strategy. While this may be true C. rodentium

infection induces a highly pro-inflammatory response in vivo leading to colitis and

murine colonic crypt hyperplasia (Collins et al., 2014). The host inflammatory

Page 162: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

162

response early during colonisation is associated with alterations in the host

commensal microbiota that is advantageous to bacterial pathogen growth (Lupp et

al., 2007; Khan et al., 2006). This balance between activation and inhibition may be

critical for C. rodentium to out-compete commensal bacteria whilst limiting host-

mediated immune clearance. Furthermore deletion of Tlr4 does not dampen the host

immune response during C. rodentium infection (Khan et al., 2006). Together this

suggests that inflammation may be directly triggered by C. rodentium to aid in

colonisation of the host. We identified that the T3SS effector NleFE69 is essential for

the early activation of NF-κB and IL-8 expression during in vitro EPEC epithelial cell

infection. While ELISAs were also carried out on supernatants post tissue culture

infection IL-8 was below detectable limits (data not shown). Although no difference

in KC (the murine homolog to IL-8) secretion or crypt hyperplasia (data not shown)

was observed upon infection of C57BL/6 mice with WT C. rodentium or the C.

rodentium ΔnleF strain, it was observed that overexpression of NleFCR in the

complemented strain increased colonic KC secretion while still successfully inhibiting

IL-18 secretion. C. rodentium and EPEC E69 share many homologous effectors but

also encode unique pathotype specific effectors (Wong et al., 2011). One such C.

rodentium effector is the pro-inflammatory GEF WxxE family protein EspT; which

regulates TNFα and KC expression in vivo (Raymond et al., 2011). Therefore, the

pro-inflammatory role of NleFCR may be redundant. Alternatively, there was little

difference in KC secretion between the WT C. rodentium infected and the PBS

control mice at day 4 p.i.. By applying more sensitive assays over a time course,

including qPCR, and through the screening of other NF-κB dependent

genes/cytokines, it may be possible to provide a more thorough analysis and

understanding of the role of NleFCR during C. rodentium infection of mice. .

Page 163: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

163

It is important to consider here that C. rodentium is only a model for EPEC infection.

Although EPEC and C. rodentium share many genes and a homologus T3SS, the

diarrhoeal disease associated with EPEC infection differs to that of C. rodentium.

6.4.2 The role of NleF in the context of EPEC infection

Both EHEC and C. rodentium trigger what is classified as an inflammatory diarrhoeal

disease, similar to the invasive and pro-inflammatory enteric pathogens Yersinia,

Salmonella, and Shigella (Hodges & Gill, 2010). On the other hand EPEC is

associated with a weak pro-inflammatory, watery diarrhoeal diease (Hodges & Gill,

2010).

EPEC E69 is exclusively an extracellular pathogen and therefore unlike other enteric

pathogens including Salmonella or Shigella it must successfully dampen

inflammation and immune cell recruitment in order to survive and colonise the host.

Contrary to this many studies including in vitro (Czerucka et al., 2001; de Grado et

al., 2001; Newton et al., 2010; Savkovic, Koutsouris & Hecht, 1997), ex vivo

(Schuller et al. 2009) and in vivo (Fagundes-Neto et al., 2000; Hill, Phillips & Walker-

Smith, 1991) EPEC infections have identified that EPEC induces IL-8 expression

and low level inflammation in both a flagellin and flagellin-independent mechanism

and that activation of NF-κB is associated with the acculumaltion of fluid and

diarrhoeal disease (Hodges & Gill, 2010). Throughout the 1980’s many histological

studies were conducted on biopsies of the small bowels from patients with acute or

persistent/chronic infection as well as from patients challenged with the prototypic

E2348/69 strain (As reviewed in Chen & Frankel, 2005). While all studies

consistently observed that similarly to C. rodentium EPEC triggers microvilli atrophy

and A/E lesion formation in vivo (Collins et al., 2014; Kallas, Patricio & Fagundes-

Neto, 1995) there were discrepancies in the ability of EPEC infection to induce

Page 164: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

164

inflammation and immune cell infiltration. Several studies identified lymphocyte,

eosinophil and/or neutrophil infiltration in response to persistent/chronic EPEC

infection (0119, O111, O125 etc.) (Fagundes-Neto et al., 2000; Hill, Phillips &

Walker-Smith, 1991) alternatively multiple studies revealed little or no change in

intestinal lymphocyte numbers in the biopsies obtained from patients diagnosed with

acute or persistent infection (Fagundes-Neto, Kallas & Patricio, 1997; Kallas, Patricio

& Fagundes-Neto, 1995). The discrepencies on the human biopsies could be

explained by differences in patient flora and immune status, strains that were

ioslated or the limitation and power of the analytical tools used (mostly microscopy).

Furthermore EPEC infection of mice (strain dependent) results in a similar infection

phenotype to that of C. rodentium, inducing inflammation and colonic inflammatory

cell recruitment (Zhang et al. 2012).

Induction of NF-κB is not only linked to immune cell recruitment. EPEC induced NF-

κB activation is also implicated in the upregulation of the galanin-1 receptor, which

increases Cl- secretion and fluid accumulation (Hodges & Gill, 2010). Thus linking

EPEC induced inflammation to the watery diarrhoeal disease associated with EPEC

infection. The current literature, in vitro and in vivo data and the avaibale biopsies

from EPEC infected infants and clinical trials support that whilst EPEC is able to

dampen the host immune response it most likely strikes a balance between pro- and

anti-inflammatory signals; resulting in a waek inflammatory diarrhoeal disease.

With regard to this a proposed function of NleFE69 in the inhibition of the host

caspase-4 dependent inflammasome whilst concurrently activating NF-κB aligns well

with the histopathological consequences and clinical symptoms of both EPEC and C.

rodentium infection.

Page 165: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

165

Chapter 7: Conclusions

Page 166: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

166

7.1 Overview

EPEC E69 and EHEC belong to a family of A/E pathogens that colonise the human

gastrointestinal tract inducing a diarrhoeal disease, which in the case of EHEC can

progress to HUS and renal failure (Wong et al., 2011). Virulence and pathogenesis is

dependent on the LEE-encoded T3SS and the translocation of numerous effectors to

manipulate host cell signalling including the innate immune response (Raymond et

al., 2013). Characterisation of several effectors has expanded our understanding of

the infection strategy of A/E pathogens and furthermore has increased our

knowledge of the host mounted immune response to clear infection. To date, the

functional role of many effectors still remains unknown. The aim of this study was to

characterise the host interacting partners and the functional role of the T3SS effector

NleF homologs during A/E pathogen infection.

7.2 Conclusions and implications

In this study we conclude that the effector NleFE69 of O126:H7 EPEC E2348/69 is

delivered during infection to stimulate the activation of the NF-κB pathway and act as

a caspase-4 inhibitor. Through the C-terminal motif LQCG NleFE69 binds to the

putative substrate binding domain of the hetero-tetramer active caspase-4 complex,

inhibiting proteolytic activity. We further confirmed that human caspase-4 is the

functional ortholog to murine caspase-11 and can functionally complement the

inflammatory caspase to stimulate IL-1α and IL-1β secretion, in response to Gram-

negative bacterial infection. Following this we further verified that human caspase-4

is essential for the secretion of active IL-18 during EPEC E69 infection of IECs and

that NleFE69 is delivered to inhibit the epithelial cell caspase-4 dependent non-

canonical inflammasome. Subsequently NleFCR dependent inhibition of

inflammasome activity in vivo inhibits IL-18 secretion and neutrophil influx during C.

Page 167: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

167

rodentium infection. This highlights a fundamental and novel role for the T3SS

effector NleF in the pathogenesis and virulence of A/E pathogens. Furthermore the

the fact that EPEC and C. rodentium have evolved mechanisms to specifically

prevent the recruitment and infiltration of neutrophils suggests that this cell type may

play an important role in the host immune response and outcome to A/E pathogen

infection.

Further to adding to our understanding of the pathogenesis and virulence

mechanisms employed by A/E pathogens this study contributes to our understanding

of the host non-canonical inflammasome response in vivo. The host inflammasome

response is a major pathogenic mediator of auto-immune, auto-inflammatory and

infectious disease (Dinarello & Fantuzzi, 2003). IL-18 specifically is a major mediator

of inflammatory bowel disease, psoriasis, myocardial pathology and sepsis (Dinarello

& Fantuzzi, 2003), leading to high morbidity and a lower quality of life, whilst being a

substantial economic burden in the developed world (Kappelman et al., 2011). IL-18

is as a possible therapeutic target for many of these complications and diseases

including IBD (Kanai, Kamada & Hisamatsu, 2013). This research further contributes

to our understanding and knowledge of how IL-18 secretion is regulated in the G.I

tract in response to bacterial infection and further expands on the role of the non-

hematopoietic inflammasome in the host innate immune response.

Page 168: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

168

Acknowledgements

I would like to thank and acknowledge the following for providing essential resources

for this study:

Dr. Anne Parle-McDermott, School of Biotechnology, Dublin City University

Dr. Olivier Marches, Centre for Immunobiology, Barts and the London, QMUL

Dr. Gunnar Schroeder, CMBI, Imperial College London

Dr. Isabel Garcia-Saez, Institut de Biologie Structurale, Grenoble

Dr. A Shenoy, CMBI, Imperial College London

Plasmids provided by Dr. A. Parle-McDermott are bound by Material transfer

agreements between Imperial College London and Dublin City University. For this

reason these plasmids cannot be transferred to a third party.

I would also like to take this opportunity to thank our collaborator Dr. Avinash Shenoy

for his expertise and guidance in immunology and the host inflammasome response.

It was pleasure working with you and I truly felt a member of your group. I am forever

grateful for your assistance and wish you and your group the best of luck in the

future

I would like to thank both Dr. Mariella Lomma and Dr. Benoit Raymond for their

invaluable guidance, training and supervision that they provided for me at the

beginning of my PhD. I would also like to extend my gratitude and thanks to the

Post-docs of the Frankel group Dr. Gunnar Schroeder and Dr. Valerie Crepin-

Sevenou for always being approachable and for the training and assistance that they

both provided throughout my PhD. I would also like to thank Dr. Olga Kotik for her

Page 169: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

169

commitment and dedication to the purification of caspase-4 and Dr. Maryam

Habibzay for her work on the in vivo role of NleFCR.

Many thanks goes to my PhD supervisor Dr. Cedric Berger. Throughout my PhD you

have been extremely helpful and patient. I am thankful for the training that you

provided and the guidance, support and feedback that you imparted throughout the

last 4 years. While my learning and training will go on forever you have helped build

the starting blocks to my research career and for that I am forever grateful –merci.

Thank you to both Dr. Vladimir Pelicic and Dr. Ray Carabeo, for taking up the roles

as my progress review advisers and for the integral feedback given for my project

proposal and both my 9 month and 18 month reports.

I am very grateful for the opportunity provided to me to carry out this research by

Prof. Gad Frankel, the Biotechnology and Biological Sciences Research Council and

Novartis. Prof. Gad Frankel has been a great source of support and guidance

throughout this research without whom it would not be possible. Words cannot

explain my gratitude and appreciation to have been provided with the chance to work

for Gadi and I will never forget my time and lessons learnt under his supervision.

Thank you Gadi!

The Frankel and Filloux group of CMBI made coming to work a joy. Each of you has

helped me through my research in one way or another and you will all be sorely

missed. Thank you all for listening time and time again and providing your opinion

and expert advice – sharing is caring. I would further like to thank Phillipa Goddard

for her assistance on the project during her Master’s project and her continuing

support and communication during her PhD. I wish you the best of luck.

Page 170: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

170

None of this would have been possible without the support of my wife Emma Pallett.

I am eternally grateful to you for listening to the latest setback, or negative result,

and for always being the optimist, to always being there for me and for being you.

You are the reason I persevered, I am very lucky and I love you.

I would also like to thank my mother and father in-law Dr. Dennis and Dr. Matilda

Buckley for their continued support through-out my research career and for

welcoming me to the family. I am forever thankful for you both.

Page 171: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

171

Bibliography

Abba, K., R. Sinfield, C.A. Hart, and P. Garner. 2009. Pathogens associated with persistent diarrhoea in children in low and middle income countries: systematic review. BMC Infect. Dis. 9:88-2334-9-88.

Abderrazak, A., T. Syrovets, D. Couchie, K. El Hadri, B. Friguet, T. Simmet, and M. Rouis. 2015. NLRP3 inflammasome: from a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biol. 4:296-307.

Allen, I.C. 2014. Non-Inflammasome Forming NLRs in Inflammation and Tumorigenesis. Front. Immunol. 5:169.

Anand, P.K., R.K. Malireddi, J.R. Lukens, P. Vogel, J. Bertin, M. Lamkanfi, and T.D. Kanneganti. 2012. NLRP6 negatively regulates innate immunity and host defence against bacterial pathogens. Nat. 488:389-393.

Baldi, D.L., E.E. Higginson, D.M. Hocking, J. Praszkier, R. Cavaliere, C.E. James, V. Bennett-Wood, K.I. Azzopardi, L. Turnbull, T. Lithgow, R.M. Robins-Browne, C.B. Whitchurch, and M. Tauschek. 2012. The type II secretion system and its ubiquitous lipoprotein substrate, SslE, are required for biofilm formation and virulence of enteropathogenic Escherichia coli. Infect. Immun. 80:2042-2052.

Berger, C.N., O. Billker, T.F. Meyer, A.L. Servin, and I. Kansau. 2004. Differential recognition of members of the carcinoembryonic antigen family by Afa/Dr adhesins of diffusely adhering Escherichia coli (Afa/Dr DAEC). Mol. Microbiol. 53:963-983.

Bergsbaken, T., S.L. Fink, and B.T. Cookson. 2009. Pyroptosis: host cell death and inflammation. Nat. Rev. Microbiol. 7:99-109.

Bielaszewska, M., T. Aldick, A. Bauwens, and H. Karch. 2014. Hemolysin of enterohemorrhagic Escherichia coli: structure, transport, biological activity and putative role in virulence. Int. J. Med. Microbiol. 304:521-529.

Black, R.E., S. Cousens, H.L. Johnson, J.E. Lawn, I. Rudan, D.G. Bassani, P. Jha, H. Campbell, C.F. Walker, R. Cibulskis, T. Eisele, L. Liu, C. Mathers, and Child Health Epidemiology Reference Group of WHO and UNICEF. 2010. Global, regional, and national causes of child mortality in 2008: a systematic analysis. Lancet 375:1969-1987.

Blasche, S. 2013. Escherichia coli as Host and Pathogen. Heidelberg University.

Blasche, S., M. Mortl, H. Steuber, G. Siszler, S. Nisa, F. Schwarz, I. Lavrik, T.M.A. Gronewold, K. Maskos, M.S. Donnenberg, D. Ullmann, P. Uetz, and M. Kogl. 2013. The E. coli Effector Protein NleF Is a Caspase Inhibitor. PLoS One 8:e58937.

Brodsky I.E., N.W. Palm, S. Sadanand, M.B. Ryndak, F.S. Sutterwala, R.A. Flavell, J.B. Bliska and R. Medzhitov. 2010. A yersinia effector protein promotes virulence by

Page 172: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

172

preventing inflammasome recognition of the type III secretion system. Cell Host Microbe 7(5):376-87.

Canetti, C.A., B.P. Leung, S. Culshaw, I.B. McInnes, F.Q. Cunha, and F.Y. Liew. 2003. IL-18 enhances collagen-induced arthritis by recruiting neutrophils via TNF-alpha and leukotriene B4. J. Immunol. 171:1009-1015.

Casson, C.N., J. Yu, V.M. Reyes, F.O. Taschuk, A. Yadav, A.M. Copenhaver, H.T. Nguyen, R.G. Collman, and S. Shin. 2015. Human caspase-4 mediates noncanonical inflammasome activation against gram-negative bacterial pathogens. Proc. Natl. Acad. Sci. U. S. A. 112:6688-6693.

Chang, K., K. Marran, A. Valentine, and G.J. Hannon. 2013. Creating an miR30-based shRNA vector. Cold Spring Harb. Protoc. 2013:631-635.

Chantret, I., A. Rodolosse, A. Barbat, E. Dussaulx, E. Brot-Laroche, A. Zweibaum, and M. Rousset. 1994. Differential expression of sucrase-isomaltase in clones isolated from early and late passages of the cell line Caco-2: evidence for glucose-dependent negative regulation. J. Cell. Sci. 107 ( Pt 1):213-225.

Chaudhary P, M. Eby, A. Jasmin, A. Kumar, L. Liu, and L. Hood. 2000. Activation of the NF-kappaB pathway by caspase 8 and its homologs. Oncogene 19:4451-60.

Chen, H.D., and G. Frankel. 2005. Enteropathogenic Escherichia coli: unravelling pathogenesis. FEMS Microbiol. Rev. 29:83-98.

Christie, P.J., N. Whitaker, and C. González-Rivera. 2014. Mechanism and structure of the bacterial type IV secretion systems. Biochimica Et Biophysica Acta (BBA) 1843:1578-1591.

Clements, A., K. Smollett, S.F. Lee, E.L. Hartland, M. Lowe, and G. Frankel. 2011. EspG of enteropathogenic and enterohemorrhagic E. coli binds the Golgi matrix protein GM130 and disrupts the Golgi structure and function. Cell. Microbiol. 13:1429-1439.

Clements, A., J.C. Young, N. Constantinou, and G. Frankel. 2012. Infection strategies of enteric pathogenic Escherichia coli. Gut Microbes 3:71-87.

Collins, J.W., K.M. Keeney, V.F. Crepin, V.A. Rathinam, K.A. Fitzgerald, B.B. Finlay, and G. Frankel. 2014. Citrobacter rodentium: infection, inflammation and the microbiota. Nat. Rev. Microbiol. 12:612-623.

Croxen, M.A., and B.B. Finlay. 2010. Molecular mechanisms of Escherichia coli pathogenicity. Nat. Rev. Microbiol. 8:26-38.

Czerucka D, S. Dahan, B. Mograbi, B. Rossi, and P. Rampal. 2001. Implication of mitogen-activated protein kinases in T84 cell responses to enteropathogenic Escherichia coli infection. Infect. Immun. 69(3):1298-305.

Page 173: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

173

Daig, R., G. Rogler, E. Aschenbrenner, D. Vogl, W. Falk, V. Gross, J. Scholmerich, and T. Andus. 2000. Human intestinal epithelial cells secrete interleukin-1 receptor antagonist and interleukin-8 but not interleukin-1 or interleukin-6. Gut 46:350-358.

Dale, G.E., C. Broger, H. Langen, A. D'Arcy, and D. Stuber. 1994. Improving protein solubility through rationally designed amino acid replacements: solubilization of the trimethoprim-resistant type S1 dihydrofolate reductase. Protein Eng. 7:933-939.

Damm, A., K. Lautz, and T.A. Kufer. 2013. Roles of NLRP10 in innate and adaptive immunity. Microbes Infect. 15:516-523.

Datsenko, K.A., and B.L. Wanner. 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.

Dautin, N. 2010. Serine protease autotransporters of enterobacteriaceae (SPATEs): biogenesis and function. Toxins (Basel) 2:1179-1206.

de Grado M, C.M. Rosenberger, A. Gauthier, B.A. Vallance, B.B. Finlay. 2001. Enteropathogenic Escherichia coli infection induces expression of the early growth response factor by activating mitogen-activated protein kinase cascades in epithelial cells. Infect. Immun. 69(10):6217-24.

Dean, P., S. Muhlen, S. Quitard, and B. Kenny. 2010. The bacterial effectors EspG and EspG2 induce a destructive calpain activity that is kept in check by the co-delivered Tir effector. Cell. Microbiol. 12:1308-1321.

Dinarello, C.A., and G. Fantuzzi. 2003. Interleukin-18 and host defense against infection. J. Infect. Dis. 187 Suppl 2:S370-84.

Donnenberg, M.S. 2013. Escherichia coli: Pathotypes and Principles of Pathogenesis. 2nd ed. Waltham, MA: Academic Press.

Donnenberg, M.S. 2002. E. coli: Genomics, Evolution and Pathogenesis. 1st ed. Waltham, MA: Academic Press.

Dufour, F., A.M. Sasseville, S. Chabaud, B. Massie, R.M. Siegel, and Y. Langelier. 2011. The ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 protect cells against TNFalpha- and FasL-induced apoptosis by interacting with caspase-8. Apoptosis 16:256-271.

Dunne, A., and L.A. O'Neill. 2003. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduction during inflammation and host defense. Sci. STKE 2003:re3.

Echtenkamp F., W. Deng, W.E. Wickham, A. Vazquez, J.L Puente, A. Thanabalasuriar, S. Gruenheid, B.B. Finlay and P.R. Hardwidge. 2008. Characterization of the NleF effector protein from attaching and effacing bacterial pathogens. FEMS Microbiol Lett 281(1):98-107.

Page 174: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

174

Elinav, E., J. Henao-Mejia, and R.A. Flavell. 2013. Integrative inflammasome activity in the regulation of intestinal mucosal immune responses. Mucosal Immunol. 6:4-13.

Elliott S.J., L.A. Wainwright, T.K. McDaniel, K.G. Jarvis, Y. Deng, L. Lai, B.P. McNamara, M.S Donnenberg, J.B Kaper. 1998. The complete sequence of the locus of enterocyte effacement (LEE) from enteropathogenic Escherichia coli E2348/69. Mol. Microbiol. 28(1):1-4.

Emmerich, C.H., A. Ordureau, S. Strickson, J.S. Arthur, P.G. Pedrioli, D. Komander, and P. Cohen. 2013. Activation of the canonical IKK complex by K63/M1-linked hybrid ubiquitin chains. Proc. Natl. Acad. Sci. U. S. A. 110:15247-15252.

Fagundes-Neto, U., S. De Martini-Costa, M.Z. Pedroso, and I.C. Scaletsky. 2000. Studies of the small bowel surface by scanning electron microscopy in infants with persistent diarrhea. Braz. J. Med. Biol. Res. 33:1437-1442.

Fagundes-Neto, U., M.R. Kallas, and F.R. Patricio. 1997. Morphometric study of the small bowel mucosa in infants with diarrhea due to enteropathogenic Escherichia coli strains. Hepatogastroenterology 44:1051-1056.

Felisberto-Rodrigues, C., E. Durand, M.S. Aschtgen, S. Blangy, M. Ortiz-Lombardia, B. Douzi, C. Cambillau, and E. Cascales. 2011. Towards a structural comprehension of bacterial type VI secretion systems: characterization of the TssJ-TssM complex of an Escherichia coli pathovar. PLoS Pathog. 7:e1002386.

Fellmann, C., T. Hoffmann, V. Sridhar, B. Hopfgartner, M. Muhar, M. Roth, D.Y. Lai, I.A. Barbosa, J.S. Kwon, Y. Guan, N. Sinha, and J. Zuber. 2013. An optimized microRNA backbone for effective single-copy RNAi. Cell. Rep. 5:1704-1713.

Frankel G and A.D. Phillips. 2008. Attaching effacing Escherichia coli and paradigms of tir-triggered actin polymerization: Getting off the pedestal. Cell Microbiol. 10(3):549-56.

Galan JE, C. Ginocchio and P. Costeas. 1992. Molecular and functional characterization of the salmonella invasion gene invA: Homology of InvA to members of a new protein family. J. Bacteriol. 174(13):4338-49.

Gao, X., X. Wang, T. Pham, L. Feuerbacher, M. Lubos, M. Huang, R. Olsen, A. Mushegian, C. Slawson, and P. Hardwidge. 2013. NleB, a Bacterial Effector with Glycosyltransferase Activity, Targets GAPDH Function to Inhibit NF-κB Activation. Cell Host Microbe 13:87-99.

Garcia-Angulo, V.A., V.I. Martinez-Santos, T. Villasenor, F.J. Santana, A. Huerta-Saquero, L.C. Martinez, R. Jimenez, C. Lara-Ochoa, J. Tellez-Sosa, V.H. Bustamante, and J.L. Puente. 2012. A distinct regulatory sequence is essential for the expression of a subset of nle genes in attaching and effacing Escherichia coli. J. Bacteriol. 194:5589-5603.

Garlanda, C., C.A. Dinarello, and A. Mantovani. 2013. The interleukin-1 family: back to the future. Immunity 39:1003-1018.

Page 175: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

175

Garmendia J, A.D. Phillips, M. Carlier, Y. Chong, S. Schüller, O. Marches, S. Dahan, E. Oswald, R.K. Shaw and S. Knutton and G. Frankel. 2004. TccP is an enterohaemorrhagic escherichia coli O157:H7 type III effector protein that couples tir to the actin-cytoskeleton? Cell Microbiol. 6(12):1167-83.

Gauthier, A., N.A. Thomas, and B.B. Finlay. 2003. Bacterial Injection Machines. J. of Biol. Chem. 278:25273-25276.

Girard, F., I. Batisson, G.M. Frankel, J. Harel, and J.M. Fairbrother. 2005. Interaction of enteropathogenic and Shiga toxin-producing Escherichia coli and porcine intestinal mucosa: role of intimin and Tir in adherence. Infect. Immun. 73:6005-6016.

Gruenheid, S., R. DeVinney, F. Bladt, D. Goosney, S. Gelkop, G.D. Gish, T. Pawson, and B.B. Finlay. 2001. Enteropathogenic E. coli Tir binds Nck to initiate actin pedestal formation in host cells. Nat. Cell Biol. 3:856-859.

Gueguen, E., and E. Cascales. 2013. Promoter swapping unveils the role of the Citrobacter rodentium CTS1 type VI secretion system in interbacterial competition. Appl. Environ. Microbiol. 79:32-38.

Gurung, P., R.K. Malireddi, P.K. Anand, D. Demon, L. Vande Walle, Z. Liu, P. Vogel, M. Lamkanfi, and T.D. Kanneganti. 2012. Toll or interleukin-1 receptor (TIR) domain-containing adaptor inducing interferon-beta (TRIF)-mediated caspase-11 protease production integrates Toll-like receptor 4 (TLR4) protein- and Nlrp3 inflammasome-mediated host defense against enteropathogens. J. Biol. Chem. 287:34474-34483.

Guzman L.M., D. Belin, M.J. Carson and J. Beckwith. 1995. Tight regulation, modulation, and high-level expression by vectors containing the arabinose PBAD promoter. J. Bacteriol. 177(14):4121-30.

Hart, E., J. Yang, M. Tauschek, M. Kelly, M.J. Wakefield, G. Frankel, E.L. Hartland, and R.M. Robins-Browne. 2008. RegA, an AraC-like protein, is a global transcriptional regulator that controls virulence gene expression in Citrobacter rodentium. Infect. Immun. 76:5247-5256.

Harwood, F.G., S. Kasibhatla, I. Petak, R. Vernes, D.R. Green, and J.A. Houghton. 2000. Regulation of FasL by NF-kappaB and AP-1 in Fas-dependent thymineless death of human colon carcinoma cells. J. Biol. Chem. 275:10023-10029.

Hemrajani, C., C.N. Berger, K.S. Robinson, O. Marches, A. Mousnier, and G. Frankel. 2010. NleH effectors interact with Bax inhibitor-1 to block apoptosis during enteropathogenic Escherichia coli infection. Proc. Natl. Acad. Sci. U. S. A. 107:3129-3134.

Hill, S.M., A.D. Phillips, and J.A. Walker-Smith. 1991. Enteropathogenic Escherichia coli and life threatening chronic diarrhoea. Gut 32:154-158.

Hitomi, J., T. Katayama, Y. Eguchi, T. Kudo, M. Taniguchi, Y. Koyama, T. Manabe, S. Yamagishi, Y. Bando, K. Imaizumi, Y. Tsujimoto, and M. Tohyama. 2004.

Page 176: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

176

Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Abeta-induced cell death. J. Cell Biol. 165:347-356.

Hodges, K., and R. Gill. 2010. Infectious diarrhea: Cellular and molecular mechanisms. Gut Microbes 1:4-21.

Hodgson, A., E.M. Wier, K. Fu, X. Sun, H. Yu, W. Zheng, H.P. Sham, K. Johnson, S. Bailey, B.A. Vallance, and F. Wan. 2015. Metalloprotease NleC Suppresses Host NF-kappaB/Inflammatory Responses by Cleaving p65 and Interfering with the p65/RPS3 Interaction. PLoS Pathog. 11:e1004705.

Iguchi, A., N.R. Thomson, Y. Ogura, D. Saunders, T. Ooka, I.R. Henderson, D. Harris, M. Asadulghani, K. Kurokawa, P. Dean, B. Kenny, M.A. Quail, S. Thurston, G. Dougan, T. Hayashi, J. Parkhill, and G. Frankel. 2009. Complete Genome Sequence and Comparative Genome Analysis of Enteropathogenic Escherichia coli O127:H6 Strain E2348/69. J. Bacteriol. 191:347-354.

Impey, S., S.R. McCorkle, H. Cha-Molstad, J.M. Dwyer, G.S. Yochum, J.M. Boss, S. McWeeney, J.J. Dunn, G. Mandel, and R.H. Goodman. 2004. Defining the CREB regulon: a genome-wide analysis of transcription factor regulatory regions. Cell 119:1041-1054.

Jeong, J.Y., H.S. Yim, J.Y. Ryu, H.S. Lee, J.H. Lee, D.S. Seen, and S.G. Kang. 2012. One-step sequence- and ligation-independent cloning as a rapid and versatile cloning method for functional genomics studies. Appl. Environ. Microbiol. 78:5440-5443.

Johnsborg, O., V. Eldholm, and L.S. Havarstein. 2007. Natural genetic transformation: prevalence, mechanisms and function. Res. Microbiol. 158:767-778.

Kallas, M.R., F.R. Patricio, and U. Fagundes-Neto. 1995. Small bowel morphometry in acute and persistent diarrhea due to classic enteropathogenic Escherichia coli. Arq. Gastroenterol. 32:91-98.

Kalman, D., O.D. Weiner, D.L. Goosney, J.W. Sedat, B.B. Finlay, A. Abo, and J.M. Bishop. 1999. Enteropathogenic E. coli acts through WASP and Arp2/3 complex to form actin pedestals. Nat. Cell Biol. 1:389-391.

Kanai, T., N. Kamada, and T. Hisamatsu. 2013. Clinical strategies for the blockade of IL-18 in inflammatory bowel diseases. Curr. Drug Targets 14:1392-1399.

Kaper, J.B., J.P. Nataro, and H.L.T. Mobley. 2004. Pathogenic Escherichia coli. Nat. Rev. Micro. 2:123-140.

Kappelman, M.D., C.Q. Porter, J.A. Galanko, S.L. Rifas-Shiman, D.A. Ollendorf, R.S. Sandler, and J.A. Finkelstein. 2011. Utilization of healthcare resources by U.S. children and adults with inflammatory bowel disease. Inflamm. Bowel Dis. 17:62-68.

Page 177: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

177

Karki, P., G.R. Dahal, and I.S. Park. 2007. Both dimerization and interdomain processing are essential for caspase-4 activation. Biochem. Biophys. Res. Commun. 356:1056-1061.

Kaur, P., A. Chakraborti, and A. Asea. 2010. Enteroaggregative Escherichia coli: An Emerging Enteric Food Borne Pathogen. Interdiscip. Perspect. Infect. Dis. 2010:

Kayagaki, N., S. Warming, M. Lamkanfi, L. Vande Walle, S. Louie, J. Dong, K. Newton, Y. Qu, J. Liu, S. Heldens, J. Zhang, W.P. Lee, M. Roose-Girma, and V.M. Dixit. 2011. Non-canonical inflammasome activation targets caspase-11. Nat. 479:117-121.

Kenny, B., R. DeVinney, M. Stein, D.J. Reinscheid, E.A. Frey, and B.B. Finlay. 1997. Enteropathogenic E. coli (EPEC) transfers its receptor for intimate adherence into mammalian cells. Cell 91:511-520.

Khan, M.A., C. Ma, L.A. Knodler, Y. Valdez, C.M. Rosenberger, W. Deng, B.B. Finlay, and B.A. Vallance. 2006. Toll-Like Receptor 4 Contributes to Colitis Development but Not to Host Defense during Citrobacter rodentium Infection in Mice. Infect. Immun. 74:2522-2536.

Knodler, L.A., S.M. Crowley, H.P. Sham, H. Yang, M. Wrande, C. Ma, R.K. Ernst, O. Steele-Mortimer, J. Celli, and B.A. Vallance. 2014. Noncanonical inflammasome activation of caspase-4/caspase-11 mediates epithelial defenses against enteric bacterial pathogens. Cell Host Microbe 16:249-256.

Knodler, L.A., S.M. Crowley, H.P. Sham, H. Yang, M. Wrande, C. Ma, R.K. Ernst, O. Steele-Mortimer, J. Celli, and B.A. Vallance. 2014. Noncanonical inflammasome activation of caspase-4/caspase-11 mediates epithelial defenses against enteric bacterial pathogens. Cell Host Microbe 16:249-256.

Kobayashi, T., M. Ogawa, T. Sanada, H. Mimuro, M. Kim, H. Ashida, R. Akakura, M. Yoshida, M. Kawalec, J.M. Reichhart, T. Mizushima, and C. Sasakawa. 2013. The Shigella OspC3 effector inhibits caspase-4, antagonizes inflammatory cell death, and promotes epithelial infection. Cell Host Microbe 13:570-583.

Kotik-Kogan, O., E.R. Valentine, D. Sanfelice, M.R. Conte, and S. Curry. 2008. Structural analysis reveals conformational plasticity in the recognition of RNA 3' ends by the human La protein. Structure 16:852-862.

Kotloff, K.L., J.P. Nataro, W.C. Blackwelder, D. Nasrin, T.H. Farag, S. Panchalingam, Y. Wu, S.O. Sow, D. Sur, R.F. Breiman, A.S. Faruque, A.K. Zaidi, D. Saha, P.L. Alonso, B. Tamboura, D. Sanogo, U. Onwuchekwa, B. Manna, T. Ramamurthy, S. Kanungo, J.B. Ochieng, R. Omore, J.O. Oundo, A. Hossain, S.K. Kumar, P., Q. Luo, T.J. Vickers, A. Sheikh, W.G. Lewis, and J.M. Fleckenstein. 2014. EatA, an immunogenic protective antigen of enterotoxigenic Escherichia coli, degrades intestinal mucin. Infect. Immun. 82:500-508.

Kyle, J.L., C.A. Cummings, C.T. Parker, B. Quinones, P. Vatta, E. Newton, S. Huynh, M. Swimley, L. Degoricija, M. Barker, S. Fontanoz, K. Nguyen, R. Patel, R.

Page 178: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

178

Fang, R. Tebbs, O. Petrauskene, M. Furtado, and R.E. Mandrell. 2012. Escherichia coli serotype O55:H7 diversity supports parallel acquisition of bacteriophage at Shiga toxin phage insertion sites during evolution of the O157:H7 lineage. J. Bacteriol. 194:1885-1896.

Lage, S.L., C. Longo, L.M. Branco, T.B. da Costa, L. Buzzo Cde, and K.R. Bortoluci. 2014. Emerging Concepts about NAIP/NLRC4 Inflammasomes. Front. Immunol. 5:309.

Lakshmanan, U., and A.G. Porter. 2007. Caspase-4 Interacts with TNF Receptor-Associated Factor 6 and Mediates Lipopolysaccharide-Induced NF-κB-Dependent Production of IL-8 and CC Chemokine Ligand 4 (Macrophage-Inflammatory Protein-1β). J. Immunol. 179:8480-8490.

Lee, J.K., S.H. Kim, E.C. Lewis, T. Azam, L.L. Reznikov, and C.A. Dinarello. 2004. Differences in signaling pathways by IL-1beta and IL-18. Proc. Natl. Acad. Sci. U. S. A. 101:8815-8820.

Levine, M., M., D. Nalin R., R. Hornick B., E. Bergquist J., D. Waterman H., C. Young R., S. Sotman, and B. Rowe. 1978. Escherichia coli strains that cause diarrhoea but do not produce heat-lable or heat-stable enterotoxins and are non-invasive. Lancet 311:1119-1122.

Li, M.D., and X. Yang. 2011. A Retrospective on Nuclear Receptor Regulation of Inflammation: Lessons from GR and PPARs. PPAR Res. 2011:742785.

Li, Q., and I.M. Verma. 2002. NF-[kappa]B regulation in the immune system. Nat. Rev. Immunol. 2:725-734.

Lim, K.H., and L.M. Staudt. 2013. Toll-like receptor signaling. Cold Spring Harb. Perspect. Biol. 5:a011247.

Liu, Z., M.H. Zaki, P. Vogel, P. Gurung, B.B. Finlay, W. Deng, M. Lamkanfi, and T.D. Kanneganti. 2012. Role of inflammasomes in host defense against Citrobacter rodentium infection. J. Biol. Chem. 287:16955-16964.

Lupp, C., M.L. Robertson, M.E. Wickham, I. Sekirov, O.L. Champion, E.C. Gaynor, and B.B. Finlay. 2007. Host-Mediated Inflammation Disrupts the Intestinal Microbiota and Promotes the Overgrowth of Enterobacteriaceae. Cell Host Microbe 2:119-129.

Marches, O., V. Covarelli, S. Dahan, C. Cougoule, P. Bhatta, G. Frankel, and E. Caron. 2008. EspJ of enteropathogenic and enterohaemorrhagic Escherichia coli inhibits opsono-phagocytosis. Cell. Microbiol. 10:1104-1115.

Mariathasan, S., and D.M. Monack. 2007. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat. Rev. Immunol. 7:31-40.

Martinon, F., and J. Tschopp. 2007. Inflammatory caspases and inflammasomes: master switches of inflammation. Cell Death Differ. 14:10-22.

Page 179: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

179

McDaniel, T.K., K.G. Jarvis, M.S. Donnenberg, and J.B. Kaper. 1995. A genetic locus of enterocyte effacement conserved among diverse enterobacterial pathogens. Proc. Natl. Acad. Sci. U. S. A. 92:1664-1668.

McEntee, G., S. Minguzzi, K. O'Brien, N. Ben Larbi, C. Loscher, C. O'Fagain, and A. Parle-McDermott. 2011. The former annotated human pseudogene dihydrofolate reductase-like 1 (DHFRL1) is expressed and functional. Proc. Natl. Acad. Sci. U. S. A. 108:15157-15162.

Miao, E.A., R.K. Ernst, M. Dors, D.P. Mao, and A. Aderem. 2008. Pseudomonas aeruginosa activates caspase 1 through Ipaf. Proc. Natl. Acad. Sci. U. S. A. 105:2562-2567.

Miao, E.A., I.A. Leaf, P.M. Treuting, D.P. Mao, M. Dors, A. Sarkar, S.E. Warren, M.D. Wewers, and A. Aderem. 2010. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat. Immunol. 11:1136-1142.

Miao, E.A., D.P. Mao, N. Yudkovsky, R. Bonneau, C.G. Lorang, S.E. Warren, I.A. Leaf, and A. Aderem. 2010. Innate immune detection of the type III secretion apparatus through the NLRC4 inflammasome. Proc. Natl. Acad. Sci. U. S. A. 107:3076-3080.

Mocarski, E.S., J.W. Upton, and W.J. Kaiser. 2011. Viral infection and the evolution of caspase 8-regulated apoptotic and necrotic death pathways. Nat. Rev. Immunol. 12:79-88.

Mogensen, T.H. 2009. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 22:240-73, Table of Contents.

Muhlen, S., M.H. Ruchaud-Sparagano, and B. Kenny. 2011. Proteasome-independent degradation of canonical NFkappaB complex components by the NleC protein of pathogenic Escherichia coli. J. Biol. Chem. 286:5100-5107.

Munday, N.A., J.P. Vaillancourt, A. Ali, F.J. Casano, D.K. Miller, S.M. Molineaux, T.T. Yamin, V.L. Yu, and D.W. Nicholson. 1995. Molecular cloning and pro-apoptotic activity of ICErelII and ICErelIII, members of the ICE/CED-3 family of cysteine proteases. J. Biol. Chem. 270:15870-15876.

Mundy, R., T.T. MacDonald, G. Dougan, G. Frankel, and S. Wiles. 2005. Citrobacter rodentium of mice and man. Cell. Microbiol. 7:1697-1706.

Mundy, R., L. Petrovska, K. Smollett, N. Simpson, R.K. Wilson, J. Yu, X. Tu, I. Rosenshine, S. Clare, G. Dougan, and G. Frankel. 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.

Mundy, R., D. Pickard, R.K. Wilson, C.P. Simmons, G. Dougan, and G. Frankel. 2003. Identification of a novel type IV pilus gene cluster required for gastrointestinal colonization of Citrobacter rodentium. Mol. Microbiol. 48:795-809.

Page 180: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

180

Nadler, C., K. Baruch, S. Kobi, E. Mills, G. Haviv, M. Farago, I. Alkalay, S. Bartfeld, T.F. Meyer, Y. Ben-Neriah, and I. Rosenshine. 2010. The Type III Secretion Effector NleE Inhibits NF-κB Activation. PLoS Pathog. 6:e1000743.

Nataro JP, J.B Kaper, R. Robins-Browne, V. Prado, P. Vial and M.M. Levine . 1987. Patterns of adherence of diarrheagenic Escherichia coli to HEp-2 cells. Pediatr. Infect. Dis. J. 6(9):829-31.

Nataro, J.P., and J.B. Kaper. 1998. Diarrheagenic Escherichia coli. Clin. Microbiol. Rev. 11:142-201.

Navarro-Garcia, F., A. Serapio-Palacios, J.E. Vidal, M.I. Salazar, and G. Tapia-Pastrana. 2014. EspC promotes epithelial cell detachment by enteropathogenic Escherichia coli via sequential cleavages of a cytoskeletal protein and then focal adhesion proteins. Infect. Immun. 82:2255-2265.

Newton, H.J., J.S. Pearson, L. Badea, M. Kelly, M. Lucas, G. Holloway, K.M. Wagstaff, M.A. Dunstone, J. Sloan, J.C. Whisstock, J.B. Kaper, R.M. Robins-Browne, D.A. Jans, G. Frankel, A.D. Phillips, B.S. Coulson, and E.L. Hartland. 2010. The Type III Effectors NleE and NleB from Enteropathogenic E. coli and OspZ from Shigella Block Nuclear Translocation of NF-κB p65. PLoS Pathog. 6:e1000898.

Nickles, D., C. Falschlehner, M. Metzig, and M. Boutros. 2012. A genome-wide RNA interference screen identifies caspase 4 as a factor required for tumor necrosis factor alpha signaling. Mol. Cell. Biol. 32:3372-3381.

Nordlander, S., J. Pott, and K.J. Maloy. 2014. NLRC4 expression in intestinal epithelial cells mediates protection against an enteric pathogen. Mucosal Immunol. 7:775-785.

Nougayrede, J.P., and M.S. Donnenberg. 2004. Enteropathogenic Escherichia coli EspF is targeted to mitochondria and is required to initiate the mitochondrial death pathway. Cell. Microbiol. 6:1097-1111.

Ochoa, T.J., and C.A. Contreras. 2011. Enteropathogenic escherichia coli infection in children. Curr. Opin. Infect. Dis. 24:478-483.

Oficjalska, K., M. Raverdeau, G. Aviello, S.C. Wade, A. Hickey, K.M. Sheehan, S.C. Corr, E.W. Kay, L.A. O'Neill, K.H. Mills, and E.M. Creagh. 2015. Protective role for caspase-11 during acute experimental murine colitis. J. Immunol. 194:1252-1260.

Olsen R.L., F. Echtenkamp, D. Cheranova, W. Deng, B.B. Finlay and P.R Hardwidge. 2013. The enterohemorrhagic Escherichia coli effector protein NleF binds mammalian Tmp21. Vet. Microbiol. 164(1–2):164-70.

Pallen, M.J., S.A. Beatson, and C.M. Bailey. 2005. Bioinformatics analysis of the locus for enterocyte effacement provides novel insights into type-III secretion. BMC Microbiol. 5:9.

Page 181: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

181

Pallett, M.A., C.N. Berger, J.S. Pearson, E.L. Hartland, and G. Frankel. 2014. The type III secretion effector NleF of enteropathogenic Escherichia coli activates NF-kappaB early during infection. Infect. Immun. 82:4878-4888.

Patel, S.K., J. Dotson, K.P. Allen, and J.M. Fleckenstein. 2004. Identification and molecular characterization of EatA, an autotransporter protein of enterotoxigenic Escherichia coli. Infect. Immun. 72:1786-1794.

Petty, N.K., R. Bulgin, V.F. Crepin, A.M. Cerdeno-Tarraga, G.N. Schroeder, M.A. Quail, N. Lennard, C. Corton, A. Barron, L. Clark, A.L. Toribio, J. Parkhill, G. Dougan, G. Frankel, and N.R. Thomson. 2010. The Citrobacter rodentium genome sequence reveals convergent evolution with human pathogenic Escherichia coli. J. Bacteriol. 192:525-538.

Puren, A.J., G. Fantuzzi, and C.A. Dinarello. 1999. Gene expression, synthesis, and secretion of interleukin 18 and interleukin 1beta are differentially regulated in human blood mononuclear cells and mouse spleen cells. Proc. Natl. Acad. Sci. U. S. A. 96:2256-2261.

Radian, A.D., L. de Almeida, A. Dorfleutner, and C. Stehlik. 2013. NLRP7 and related inflammasome activating pattern recognition receptors and their function in host defense and disease. Microbes Infect. 15:630-639.

Rastelli, G., W. Sirawaraporn, P. Sompornpisut, T. Vilaivan, S. Kamchonwongpaisan, R. Quarrell, G. Lowe, Y. Thebtaranonth, and Y. Yuthavong. 2000. Interaction of pyrimethamine, cycloguanil, WR99210 and their analogues with Plasmodium falciparum dihydrofolate reductase: structural basis of antifolate resistance. Bioorg. Med. Chem. 8:1117-1128.

Rathinam, V.A., S.K. Vanaja, L. Waggoner, A. Sokolovska, C. Becker, L.M. Stuart, J.M. Leong, and K.A. Fitzgerald. 2012. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell 150:606-619.

Raymond, B., V.F. Crepin, J.W. Collins, and G. Frankel. 2011. The WxxxE effector EspT triggers expression of immune mediators in an Erk/JNK and NF-κB-dependent manner. Cell. Microbiol. 13:1881-1893.

Raymond, B., J.C. Young, M. Pallett, R.G. Endres, A. Clements, and G. Frankel. 2013. Subversion of trafficking, apoptosis, and innate immunity by type III secretion system effectors. Trends Microbiol. 21:430-441.

Rider, P., Y. Carmi, O. Guttman, A. Braiman, I. Cohen, E. Voronov, M.R. White, C.A. Dinarello, and R.N. Apte. 2011. IL-1alpha and IL-1beta recruit different myeloid cells and promote different stages of sterile inflammation. J. Immunol. 187:4835-4843.

Roschitzki-Voser, H., T. Schroeder, E.D. Lenherr, F. Frolich, A. Schweizer, M. Donepudi, R. Ganesan, P.R. Mittl, A. Baici, and M.G. Grutter. 2012. Human caspases in vitro: expression, purification and kinetic characterization. Protein Expr. Purif. 84:236-246.

Page 182: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

182

Rothbaum, R.J., J.C. Partin, K. Saalfield, and A.J. McAdams. 1983. An ultrastructural study of enteropathogenic Escherichia coli infection in human infants. Ultrastruct. Pathol. 4:291-304.

Ruiz-Perez, F., and J.P. Nataro. 2014. Bacterial serine proteases secreted by the autotransporter pathway: classification, specificity, and role in virulence. Cell Mol. Life Sci. 71:745-770.

Sakkejha, H., L. Byrne, A.J. Lawson, and C. Jenkins. 2013. An update on the microbiology and epidemiology of enteropathogenic Escherichia coli in England 2010-2012. J. Med. Microbiol. 62:1531-1534.

Saldana, Z., A.L. Erdem, S. Schuller, I.N. Okeke, M. Lucas, A. Sivananthan, A.D. Phillips, J.B. Kaper, J.L. Puente, and J.A. Giron. 2009. The Escherichia coli common pilus and the bundle-forming pilus act in concert during the formation of localized adherence by enteropathogenic E. coli. J. Bacteriol. 191:3451-3461.

Samba-Louaka, A., J.P. Nougayrede, C. Watrin, E. Oswald, and F. Taieb. 2009. The enteropathogenic Escherichia coli effector Cif induces delayed apoptosis in epithelial cells. Infect. Immun. 77:5471-5477.

Savkovic, S.D., A. Koutsouris, and G. Hecht. 1997. Activation of NF-κB in intestinal epithelial cells by enteropathogenic Escherichia coli. Am. J. Physiol. 273:C1160-C1167.

Savkovic, S.D., A. Ramaswamy, A. Koutsouris, and G. Hecht. 2001. EPEC-activated ERK1/2 participate in inflammatory response but not tight junction barrier disruption. Am. J. Physiol. Gastrointest. Liver Physiol. 281:G890-G898.

Schauer, D.B., and S. Falkow. 1993. Attaching and effacing locus of a Citrobacter freundii biotype that causes transmissible murine colonic hyperplasia. Infect. Immun. 61:2486-2492.

Schlosser-Silverman, E., M. Elgrably-Weiss, I. Rosenshine, R. Kohen, and S. Altuvia. 2000. Characterization of Escherichia coli DNA lesions generated within J774 macrophages. J. Bacteriol. 182:5225-5230.

Schroder, K., and J. Tschopp. 2010. The inflammasomes. Cell 140:821-832.

Schuller, S., M. Lucas, J.B. Kaper, J.A. Giron, and A.D. Phillips. 2009. The ex vivo response of human intestinal mucosa to enteropathogenic Escherichia coli infection. Cell. Microbiol. 11:521-530.

Schweitzer, B.I., A.P. Dicker, and J.R. Bertino. 1990. Dihydrofolate reductase as a therapeutic target. Faseb j. 4:2441-2452.

Scott, A.M., and M. Saleh. 2007. The inflammatory caspases: guardians against infections and sepsis. Cell Death Differ. 14:23-31.

Page 183: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

183

Servin, A.L. 2005. Pathogenesis of Afa/Dr diffusely adhering Escherichia coli. Clin. Microbiol. Rev. 18:264-292.

Sham, H.P., S.R. Shames, M.A. Croxen, C. Ma, J.M. Chan, M.A. Khan, M.E. Wickham, W. Deng, B.B. Finlay, and B.A. Vallance. 2011. Attaching and effacing bacterial effector NleC suppresses epithelial inflammatory responses by inhibiting NF-kappaB and p38 mitogen-activated protein kinase activation. Infect. Immun. 79:3552-3562.

Shenoy, A.R., D.A. Wellington, P. Kumar, H. Kassa, C.J. Booth, P. Cresswell, and J.D. MacMicking. 2012. GBP5 promotes NLRP3 inflammasome assembly and immunity in mammals. Science 336:481-485.

Shi, J., Y. Zhao, Y. Wang, W. Gao, J. Ding, P. Li, L. Hu, and F. Shao. 2014. Inflammatory caspases are innate immune receptors for intracellular LPS. Nat. 514:187-192.

Shin, S., M.P. Castanie-Cornet, J.W. Foster, J.A. Crawford, C. Brinkley, and J.B. Kaper. 2001. An activator of glutamate decarboxylase genes regulates the expression of enteropathogenic Escherichia coli virulence genes through control of the plasmid-encoded regulator, Per. Mol. Microbiol. 41:1133-1150.

Sircili, M.P., M. Walters, L.R. Trabulsi, and V. Sperandio. 2004. Modulation of enteropathogenic Escherichia coli virulence by quorum sensing. Infect. Immun. 72:2329-2337.

Sollberger, G., G.E. Strittmatter, M. Kistowska, L.E. French, and H.D. Beer. 2012. Caspase-4 is required for activation of inflammasomes. J. Immunol. 188:1992-2000.

Song-Zhao, G.X., N. Srinivasan, J. Pott, D. Baban, G. Frankel, and K.J. Maloy. 2014. Nlrp3 activation in the intestinal epithelium protects against a mucosal pathogen. Mucosal Immunol. 7:763-774.

Spehlmann, M.E., S.M. Dann, P. Hruz, E. Hanson, D.F. McCole, and L. Eckmann. 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.

Sperandio, V., A.G. Torres, B. Jarvis, J.P. Nataro, and J.B. Kaper. 2003. Bacteria-host communication: the language of hormones. Proc. Natl. Acad. Sci. U. S. A. 100:8951-8956.

Tahoun, A., G. Siszler, K. Spears, S. McAteer, J. Tree, E. Paxton, T.L. Gillespie, I. Martinez-Argudo, M.A. Jepson, D.J. Shaw, M. Koegl, J. Haas, D.L. Gally, and A. Mahajan. 2011. Comparative analysis of EspF variants in inhibition of Escherichia coli phagocytosis by macrophages and inhibition of E. coli translocation through human- and bovine-derived M cells. Infect. Immun. 79:4716-4729.

Tobe, T., S.A. Beatson, H. Taniguchi, H. Abe, C.M. Bailey, A. Fivian, R. Younis, S. Matthews, O. Marches, G. Frankel, T. Hayashi, and M.J. Pallen. 2006. An extensive

Page 184: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

184

repertoire of type III secretion effectors in Escherichia coli O157 and the role of lambdoid phages in their dissemination. Proc. Natl. Acad. Sci. U. S. A. 103:14941-14946.

Tree, J.J., E.B. Wolfson, D. Wang, A.J. Roe, and D.L. Gally. 2009. Controlling injection: regulation of type III secretion in enterohaemorrhagic Escherichia coli. Trends Microbiol. 17:361-370.

Tseng, T.T., B.M. Tyler, and J.C. Setubal. 2009. Protein secretion systems in bacterial-host associations, and their description in the Gene Ontology. BMC Microbiol. 9 Suppl 1:S2-2180-9-S1-S2.

van der Meer, J.W. 1988. The effects of recombinant interleukin-1 and recombinant tumor necrosis factor on non-specific resistance to infection. Biotherapy 1:19-25.

Vandermark, E.R., K.A. Deluca, C.R. Gardner, D.F. Marker, C.N. Schreiner, D.A. Strickland, K.M. Wilton, S. Mondal, and C.D. Woodworth. 2012. Human papillomavirus type 16 E6 and E 7 proteins alter NF-kB in cultured cervical epithelial cells and inhibition of NF-kB promotes cell growth and immortalization. Virology 425:53-60.

Verri, W.A.,Jr, T.M. Cunha, S.H. Ferreira, X. Wei, B.P. Leung, A. Fraser, I.B. McInnes, F.Y. Liew, and F.Q. Cunha. 2007. IL-15 mediates antigen-induced neutrophil migration by triggering IL-18 production. Eur. J. Immunol. 37:3373-3380.

Vidal, J.E., and F. Navarro-Garcia. 2008. EspC translocation into epithelial cells by enteropathogenic Escherichia coli requires a concerted participation of type V and III secretion systems. Cell. Microbiol. 10:1975-1986.

Vijayakumar, V., A. Santiago, R. Smith, M. Smith, R.M. Robins-Browne, J.P. Nataro, and F. Ruiz-Perez. 2014. Role of class 1 serine protease autotransporter in the pathogenesis of Citrobacter rodentium colitis. Infect. Immun. 82:2626-2636.

Vladimer, G.I., R. Marty-Roix, S. Ghosh, D. Weng, and E. Lien. 2013. Inflammasomes and host defenses against bacterial infections. Curr. Opin. Microbiol. 16:23-31.

Vossenkämper, A., O. Marchès, P.D. Fairclough, G. Warnes, A.J. Stagg, J.O. Lindsay, P.C. Evans, L.A. Luong, N.M. Croft, S. Naik, G. Frankel, and T.T. MacDonald. 2010. Inhibition of NF-κB Signaling in Human Dendritic Cells by the Enteropathogenic Escherichia coli Effector Protein NleE. J. Immunol. 185:4118-4127.

Wang, S., M. Miura, Y.K. Jung, H. Zhu, E. Li, and J. Yuan. 1998. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell 92:501-509.

Weintraub, A. 2007. Enteroaggregative Escherichia coli: epidemiology, virulence and detection. J.Med. Microbiol. 56:4-8.

Page 185: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

185

Weiss, A., and J. Brockmeyer. 2012. Prevalence, biogenesis, and functionality of the serine protease autotransporter EspP. Toxins 5:25-48.

Wiles, S., S. Clare, J. Harker, A. Huett, D. Young, G. Dougan, and G. Frankel. 2004. Organ specificity, colonization and clearance dynamics in vivo following oral challenges with the murine pathogen Citrobacter rodentium. Cell. Microbiol. 6:963-972.

Wlodarska, M., C.A. Thaiss, R. Nowarski, J. Henao-Mejia, J.P. Zhang, E.M. Brown, G. Frankel, M. Levy, M.N. Katz, W.M. Philbrick, E. Elinav, B.B. Finlay, and R.A. Flavell. 2014. NLRP6 inflammasome orchestrates the colonic host-microbial interface by regulating goblet cell mucus secretion. Cell 156:1045-1059.

Wong Fok Lung, T., J.S. Pearson, R. Schuelein, and E.L. Hartland. 2014. The cell death response to enteropathogenic Escherichia coli infection. Cell. Microbiol. 16:1736-1745.

Wong, A.R.C., J.S. Pearson, M.D. Bright, D. Munera, K.S. Robinson, S.F. Lee, G. Frankel, and E.L. Hartland. 2011. Enteropathogenic and enterohaemorrhagic Escherichia coli: even more subversive elements. Mol. Microbiol. 80:1420-1438.

Wong, A.R., A. Clements, B. Raymond, V.F. Crepin, and G. Frankel. 2012. The interplay between the Escherichia coli Rho guanine nucleotide exchange factor effectors and the mammalian RhoGEF inhibitor EspH. Mbio. 3:10.1128/mBio.00250-11. Print 2012.

Xie, P. 2013. TRAF molecules in cell signaling and in human diseases. J. Mol. Signal. 8:7-2187-8-7.

Yang, J., Y. Zhao, J. Shi, and F. Shao. 2013. Human NAIP and mouse NAIP1 recognize bacterial type III secretion needle protein for inflammasome activation. Proc. Natl. Acad. Sci. U. S. A. 110:14408-14413.

Yona-Nadler, C., T. Umanski, S. Aizawa, D. Friedberg, and I. Rosenshine. 2003. Integration host factor (IHF) mediates repression of flagella in enteropathogenic and enterohaemorrhagic Escherichia coli. Microbiol. 149:877-884.

Yoneyama, M., and T. Fujita. 2007. Cytoplasmic double-stranded DNA sensor. Nat. Immunol. 8:907-908.

Zhang, L., X. Ding, J. Cui, H. Xu, J. Chen, Y. Gong, L. Hu, Y. Zhou, J. Ge, Q. Lu, S. Chen, and F. Shao. 2011. Cysteine methylation disrupts ubiquitin-chain sensing in NF-κB activation. Nat. 481:204-208.

Zhang, Q., Q. Li, C. Wang, N. Li, and J. Li. 2012. Redistribution of tight junction proteins during EPEC infection in vivo. Inflammation 35:23-32.

Zhang, Y., C. Laing, M. Steele, K. Ziebell, R. Johnson, A.K. Benson, E. Taboada, and V.P. Gannon. 2007. Genome evolution in major Escherichia coli O157:H7 lineages. BMC Genomics 8:121.

Page 186: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

186

Zhao, Y., J. Yang, J. Shi, Y.N. Gong, Q. Lu, H. Xu, L. Liu, and F. Shao. 2011. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nat. 477:596-600.

Zhou, M., Z. Guo, Q. Duan, P.R. Hardwidge, and G. Zhu. 2014. Escherichia coli type III secretion system 2: a new kind of T3SS? Vet. Res. 45:32-9716-45-32.

Page 187: Characterisation of the type III secretion effector NleF › bitstream › 10044 › 1 › ...effectors into the host cytosol to subvert host cell signalling including: actin dynamics,

187

Permissions for use of unpublished data