regulation of metalloproteinase-dependent ectodomain ......regulation of metalloproteinase-dependent...

219
Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation by Aditya K. Murthy A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Medical Biophysics University of Toronto © Copyright by Aditya K. Murthy 2011

Upload: others

Post on 05-Oct-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

Regulation of metalloproteinase-dependent ectodomain shedding in

cytokine biology and inflammation

by

Aditya K. Murthy

A thesis submitted in conformity with the requirements for the degree of

Doctor of Philosophy

Graduate Department of Medical Biophysics

University of Toronto

© Copyright by Aditya K. Murthy 2011

Page 2: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

ii

Abstract

Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology

and inflammation.

Aditya Murthy, Doctor of Philosophy 2011. Department of Medical Biophysics,

University of Toronto.

In 1962, Gross and Lapiere described collagenolytic activity in the degradation of

tadpole tails during amphibian metamorphosis. This activity was later attributed to a

collagenase enzyme belonging to the matrix metalloproteinase family. Over the past 49

years, steady growth in the field of metalloproteinase biology has uncovered that

degradation of extracellular matrix components represents only a fraction of the functions

performed by these enzymes. The regulatory roles of these enzymes in numerous aspects

of mammalian biology remains poorly understood.

This thesis investigates the metalloproteinase ADAM17 and its natural inhibitor

TIMP3 in acute and chronic inflammation. My work describes the generation of new

murine experimental systems of compartmentalized ADAM17 or TIMP3 deficiency and

their applications in acute liver inflammation (i.e. fulminant hepatitis and T-cell mediated

autoimmune hepatitis) and atopic dermatitis. Loss of Timp3 protected mice against

fulminant hepatic failure caused by activation of the death receptor Fas. We determined

that TIMP3 simultaneously promotes pro-apoptotic signaling through TNFR1 while

suppressing anti-apoptotic EGFR activation in the liver. Mechanistically, we identified

Page 3: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

iii

that ADAM17 is critical in shedding TNFR1 and EGFR ligands (e.g. Amphiregulin, HB-

EGF, TGF) and extended this finding to clinically relevant drug-induced hepatitis.

Adult TIMP3 deficient mice also exhibited spontaneous accumulation of CD4+ T

cells in the liver. Consequently, polyclonal T cell activation with the lectin Concanavalin

A (con A) in a model of autoimmune hepatitis resulted in accelerated liver injury. We

identified that this immunopathology relied on TNF bioavailability as mice lacking both

Timp3 and Tnf were resistant to con A. Using bone marrow chimeras we established that

non-hematopoietic tissues were the physiologically relevant source of TIMP3 in vivo,

thereby highlighting an immunosuppressive role for this stromal metalloproteinase

inhibitor in cellular immunity.

Finally, we investigated epithelial:immune crosstalk in the epidermis by

generating tissue-specific ADAM17 deficiency in basal keratinocytes. These mice

developed spontaneous inflammatory skin disease that was physiologically consistent

with atopic dermatitis. Focused investigation of keratinocyte-specific signaling

deregulated by ADAM17 deficiency revealed its requirement for tonic Notch activation,

which in turn antagonized transcriptional activity of AP-1 transcription factors on the

promoters of epithelial cytokines TSLP and G-CSF.

In summary, these works identify cellular mechanisms governing cytokine-

mediated communication between epithelial and immune cells to modulate inflammation.

The findings that TIMP3 and ADAM17 act as regulators of key inflammatory,

proliferative and developmental pathways provide impetus to expand our understanding

of this important family of enzymes in mammalian signal transduction.

Page 4: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

iv

Acknowledgements

My most sincere thanks go to my supervisor Dr. Rama Khokha. I could not have

asked for a more ideal environment to learn and grow as a student. You have consistently

been an example of a true mentor, and your genuine curiosity, sincerity, openness and

camaraderie are qualities I hope to emulate throughout my career. Thank you. I would

like to acknowledge my committee members Dr. Razq Hakem and Dr. Rob Rottapel. Our

meetings were always challenging and rewarding. Your critiques and advice pushed my

inquiries to the next level, and for that I am grateful. A special thanks to Dr. Juan Carlos

Zúñiga-Pflücker - you sparked my budding interest in immunology, and have gone above

and beyond to introduce me to new avenues in the field. To my labmates and colleagues,

I thank you for your friendship and constant support over the last six years.

I also thank my family – Ma, Baba, Bhaskar, Farhana and the Berghoefs. You

have been examples for me in more ways than you can imagine. Your never-ending

support and interest in my work has made this thesis a success.

To my partner Naomi, I can confidently say that none of this would be possible

without your presence in my life. You encouraged me and fed my scientific curiosity

from the day I started this journey. I look forward to many more journeys to come.

Page 5: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

v

1 Table of Contents

Abstract .............................................................................................................................. ii

Acknowledgements .......................................................................................................... iv

Table of Contents .............................................................................................................. v

List of Figures and Tables ............................................................................................... xi

List of Abbreviations ..................................................................................................... xvi

List of Publications ......................................................................................................... xx

CHAPTER 1 ...................................................................................................................... 1

1.1 Ectodomain shedding ........................................................................................... 2

1.1.1 Tissue Inhibitors of Metalloproteinases (TIMPs) ..................................... 3

1.1.1.1 The structure and evolution of TIMPs ................................................... 4

1.1.1.2 Targets of TIMPs and phenotypes of Timp deficiency ......................... 6

1.1.1.3 TIMP1 .................................................................................................. 10

1.1.1.4 TIMP2 .................................................................................................. 12

1.1.1.5 TIMP3 .................................................................................................. 12

1.1.1.6 TIMP4 .................................................................................................. 15

1.1.2 Summary ................................................................................................. 15

1.1.3 Matrix Metalloproteinases (MMPs) ........................................................ 16

1.1.4 Disintegrin and Metalloproteinases (ADAMs) ....................................... 18

1.1.4.1 ADAMs in receptor crosstalk .............................................................. 19

1.1.4.2 ADAMs in receptor signaling cascades............................................... 20

1.1.4.3 ADAM10 ............................................................................................. 21

1.1.4.4 ADAM17 ............................................................................................. 22

Page 6: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

vi

1.2 Ectodomain shedding in physiological processes .............................................. 23

1.2.1 Cell proliferation ..................................................................................... 23

1.2.2 Apoptosis ................................................................................................. 25

1.2.3 Ectodomain shedding in immunity ......................................................... 26

1.2.3.1 Metalloproteinases facilitate lymphocyte development and

proliferation............................................................................................................ 28

1.2.3.2 Myeloid and lymphoid cell migration is dependent on

metalloproteinase activity ...................................................................................... 33

1.2.3.3 Shedding of cell surface molecules regulates humoral and cell based

immune effector function ....................................................................................... 36

1.2.4 Summary ................................................................................................. 40

1.3 Thesis outline ..................................................................................................... 43

1.3.1 Study Rationale ....................................................................................... 43

1.3.2 Thesis Objectives .................................................................................... 44

CHAPTER 2 .................................................................................................................... 45

2.1 Abstract .............................................................................................................. 46

2.2 Introduction ........................................................................................................ 47

2.3 Results ................................................................................................................ 49

2.3.1 TNF signaling sensitizes hepatocytes to Fas-mediated apoptosis ........... 49

2.3.2 Delay of Fas-induced apoptosis in Timp3−/−

livers ................................. 52

2.3.3 TNFR1 shedding dampens JNK phosphorylation and NF-B activation in

Timp3−/−

liver ............................................................................................................ 55

2.3.4 Signaling through AKT or AMPK does not contribute to

hepatoprotection in Timp3−/−

mice ............................................................................ 62

2.3.5 Compound deletions of Timp3/Tnf or Timp3/Tnfr1 completely prevent

hepatic failure and involve enhanced ERK1/2 phosphorylation ............................... 62

Page 7: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

vii

2.3.6 TIMP3 inhibits metalloproteinase-dependent EGFR signaling .............. 66

2.3.7 Increased EGFR ligand shedding is hepatoprotective............................. 67

2.3.8 Hepatocyte-specific loss of ADAM17 or EGFR promotes Fas-induced

killing ................................................................................................................. 69

2.3.9 Inhibitors of MAPK, EGFR or ADAM17 reverse the Timp3−/−

resistance

to Fas ................................................................................................................. 70

2.3.10 Adenoviral ADAM17 prevents acute liver failure in drug-induced

toxicity ................................................................................................................. 74

2.4 Discussion .......................................................................................................... 76

2.4.1 Ectodomain shedding of TNF is a crucial step in hepatotoxicity ........... 77

2.4.2 TIMP3 regulates pro-survival and pro-apoptotic signaling in hepatocytes

................................................................................................................. 78

2.5 Methods .............................................................................................................. 83

2.5.1 Mice ......................................................................................................... 83

2.5.2 Fas-induced hepatotoxicity ..................................................................... 83

2.5.3 APAP-induced hepatotoxicity and adenoviral delivery of ADAM17 .... 84

2.5.4 Primary hepatocyte culture and apoptosis assays ................................... 84

2.5.5 LPA treatment of MEFs .......................................................................... 85

2.5.6 Immunoblotting ....................................................................................... 86

2.5.7 Caspase activity assays............................................................................ 87

2.5.8 Electrophoretic Mobility Shift Assay (EMSA) ....................................... 87

2.5.9 Enzyme-Linked ImmunoSorbent Assay (ELISA) .................................. 88

2.5.10 Histology & Immunohistochemistry ....................................................... 88

2.5.11 RNA preparation and quantitative RT-PCR............................................ 89

2.5.12 Statistical Analyses ................................................................................. 89

Page 8: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

viii

CHAPTER 3 .................................................................................................................... 91

3.1 Abstract .............................................................................................................. 92

3.2 Introduction ........................................................................................................ 92

3.3 Results ................................................................................................................ 94

3.3.1 A basal increase in CD4+ T cell and NKT cell populations in Timp3

−/−

livers ................................................................................................................. 94

3.3.2 TIMP3 deficiency sensitizes mice to T-cell mediated hepatitis induced by

concanavalin A .......................................................................................................... 96

3.3.3 Enhanced TNF signaling and Th1 cytokine response drives liver damage

in Timp3−/−

mice ........................................................................................................ 97

3.3.4 Cell-intrinsic TIMP3 is not required for CD4+ T cell activation .......... 103

3.3.5 Stromal TIMP3 protects against con A-induced hepatitis .................... 103

3.4 Discussion ........................................................................................................ 105

3.5 Methods ............................................................................................................ 109

3.5.1 Mice ....................................................................................................... 109

3.5.2 Induction of hepatitis & generation of bone marrow chimeras ............. 109

3.5.3 CD4+ T cell culture ............................................................................... 109

3.5.4 Serum analysis....................................................................................... 110

3.5.5 Histology & Immunoblotting ................................................................ 110

3.5.6 Flow cytometry ..................................................................................... 111

3.5.7 Cell culture ............................................................................................ 111

3.5.8 Immunoblotting ..................................................................................... 112

3.5.9 RNA preparation and quantitative RT-PCR.......................................... 112

3.5.10 Statistical Analyses ............................................................................... 113

Page 9: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

ix

CHAPTER 4 .................................................................................................................. 115

4.1 Abstract ............................................................................................................ 116

4.2 Introduction ...................................................................................................... 116

4.3 Results .............................................................................................................. 119

4.3.1 Onset of spontaneous atopic dermatitis upon deletion of epidermal

Adam17 ............................................................................................................... 119

4.3.2 Th2-polarized cellular immunity in skin-draining lymph nodes and

myeloproliferative disease in bone marrow of Adam17ep

mice ............................. 121

4.3.3 Precocious differentiation of ADAM17 deficient keratinocytes in vivo

and in vitro ............................................................................................................... 122

4.3.4 Inducible deletion of epidermal Adam17 in adult mice recapitulates

atopic dermatitis and myeloproliferative disease .................................................... 124

4.3.5 TNFR1 signaling is dispensable for atopic dermatitis and

myeloproliferative disease in Adam17ep

mice ....................................................... 130

4.3.6 Loss of Adam17 compromises tonic Notch signaling in the epidermis 133

4.3.7 Active Notch antagonizes c-Fos/AP-1 transcription of Tslp in human

keratinocytes ............................................................................................................ 136

4.3.8 Ectopic Notch rescues the enhanced AP-1 driven stress response in

ADAM17 deficient keratinocytes ............................................................................ 139

4.3.9 Adenoviral delivery of active Notch rescues local Th2 immunity and

myeloproliferation in Adam17ep

mice ................................................................... 142

4.4 Discussion ........................................................................................................ 143

4.4.1 Contexts of ADAM10 and ADAM17 function in Notch signaling ...... 146

4.5 Methods ............................................................................................................ 148

4.5.1 Mice ....................................................................................................... 148

4.5.2 Microarray Gene Expression Analysis .................................................. 148

Page 10: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

x

4.5.3 In vivo delivery of active Notch to rescue epidermal inflammation ..... 149

4.5.4 Isolation and culture of primary keratinocytes ...................................... 149

4.5.5 AP-1 luciferase assay and PMA treatment of mouse embryonic

fibroblasts (MEFs) ................................................................................................... 150

4.5.6 Culture and treatment of HaCaT cells and chromatin

immunoprecipitation (ChIP) .................................................................................... 151

4.5.7 Flow cytometry ..................................................................................... 151

4.5.8 Histology & cytokine analysis .............................................................. 152

4.5.9 Immunoblotting and AP-1 binding assay .............................................. 152

4.5.10 RNA preparation and quantitative RT-PCR.......................................... 153

4.5.11 Statistical Analyses ............................................................................... 153

CHAPTER 5 .................................................................................................................. 156

5.1 Manipulating ADAM17 and TIMPs in models of adult skin inflammation .... 159

5.1.1 Antigen-specific atopic dermatitis ........................................................ 159

5.1.2 Mechanical cutaneous injury ................................................................. 163

5.2 Redundancy of TIMPs in hematopoietic niches .............................................. 164

5.2.1 TIMPs in the thymic stroma .................................................................. 164

5.2.1.1 Cell autonomous and stromal effects of Timps on T lymphopoiesis 164

5.2.1.2 Mechanisms underlying altered T cell development in Timp or

Adam17 null mice ................................................................................................ 170

5.3 Concluding Remarks ........................................................................................ 174

References ...................................................................................................................... 175

Page 11: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xi

List of Figures and Tables

Figure 1.1 – Structure and evolution of Tissue Inhibitors of Metalloproteinases (TIMP) . 7

Figure 1.2The processes of clipping, shedding and RIPping ....................................... 29

Figure 1.3Cell surface sheddases influence adaptive immunity. .................................. 32

Figure 1.4 – Localization of metalloproteinases and their substrates impacting immunity.

........................................................................................................................................... 42

Figure 2.1 Ablation of TNF signaling delays Fas-mediated hepatotoxicity. ................. 51

Figure 2.2 – Caspase 3/7 activation in TNF and TNFR1 deficient hepatocytes, hepatic

Adam and Mmp expression following Jo-2 administration, and decreased caspase

activation in Timp3

mice. ............................................................................................. 53

Figure 2.3 Histology of liver damage upon Jo-2 treatment across all genotypes. ......... 54

Figure 2.4Delay of Fas-mediated apoptosis and hepatotoxicity in Timp3 −/−

mice. .... 56

Figure 2.5 – Comparable Fas expression in WT and T3

liver....................................... 57

Figure 2.6 – PARP cleavage in primary hepatocytes, Adam17, Tnfr1 and Tnfr2

expression in liver following Jo-2 treatment. ................................................................... 58

Figure 2.7Decreased hepatocyte apoptosis correlates with enhanced TNFR1 shedding

and abrogated TNF signaling upon loss of Timp3............................................................ 60

Figure 2.8Comparable AKT and AMPKβ phosphorylation in WT and T3−/−

livers upon

induction of Fas mediated toxicity. ................................................................................... 63

Figure 2.9Compound loss of Timp3 and Tnf or Tnfr1 completely prevents Fas-

mediated hepatotoxicity and reveals accelerated ERK1/2 phosphorylation. .................... 64

Page 12: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xii

Figure 2.10Loss of Timp3 enhances hepatoprotective EGFR ligand shedding and

downstream ERK1/2 phosphorylation. ............................................................................. 68

Figure 2.11 – Enhanced EGFR phosphorylation in T3

MEFS, increased EGFR target

gene expression in T3

livers. ........................................................................................ 71

Figure 2.12ADAM17 and EGFR are individually hepatoprotective against Fas-

mediated hepatotoxicity. ................................................................................................... 72

Figure 2.13TIMP3 is an upstream regulator of EGFR and MAPK activity in Fas-

induced hepatotoxocity. ................................................................................................... 73

Figure 2.14 Ectopic ADAM17 delivery protects against acetaminophen-driven

fulminant hepatitis. ........................................................................................................... 75

Figure 2.15Inhibition of cell surface ADAM17 impacts MAP kinases during stress

responses and provides stromal regulation of cell survival. ............................................. 80

Figure 3.1 Timp3−/−

mice exhibit spontaneous accumulation and activation of hepatic

CD4+ T cells and NKT cells, and enhanced sensitization to con A-induced hepatitis. .... 95

Figure 3.2Enhanced necrosis, lymphocyte infiltration and macrophage activity in

Timp3−/−

livers. ................................................................................................................. 98

Figure 3.3 Loss of Timp3 enhances TNF signaling and Th1 cytokine response during

autoimmune hepatitis. ....................................................................................................... 99

Figure 3.4Timp3−/−

;Tnf −/−

compound deletion elevates serum IL-10, IL-17A and

depletes splenic and hepatic CD4+ T cells. ..................................................................... 101

Page 13: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xiii

Figure 3.5 – TIMP3 is dispensable for cell-autonomous activation and proliferation of

CD4+ T cells. ................................................................................................................... 102

Figure 3.6Stromal TIMP3 protects against a Th1 pro-inflammatory cyokine response

and autoimmune hepatitis. .............................................................................................. 104

Figure 4.1Epidermal ADAM17 deficiency causes atopic dermatitis (AD). ............... 120

Figure 4.2Skin-specific Th2 lymphocyte activation and myeloproliferative disease

(MPD) in Adam17ep

mice. ............................................................................................. 123

Figure 4.3Accelerated differentiation of Adam17ep

keratinocytes. ........................... 125

Figure 4.4Inducible deletion of epidermal Adam17 in adult mice recapitulates atopic

dermatitis and skin-specific Th2 lymphocyte activation. ............................................... 128

Figure 4.5 – Onset of MPD upon tamoxifen-induced deletion of epidermal Adam17 in

adult mice. ....................................................................................................................... 129

Figure 4.6 – Tnfr1 deletion does not rescue AD in Adam17ep

mice. ............................ 131

Figure 4.7 – MPD persists in Adam17ep

;Tnfr1

double deficient mice. ..................... 132

Figure 4.8ADAM17 provides ligand-independent Notch signaling and suppresses c-

Fos activity in keratinocytes. .......................................................................................... 134

Figure 4.9Stratification of atopic dermatitis, atopic eczema and psoriasis patients based

on expression analysis of genes involved in Notch signaling......................................... 135

Figure 4.10ADAM17 activates Notch to suppress c-Fos transcriptional activity on

cytokine promoters.......................................................................................................... 138

Page 14: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xiv

Figure 4.11Diminished Notch activation upon ADAM17 deficiency enhances c-

Fos/AP-1 driven stress signaling in keratinocytes. ......................................................... 140

Figure 4.12AdCre-deletion of Adam17 in keratinocytes enhances Csf2 gene expression

in an AP-1 dependent manner but does not impact c-Jun, JunB, JunD, Fra1 or Il33

expression. ...................................................................................................................... 141

Figure 4.13Ectopic Notch activity rescues atopic dermatitis and myeloproliferative

disease in Adam17ep

mice. ............................................................................................. 144

Figure 4.14 – An illustration of epithelial-immune crosstalk as directed by the

ADAM17/Notch/c-Fos triad. .......................................................................................... 145

Figure 5.1 – Methods of analyzing antigen-specific responses in atopic dermatitis. ..... 162

Figure 5.2 Expression of Timp genes in bone marrow stromal cell line OP9-DL1

compared to OP9-GFP. ................................................................................................... 166

Figure 5.3 Methods of analyzing hematopoietic vs. epithelial contributions of TIMPs

and ADAM17 in thymopoiesis using fetal thymus organ culture (FTOC). ................... 167

Figure 5.4 – Pharmacological inhibition of ADAM17 activity arrests T cell development

in a dose-dependent manner............................................................................................ 168

Figure 5.5 – Additive loss of Timp genes impacts T cell development with a gene-

dependent decrease in double-negative 1 (DN1) populations. ....................................... 169

Figure 5.6 – Decreased thymic cellularity in K14-Cre;Adam17fl/fl

thymus. ................... 172

Page 15: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xv

Table 1.1 – Profiling TIMP target MMPs and ADAMs, and their downstream substrates.9

Table 1.2 – Murine phenotypes arising from individual Timp knockouts. ...................... 11

Table 1.3TIMP effects on apoptosis and proliferation in vitro .................................... 24

Table 2.1Primer sequences used for quantitative RT-PCR analysis. ........................... 89

Table 3.1 – Primer sequences for quantitative RT-PCR analysis and genotyping. ........ 113

Table 4.1 – Probelists and significance values for differentially expressed genes in all

studies. (See Appendix A online). .................................................................................. 154

Table 4.2Primer sequences used for qRT-PCR and ChIP analysis. ........................... 154

Page 16: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xvi

List of Abbreviations

AD Atopic Dermatitis

ADAM A Disintegrin And Metalloproteinase

ADAM-TS A Disintegrin And Metalloproteinase with Thrombospondin motifs

AICD Activation Induced Cell Death

AKT/PI3K Phosphoinositide 3-Kinase

ALT Alanine Transaminase

AMPK Adenosine Monophosphate-activated Protein Kinase

AP-1 Activating Protein 1

APAP N-acetyl-p-aminophenol (acetaminophen)

APC Antigen Presenting Cell

AR Amphiregulin

AST Aspartate Transaminase

APP Beta Amyloid Precursor Protein

BM Bone Marrow

CBA Cytokine Bead Array

CD Cluster Domain

cDNA complementary Deoxyribonucleic Acid

c-FLIP FLICE Inhibitory Protein

CLL Chronic Lymphocytic Leukemia

Con A Concanavalin A

DAPT N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl

ester

DC Dendritic Cell

DLL Delta Like Ligand

dLN draining Lymph Node

DMSO Dimethyl Sulfoxide

DN Double Negative

DNA Deoxyribonucleic acid

DR Death Receptor

EC Epithelial Cell

ECM Extracellular Matrix

EGFR Epidermal Growth Factor Receptor

ELISA Enzyme-Linked Immunosorbent Assay

EMSA Electrophoretic Mobility Shift Assay

EPA Erythroid Potentiating Activity

ERK Extracellular signal-regulated Kinase

ERp5 Endoplasmic Reticulum Protein 5

ES cell Embryonic Stem Cell

Page 17: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xvii

FACS Flow Assisted Cell Sorting

FADD Fas Assodicated via Death Domain

FAK Focal Adhesion Kinase

FasL Fas Ligand

FGF Fibroblast Growth Factor

FGFR Fibroblast Growth Factor Receptor

FTOC Fetal Thymic Organ Culture

G-CSF Granulocyte Colony Stimulating Factor

GFP Green Fluorescent Protein

GM-CSF Granulocyte Macrophage Colony Stimulating Factor

GPCR G-protein Coupled Receptor

GSK Glycogen Synthase Kinase

HB-EGF Heparin Binding EGFR-like Growth Factor

HCC Hepatocellular Carcinoma

HDMEC Human Dermal Microvascular Endothelial Cells

HEMVEC Human Endometrial Microvascular Endothelial Cell

HGF Hepatocyte Growth Factor

HLA Human Leukocyte Antigen

HMVEC Human Microvascular Endothelial Cells

HSC Hepatic Stellate Cell

HUVEC Human Umbilical Vein Endothelial Cell

IAP Inhibitor of Apoptosis

ICAM Inter-Cellular Adhesion Molecule

IFN Interferon

IGF Insulin-like Growth Factor

IGFBP Insulin-like Growth Factor Binding Protein

IB nuclear factor of Kappa light polypeptide gene enhancer in B cells,

Inhibitor

IKK nuclear factor Kappa B inhibitor kinase beta

IL-(x) Interleukin - (number)

IL-(x)R Interleukin - (number) Receptor

JNK c-Jun N-terminal Kinase

KC Keratinocyte Chemoattractant

LAG Lymphocyte Activation Gene

LPA Oleoyl-L-α-lysophosphatidic acid sodium salt

LPS

LV

Lipopolysaccharide

Left Ventricle

MAML Mastermind Like

MAPK Mitogen Associated Protein Kinase

MCP Monocyte Chemoattractant Protein

Page 18: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xviii

M-CSF Macrophage Colony Stimulating Factor

MEF Mouse Embryonic Fibroblast

MEKK Mitogen Associated Protein Kinase Kinase

MHC Major Histocompatibility Complex

MICA MHC class I chain-related A

microCT Micro Computed Tomography

MIG Monokine Induced by Interferon Gamma

MIP Macrophage Inflammatory Protein

miRNA micro-RNA

mLN mesenteric Lymph Node

MMP Matrix Metalloproteinase

MNC Mononuclear cell

MPD Myeloproliferative Disease

mRNA Messenger Ribonucleic Acid

MT1-MMP Membrane Type 1 Matrix Metalloproteinase

MUC1 Mucin 1, cell surface associated

NECD Notch Extra-Cellular Domain

NEMO NF kappa B Essential Modulator

NF-B Nuclear Factor kappa B

NGFR Nerve Growth Factor Receptor

NICD Notch Intra-Cellular Domain

NK Natural Killer

NKG2D Natural Killer G2D

NKT Natural Killer T cell

OVA Ovalbumin

PARP Poly-ADP Ribose Polymerase

pDC Plasmacytoid Dendritic Cell

PECAM Platelet Endothelial Cell Adhesion Molecule

PI Propidium Iodide

PIP2 Phosphatidylinositol 4,5-bisphosphate

PKC Protein Kinase C

PLC Phospholipase C

PMA phorbol-12-myristate-13-acetate

PMN Polymorphonuclear cell

PTEN Phosphatase and Tensin homolog

qRT-PCR Quantitative Reverse Transcriptase Polymerase Chain Reaction

RA Rheumatoid Arthritis

RANTES Regulated on Activation Normal T cell Expressed and Secreted

RBP-j Recombination Signal-Binding Protein 1 for J-Kappa

RECK Reversion-inducing-cysteine-rich protein with Kazal motifs

Page 19: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xix

RIP Receptor Intramembrane Proteolysis

RNA Ribonucleic Acid

ROS Reactive Oxygen Species

RTK Receptor Tyrosine Kinase

SDF Stromal Derived Factor

SFD Sorsby's Fundus Dystrophy

SHP SH2 domain containing Phosphatase

STAT Signal Transducer and Activator of Transcription

SV40/TAg Simian Virus 40 large T antigen

SVMP Snake Venom Metalloproteinase

TACE TNF Alpha Converting Enzyme

TanIIA Tanshinone II A

TAPI TNF Alpha Protease Inhibitor

TCR T Cell Receptor

TGF Transforming Growth Factor

TH T-helper

TIMP Tissue Inhibitor of Metalloproteinases

TNF Tumor Necrosis Factor

TNFR Tumor Necrosis Factor Receptor

TPA 12-O-tetradecanoylphorbol-13-acetate

TRADD Tumor necrosis factor recetpor type 1 Associated Death Domain

TRAIL Tumor necrosis factor Related Apoptosis Inducing Ligand

TRAILR Tumor necrosis factor Related Apoptosis Inducing Ligand Receptor

TRANCE TNF Related Activation Induced Cytokine

TRE TPA Responsive Element

Treg Regulatory T cell

TSLP Thymic Stormal Lymphoprotein

TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling

TYK Tyrosine Kinase

UV Ultraviolet

VCAM Vascular cell adhesion molecule

VE-cadherin Vascular Endothelial Cadherin

VEGF Vascular Endothelial Growth Factor

VEGFR Vascular Endothelial Growth Factor Receptor

WT Wildtype

Page 20: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

xx

List of Publications

Murthy, A., Cruz-Munoz, W., Khokha, R. (2008). TIMPs: Extracellular Modifiers in

Cancer Development. In The Cancer Degradome. Proteases and Cancer Biology

(pp. 371-401). New York, NY: Springer Science.

Murphy, G., Murthy, A., Khokha, R. (2008). Clipping, shedding and RIPping keep

immunity on cue. Trends in Immunology. Feb;29(2):75-82. Review

Murthy, A., Defamie, V., Smookler, D.S., Di Grappa, M.A., Horiuchi, K., Federici, M.,

Sibilia, M., Blobel, C.P., Khokha, R. (2010). Ectodomain shedding of EGFR

ligands and TNFR1 dictates hepatocyte apoptosis during fulminant hepatitis in

mice. Journal of Clinical Investigation. Aug 2;120(8):2731-44.

Murthy, A., Shao, Y.W., Narala, S.R., Molyneux, S.D., Zúñiga-Pflücker, J.C., Khokha,

R. (2011). ADAM17 activation of epidermal Notch regulates atopic barrier

immunity and myeloproliferation by suppressing c-Fos-driven transcription of

epithelial cytokines. (Under review).

Murthy, A., Shao, Y.W., Wedeles, C., Smookler, D.S., Khokha, R. (2011). Stromal

TIMP3 regulates liver lymphocyte populations and provides protection against

Th1 T-cell driven autoimmune hepatitis. (Submitted).

Page 21: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

1

CHAPTER 1

GENERAL INTRODUCTION

This chapter includes two published reviews:

Murthy, A., W. Cruz-Munoz, and R. Khokha, TIMPs: Extracellular Modifiers in Cancer

Development, in The Cancer Degradome, D. Edwards, et al., Editors. 2008, Springer

New York. p. 373-400.

Murphy, G., A. Murthy, and R. Khokha, Clipping, shedding and RIPping keep immunity

on cue. Trends Immunol, 2008. 29(2): p. 75-82.

Page 22: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

2

1.1 Ectodomain shedding

Identifying how cells communicate, both with each other and the extracellular

microenvironment, remain central questions in biology. It is now recognized that cells

use physical attachment to the extracellular matrix to modulate numerous processes such

as differentiation of stem cells, establishing a polarized identity, promoting cell survival

and migration. Cells also produce and release soluble factors such as cytokines to interact

with each other across larger physiological spaces. Initial studies on the extracellular

matrix identified a process by which cell surface molecules like heparan sulfate

proteoglycans can regulate the binding of a cell to extracellular matrix components. This

process, termed „ectodomain shedding‟, involved enzymatic cleavage of the extracellular

component of heparin sulfate proteoglycan and suggested a mechanism of detaching

epithelial cells to allow for movement during normal biology; it also identified a potential

role for ectodomain shedding in cancer cell invasion [1, 2]. Subsequent studies added

cytokines, growth factors and their receptors as additional targets of ectodomain shedding

[3]. The current landscape of cytokine biology identifies this process as a key step in

allowing autocrine, juxtacrine or systemic signaling. A family of enzymes termed

metalloproteinases harbours the principal proteinases which perform ectodomain

shedding at the cell surface. Their function is controlled by four tissue inhibitors of

metalloproteinses (TIMPs) [4]. Together, the TIMP:metalloproteinase axis directs lines

of communication between cells and the microenvironment during health and disease.

Page 23: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

3

1.1.1 Tissue Inhibitors of Metalloproteinases (TIMPs)

The tissue inhibitor of metalloproteinase (TIMP) family has evolved to regulate

tissue homeostasis through its ability to operate at the stromal:cellular interface. It

controls remodeling of the extracellular matrix (ECM) as well as the cell surface by

inhibiting the activity of several classes of metalloproteinases. TIMPs are ancient proteins

found in invertebrates and vertebrates including nematodes, insects, fish and mammals.

However TIMPs have not been reported in plants despite the known existence of

metalloproteinases in this kingdom [5]. The mammalian genome contains 4 distinct

TIMP proteins, each with different yet overlapping metalloproteinase inhibitory profiles

[6]. The mammalian TIMP is a two-domain protein consisting of N- and C-termini and 6

disulphide linkages, and can exist in glycosylated and unglycosylated forms. Well-known

targets of TIMPs include the enzymes from matrix metalloproteinase (MMPs), a

disintegrin and metalloproteinase (ADAMs) and ADAM with thrombospondin motif

(ADAM-TS) classes. During metalloproteinase inhibition, a single netrin domain forms a

wedge-like structure to interact with the active site of the enzyme forming a 1:1

stoichiometric complex that sterically inhibits metalloproteinase activity [7].

TIMP was independently identified as a collagenase inhibitor [8] and for its

erythroid potentiating activity [9], and additionally murine Timp1 was identified as a cell

cycle responsive gene [10]. It was then discovered that antisense RNA-mediated

downregulation of TIMP1 conferred oncogenic properties on immortal but non-

transformed murine fibroblasts showing a central role for TIMP in tumorigenesis [11].

The systems initially used to examine TIMPs included reproductive biology [12, 13],

Page 24: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

4

wound healing (reviewed in [14]), tumorigenesis and metastasis [11, 15]. The cloning of

all four human and murine Timp genes [9, 16-20], followed by analysis of their

regulatory elements and genetic expression studies [21-25] have revealed a more

complex role in orchestrating tissue homeostasis. Cellular and matrix turnover are both

affected by TIMPs, and each TIMP distinctly influences cell function. Clinical studies

covering diverse human cancers also document the tremendous heterogeneity of TIMP

expression and their correlation with disease stage and prognosis. Furthermore, the

understanding of TIMP function has co-evolved with that of the expanding field of

metalloproteinase biology, and together they have highlighted complementary concepts

essential for the health and survival of the organism.

1.1.1.1 The structure and evolution of TIMPs

Invertebrate and vertebrate TIMPs differ significantly in both structure and

function. The two C. elegans TIMPs are single domain proteins consisting of only the N-

terminal region of mammalian TIMP, whereas the single D. melanogaster TIMP has the

2-domain structure, and functionally resembles the mammalian TIMP3. It is able to

inhibit the activity of D. melanogaster Kuzbanian/ADAM10 along with MMP inhibition

[26, 27] and associates strongly with the ECM via interactions with hyaluronic acid, both

properties unique to the mammalian TIMP3. Until recently, the N-terminal end of TIMP3

was thought to be solely responsible for the TIMP3:ECM binding. However elegant work

by Lee et al [28] has revealed that in fact both N- and C-termini are involved in ECM-

binding. Both termini utilize basic amino acids such as Lysine and Arginine in this

Page 25: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

5

interaction, as exhaustive mutations to specific amino acids at either terminus, together

with domain swapping to generate ECM-adhering TIMP1 clearly identify the 6 amino

acids (N-terminus: Lys-26, 27, 30, and 76; C-terminus: Lys-165 and Arg-163) required

for ECM binding. Figure 1.1A represents a schematic of the prototypical 2-domain

TIMP, with the MMP-inhibitory and ECM-binding residues indicated. As TIMP1 and

TIMP3 are structurally similar, the figure also illustrates the residues of TIMP3 that are

absent in TIMP1. The crystal structure of a full length TIMP has yet to be solved, as only

the N-termini in isolation or complexed with MMP have been obtained thus far [29-31].

The significant overlap in TIMP inhibition of metalloproteinases has likely arisen due to

the events leading to the generation of each TIMP, and phylogenetic analysis shows that

the Timp genes were created by multiple duplication events [32] (Figure 1.1B).

Comparative genomic analyses of Timp evolution between invertebrate and vertebrate

genomes provide insight into these events. The vertebrate Timp family arose from an

earlier whole genome duplication before vertebrate and invertebrate divergence [33].

Furthermore, whole genome duplications are probably responsible for the formation of

the four mammalian Timp genes, before the divergence of tetrapods and teleosts. Timp1,

Timp3, and Timp4 genes are located within introns of three Synapsin genes suggesting

that the Syn-Timp locus duplicated at least three times in M. musculus and H. sapiens.

Timp2 on the other hand is not located within a Synapsin intron, and could have arisen

via duplication of Timp alone or degeneration of the Syn locus after duplication. While C.

intestinalis has a single Timp orthologous to the 4 human Timps, D. rerio has 4 Timp2

genes (Timp2a,b,c,d) orthologous to the human Timp2 [32]. This indicates that

Page 26: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

6

duplication events occurred in the Timp2 locus in D. rerio after the tetrapod/teleost

divergence. Among the four mammalian Timp genes, Timp1 is considered to most closely

resemble the ancestral Timp, as it demonstrates the lowest rate of evolutionary change

[7]. Figure 1.1B illustrates the sequence of gene duplications leading to the creation of

each Timp gene. Timp3 is the second most ancient while Timp2 and 4 arose from the final

duplication event and are the newest members of the family.

1.1.1.2 Targets of TIMPs and phenotypes of Timp deficiency

As the number of published substrates processed by each MMP, ADAM and

ADAMTS steadily increases, the biological importance of each TIMP in regulating tissue

homeostasis comes to light. Table 1.1 lists the inhibitory capacity of individual TIMPs on

some common metalloproteinases (MMP2, MMP7, MMP9, MT1-MMP, ADAM10,

ADAM12, ADAM7, ADAM33 and ADAMTS4), as well as the substrates processed by

each of these enzymes. Given the substrate repertoire that represents ECM and cell

surface molecules, the complexity is evident in TIMP regulation of multiple signaling

pathways. At the stromal:cellular interface, TIMPs can elicit a paracrine response by

ligand processing or modulate cell autonomous function. The immune response is a

classic example where such proteolytic processing alters immune cell activation,

migration, function of clearing antigen and finally resolution of inflammation as reviewed

in [34]. Extracellular proteolytic cascades also trigger „start‟ or „stop‟ signals for

proliferation to guide cell division. The process of liver regeneration, where ~70% of the

liver is surgically removed to initiate compensatory hepatocyte proliferation, offers a

Page 27: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

7

Figure 1.1 – Structure and evolution of Tissue Inhibitors of Metalloproteinases (TIMP)

(A) Proposed structure of the prototypical TIMP. The two domains (N-terminal and C-terminal) are shaded

in green, and major differences between TIMP1 and TIMP3 architecture are highlighted. The conserved

VIRAK sequence in TIMP is required for MMP inhibition. (B) Evolutionary pathway in the generation of

the four mammalian TIMPs. Ancestral TIMP underwent a duplication event to generate two paralogous

TIMPs thereby creating TIMP1. A second duplication event created TIMP3 and the ancestor to TIMP2 and

4. A final duplication resulted in the generation of TIMP2 and 4. TIMP1 most highly resembles the

ancestral TIMP as it has accumulated the fewest mutations across species.

Page 28: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

8

powerful in vivo system to study the contribution of metalloproteinases and TIMPs in cell

division. Factors important for liver regeneration, as demonstrated by genetic mutant

models, are often direct or indirect target of metalloproteinases, and therefore regulated

by TIMPs as reviewed in [35].

Although there is significant overlap in the repertoire of metalloproteinases

inhibited by each TIMP, the expression and localization patterns of these inhibitors limit

the ability of an individual TIMP to comprehensively regulate MMP activity in vivo.

Murine expression analyses indicate that Timp1 is highly expressed in the muscle, lung,

and bone, Timp2 is ubiquitously expressed, Timp3 is enriched in the heart, kidney, lung,

and thymus, and Timp4 in heart, brain and muscle [19, 24, 36, 37]. Reproductive organs

are enriched in most Timps and demonstrate cell type specificity within each tissue. For

instance, Timp2 is present in stromal cells, and Timp3 and Timp4 in epithelial cells of the

developing mouse mammary gland [36, 37]. Therefore despite having common targets,

each TIMP can regulate unique cellular processes by inhibiting an ADAM, ADAM-TS or

MMP in a specific tissue compartment [38]. It is important to note that in addition to

inhibiting metalloproteinase function, TIMPs have been shown to operate via MMP-

independent mechanisms. Below we discuss phenotypes arising in TIMP deficient mice

and attempt to describe the many consequences of regulating metalloproteinase function,

connecting ECM remodeling, intracellular signaling and pathology.

Page 29: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

9

Table 1.1 – Profiling TIMP target MMPs and ADAMs, and their downstream substrates.

TIMP1

TIMP2

TIMP3

TIMP4

Target MMP2 MMP7 MMP9 MT1-MMP ADAM10 ADAM12

ADAM17

[39] ADAM33 ADAMTS4

ECM

Collagen I[40],

IV[41], VII[42] 4 Integrin 2M 5 Integrin Aggrecan I 2 Actinin L-Selectin 9 Integrin

Decorin[43] Collagen IV Dystroglycan Collagen I, II, III Collagen IV 91 Integrin Aggrecan I

Fibrillin[44] Decorin[43] Collagen IV, V, XI Fibronectin MBP Aggrecan I Fibronectin

Galectin-3[45] E-Cadherin Elastin Gelatin

Gelatin[46] Elastin Fibrillin Laminin

Plasminogen[47] Fibronectin I MBP Progelatinase A

Laminin TGF1 ProMMP2

Nidogen Syndecan 1

Testican 1

Cell

Surface

FGF1[48] FasL CCL7 CCL7 APP peptide HB-EGF CD30 APP

peptide

MT4-MMP

FGFR1[49] IL6R CCL11 CXCL12 Delta-Like1 IGFBP3 c-Met CD23

Pro-HGF[50] CXCL12 CD23 IGFBP5 Fractalkine Kit-L1

peptide

ICAM1 CD40 GHBP Insulin

chain

IGF2 NGFR IL1R II TRANCE

IGFBP3 IL6R

IL8 IL15RA

RECK MUC1

Pro-HGF[50] MxL1

Notch

TGF

TNF

TNFR1

TNFR2

indicates inhibition by the respective TIMP

Page 30: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

10

1.1.1.3 TIMP1

TIMP1 was identified as erythroid potentiating activity (EPA) owing to its ability

to augment red blood cell colony formation [17]. TIMP1 deficient mice display mild

phenotypes when challenged in several models including elevated cardiac ECM

breakdown [51-53], enhanced hepatocyte proliferation [54] and altered metabolic control

of obesity [55] (Table 1.2). In accord with its high expression in reproductive tissue, male

TIMP1 deficient mice have modestly lower testosterone levels. Importantly each TIMP

exhibits a unique pattern of expression during sexual maturation in both females and

males, suggesting their specific roles at puberty [56]. The well-studied MMPs and

ADAMs inhibited by TIMP1 include MMP1, MMP9, ADAM10 and ADAM-TS4 (Table

1.1); TIMP1 is a poor inhibitor of membrane type matrix metalloproteinases (MT-

MMPs). The MMP-inhibitory function of TIMP1 correlates well with the phenotypes of

heart tissue remodeling and ventricle function observed in Timp1

mice [51-53]. TIMP1

has also been shown to regulate cell proliferation by inhibiting the release of hepatocyte

growth factor (HGF) by MMP2 and MMP9. HGF is an important growth factor required

for liver regeneration [57, 58], and hepatocytes of Timp1

mice show an accelerated

entry into the cell cycle. Specifically, Timp1

mice exhibit elevated HGF signaling

culminating in accelerated hepatocyte cell division. In this model the MMP-inhibitory

function of TIMP1 is important in regulating cell proliferation [50, 54].

Page 31: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

11

Table 1.2 – Murine phenotypes arising from individual Timp knockouts.

Genotype Phenotypes Reference

Timp1-/-

Reduced luminal obliteration/increased re-epithelialization after tracheal transplantation [59]

Enhanced acute lung injury after bleomycin exposure [60]

Increased HGF activity in regenerating livers [54]

Altered LV geometry and cardiac function [53]

Exacerbated LV remodeling after myocardial infarction [51, 52]

Enhanced estrogen-induced uterine edema [61]

Decreased serum total testosterone levels [56]

Reduced serum progesterone levels during corpus luteum development [62]

Decreased adipose tissue development during nutritionally induced obesity [55]

Timp2-/-

Increased nerve branching and acetylcholine receptor expression [63]

Weakened muscle and reduced fast-twitch muscle mass [64]

Deficits in pre-attentional sensorimotor gating [65]

Required for efficient pro-MMP-2 activation both in vivo and in vitro [66, 67]

Timp3-/-

Enhanced metastatic dissemination to multiple organs [68]

Increased susceptibility to LPS-induced mortality [69]

Enhanced tumor angiogenesis in response to FGF-2 [70]

LV dilation and dilated cardiomyopathy following aortic banding [71]

Increased pulmonary compliance following LPS challenge [72]

Increased inflammatory response to intra-articular antigen injection & TNF-alpha [73]

Spontaneous LV dilatation, cardiomyocyte hypertrophy, and contractile dysfunction [74]

Impaired bronchiole branching morphogenesis [75]

Chronic hepatic inflammation and failure of liver regeneration [76]

Spontaneous air space enlargement and impaired lung function in aged mice [77]

Accelerated apoptosis during mammary gland involution [78]

Exacerbated systolic/diastolic dysfunction following myocardial infarction [79]

Increased corneal neovascularization [80]

Timp3+/-

Acceleration of type 2 diabetes when combined with insulin receptor heterozygosity [81]

Timp4-/-

Compromised cardiac function, higher susceptibility to myocardial infarction [82]

Page 32: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

12

1.1.1.4 TIMP2

The well-established biological function of the TIMP family is to inhibit activated

metalloproteinases. While this is role is performed by all TIMPs, TIMP2 paradoxically

has a central function in MMP2 activation at the cell surface. It acts as an adaptor for

MMP2 by allowing the formation of a trimolecular complex involving MT1-

MMP/TIMP2/Pro-MMP2, where pro-MMP-2 is activated in a two-step process [66, 67,

83]. Thus TIMPs are capable of regulating metalloproteinase activity via multiple

mechanisms [67, 84]. Despite its previously mentioned ubiquitous expression pattern,

only neurological phenotypes have been reported in Timp2 null mice [63, 65]. Lluri et al

[64] have shown that Timp2 is expressed at neuromuscular junctions and colocalizes with

ß1 integrin. Interestingly, ß1 integrin (Itgb1) gene expression is decreased in TIMP2

deficient muscle, suggesting a role for this cell adhesion molecule in maintaining muscle

fiber integrity. The molecular mechanism explaining the decrease in Itgb1 expression in

Timp2

tissue has not been investigated, but one can speculate that the resulting loss of

ECM stability and enhanced metalloproteinase activity indirectly contributes to integrin

proteolysis [85] (Table 1.2).

1.1.1.5 TIMP3

TIMP3 is the only TIMP genetically linked to a human disease. Individuals

harbouring mutations in the C-terminal of TIMP3 suffer from a macular degenerative

disease termed Sorsby‟s fundus dystrophy (SFD) [86, 87]. Interestingly the MMP-

inhibitory property of the mutant TIMP3 is maintained in SFD patients, indicating that

Page 33: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

13

the mechanism underlying the disease is MMP-independent. In fact the elevated

production and resulting accumulation of mutant TIMP3 in the Bruch‟s membrane of

SFD patients is causal to macular degeneration [88]. Additional clinical studies show that

deregulation of epigenetic and microRNA mediated control leading to decreased of

Timp3 expression are linked to age-related macular degeneration [89, 90] and epithelial

tumorigenesis [91-94].

Our group has investigated the physiological role of TIMP3 in multiple tissues by

exposing Timp3

mice to specific stimuli (Table 1.2). Two of the earliest phenotypes

identified were that of air space enlargement in the lung and accelerated involution of the

mammary gland of Timp3 knockout mice [77, 78]. Study by Fata et al [78] shows that the

loss of Timp3 is conducive to accelerated apoptosis during mammary involution, in part

owing to greater matrix proteolysis; the molecular mechanisms contributing to cell death

still remain to be understood. TIMP3 is a potent inhibitor of many MMP, ADAM and

ADAM-TS enzymes (Table 1.2), and the observations of pulmonary air space

enlargement [77], dilated cardiomyopathy [74] and cartilage degradation [95] in

Timp3mice indicate that enhanced metalloproteinase activity leads to compromised

ECM homeostasis as a function of aging. Subsequent studies by Mohammed et al [76],

Smookler et al [69] and Mahmoodi et al [73] investigated the role of TIMP3 in regulating

inflammation and proliferation dependent on the TNF signaling pathway, establishing

TIMP3 as an important negative regulator of TNF bioavailability owing to its unique

ability to inhibit ADAM17. The lack of Timp3 results in increased circulating levels of

TNF as well as its two receptors, TNFR1 and TNFR2. Federici et al [81] identified a

Page 34: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

14

novel role for TIMP3 in providing protection from type 2 diabetes via modulation of TNF

shedding by ADAM17. Here, elevated vascular inflammation accompanied by

insensitivity to insulin signals caused the development of glucose intolerance and

hyperglycemia in Timp3+/

;InsR+/

mice at 6 months of age (Table 1.2). TNF signaling is

also crucial during myogenesis, and TIMP3 was shown to suppress myogenesis in adult

muscle by inhibiting ADAM17 activity and consequently TNF shedding. In their study,

Liu et al [96] identified that the microRNA miR-206 suppressed Timp3 gene expression

during muscle regeneration, showing that TIMP3 acts as a switch during myogenic

differentiation. Using a heart disease model of pressure overload, Kassiri et al [97]

dissected the dysregulation of MMP and ADAM activities in Timp3-/-

mice and found

that increased TNF transcriptionally upregulated several specific MMPs (MMP2, MT1-

MMP, MMP13) while exerting no effect on others (MMP7, MMP9). Intriguingly, the

combination of Timp3 and Tnf

- deletion led to a greater neutrophil influx and production

of MMP8 in cardiac tissue. Here, TIMP3 molecularly linked ECM turnover with that of

cytokine activity when cardiac tissue homeostasis was perturbed. More recently, we

showed that TIMP3 simultaneously regulates EGFR and TNFR1 signaling in the liver

following death receptor activation, thereby balancing cell survival [98]. This reveals a

role for the host genome in responding to acute stress in systems where previously

heterogeneity in the inducing agent(s) were thought to be the primary source of variations

in survival outcomes [99].

Page 35: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

15

1.1.1.6 TIMP4

The last member of the TIMP family, TIMP4 protein is present in cardiomyocytes

and smooth muscle cells, with lower levels in the brain and muscle [100]. Its

colocalization with inflammatory cells such as macrophages and CD3+ T cells suggests a

role in inflammatory cardiac pathologies such as atherosclerotic lesions where it localizes

within necrotic regions. As Timp4 exhibits the most restricted expression pattern of all

the four Timps in mice, its effects may be limited to the target organs despite its property

of being a secreted protein. Generation of the Timp4

mouse by Koskivirta et al [82]

highlighted the in vivo function of this inhibitor in cardiovascular physiology. The mice

were highly prone to ventricular wall rupture in a model of myocardial infarction in an

MMP2-dependent manner. Corroborating evidence supports the protective role of TIMP4

in acute cardiovascular pathology as its expression is induced rat myocardium following

ischemia-reperfusion injury [101], and patients suffering from dilated cardiomyopathy

also exhibit decreased Timp4 gene expression in myocardial tissue [102].

1.1.2 Summary

Individual knockouts of Timp genes do not exhibit in utero lethality, although D.

melanogaster Timp mutant phenocopies integrin mutants displaying inflated wings and

premature lethality [103]. Overall, we see regulation of three important systems from the

in vivo analysis of TIMP function: ECM remodeling, cytokine and growth factor

bioavailability, and inflammatory cell function, critical in maintaining tissue homeostasis.

It is evident that deficiency of a single TIMP does not result in a complete loss of

Page 36: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

16

regulation of these processes, raising the possibility of functional compensation by other

three TIMPs. Interestingly, TIMP3 demonstrates the ability to simultaneously affect all

three systems. These systems are intricately connected in vivo as ECM cleavage releases

not only the structural constraints but also ECM-bound ligands, and TIMP regulation of

receptor shedding influences ligand:receptor kinetics. Thus, TIMPs alter the amplitude of

a signaling stimulus as well as the triggers that serve to recruit infiltrating cells. The

tissue- and stimulus-specific requirement of each TIMP makes them important in

maintaining tissue homeostasis.

1.1.3 Matrix Metalloproteinases (MMPs)

This family of extracellular proteases has long been associated with cellular

invasion and motility, as its initially identified proteolytic activity was to degrade

extracellular matrix (ECM) components such as collagen and fibronectin. The MMPs

share similar modular structures beginning with a signal peptide, a pro-domain containing

the characteristic cysteine-switch motif (PRCGXPD), followed by the catalytic domain

which harbors a zinc-binding motif (HEXGHXXGXXH), a hinge region and a

hemopexin domain. The cysteine-switch and zinc-binding motifs are signatures of

MMPs. Structure/function properties have been used to generate six groups of MMPs:

Collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs and a final

group of unclassified MMPs. Collagenases (MMP1, MMP8, MMP13) are identified by

their ability to cleave collagens I, II and III in addition to non-ECM targets. Gelatinases

(MMP2, MMP9) have structural modifications in their catalytic domains consisting of

Page 37: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

17

fibronectin repeats, which allow them to associate with collagens and laminin. They

degrade gelatin, and MMP2 can also cleave collagens I, II and III. Stromelysins (MMP3,

MMP10, MMP11) can regulate the activity of other MMPs, for instance MMP3 activates

pro-MMP1 to its active form. Matrilysins are identified by their lack of a hemopexin

domain. MMP7 is the prototypical matrilysin and is efficient at cleaving a number of cell

surface proteins such as FasL, syndecan and E-cadherin among others. Membrane-type

MMPs (MMP14, MMP15, MMP16, MMP17, MMP24, MMP25) are transmembrane

proteins with the capacity to activate pro-MMP2. This interesting group of MMPs is

emerging as a significant regulator of several developmental processes, as mice lacking

MMP14 exhibit severe defects in skeletal development and angiogenesis [104, 105].

Metalloproteinases comprise the largest class of proteolytic enzymes in the human

genome [106]. 23 MMPs have been identified, and significant advances have been made

regarding their function in tumor metastasis and immune cell infiltration into tissues. For

example, MMP2 cleavage of fibronectin has been associated with ovarian cancer cell

invasion in humans [107]. On the other hand MMP8 has anti-tumor and anti-

inflammatory properties in mouse models since it degrades Macrophage Inflammatory

Protein 1 (MIP1) and downregulates immune cell influx into epithelial tissues [108,

109]. Membrane type metalloproteinases such as MT1-MMP are critical promoters of

invasion in 3-D collagen gels. Comparative analysis of multiple MMPs with MT1-MMP

has shown that MT1-MMP activity is required for motility of several cell types [110-

113]. This metalloproteinase is a potent collagenase and provides the anchoring

mechanism for MMP2 at the cell surface in addition to mediating its activation. TIMPs

Page 38: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

18

display variable efficiencies at inhibiting MT1-MMP. For instance, TIMP1 is unable to

inhibit this metalloproteinase, while TIMP2 is the critical adaptor between MT1-MMP

and MMP2 at the cell surface [114]. At higher concentrations TIMP2 is an effective

inhibitor of MT1-MMP. Comparative analysis with mouse embryonic fibroblasts

deficient in individual TIMP, in a culture system designed to study MT1-MMP activity

and pro-MMP-2 activation at the cell surface, demonstrates that TIMP2 and 3 are key

modulators of this metalloproteinase [83]. How this complex may operate in vivo, upon

individual and combined TIMP2 and TIMP3 deficiencies, to regulate cell motility is an

important question that remains to be answered.

1.1.4 Disintegrin and Metalloproteinases (ADAMs)

Like MMPs, ADAMs also consist of secreted and transmembrane proteins that

perform ectodomain shedding of numerous substrates. The structure of ADAMs is a

distinguishing property from MMPs, as they harbor disintegrin-like domains that allow

for co-localization with cell surface integrins. Disintegrins were originally identified in

viper venom (hemorrhagic snake venom metalloproteinases; SVMPs) with the capacity to

bind platelet IIb3 receptors and inhibit its interaction with fibrinogen, thereby

preventing platelet coagulation. Structure analysis has identified two groups of ADAM

proteases: Conventional ADAMs and ADAMs harboring thrombospondin repeats

(ADAM-TS). All ADAMs are type I transmembrane proteins and consist of several

domains conserved with MMPs: A signal peptide, a pro-domain followed by a catalytic

domain containing the zinc-binding motif, a disintegrin domain, a cysteine rich domain,

Page 39: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

19

EGF-like domain, a transmembrane domain and a cytoplasmic domain. ADAM-TSs have

thrombospondin motifs but lack a transmembrane domain, are known to cleave pro-

collagen, aggrecan and versican, and are play a functional role in cartilage remodeling

[115] The human genome contains 40 ADAMs and 19 ADAM-TSs, of which 13

ADAMs and 8 ADAM-TSs have shown proteolytic activity. The most intriguing

functions of ADAMs are their capacity to cleave growth factors, cytokines and their

receptors from the cell surface in a process termed „ectodomain shedding‟. The process of

ectodomain shedding regulates cytokine and growth factor signaling by either promoting

cytokine bioavailability (a positive regulation), or generating soluble receptors to

abrogate signaling (negative regulation). This post-translational control is a rapid means

of maintaining signal homeostasis in response to stimuli. It is important to note here that

ectodomain shedding also occurs in the golgi to generate pools of „shed‟ ligand or

receptor that is then mobilized to the cell surface via exocytosis.

1.1.4.1 ADAMs in receptor crosstalk

Study of crosstalk between ADAMs and other membrane-associated pathways

shows the capacity of metalloproteinases to regulate signal transduction. Ullrich and

colleagues have established that G-protein coupled receptors (GPCRs) and ADAMs

interact to activate receptor tyrosine kinases (RTKs) in a process termed “triple-

membrane pass” signaling. Specifically, ligand-stimulation of heterotrimeric G-proteins

consisting of Gi or G11 subunits have been reported to activate numerous ADAMs,

resulting in ectodomain shedding of EGFR ligands such as amphiregulin, HB-EGF or

Page 40: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

20

TGF culminating in EGFR stimulation. The targeted activation of G11 is relevant since

stimulating cells with phorbol esters (e.g. PMA) results in similar activation of ADAMs

and RTK stimulation. Both G11 and PMA activate protein kinase C (PKC): G11 induces

PLC-mediated conversion of phosphatydilinositol 4,5-biphosphate (PIP2) to inositol

(1,4,5)-triphosphate (IP3) and diacyl glycerol (DAG). PMA is a DAG analog, and both

act as second messengers for PKC activation [116-118]. A cellular consequence of these

events is the activation of ADAM proteases, although the precise nature of biophysical

interactions between G-protein dependent signal modulator(s) and ADAM structural

domains is not fully elucidated. ADAMs in turn cleave targets such as EGFR ligands,

which once solubilized may act in juxtacrine, paracrine or systemic manners to stimulate

EGFR and induce a functional physiological response. This complex sequence in the

triple membrane pass signal occurs swiftly and relies on ADAM activity.

1.1.4.2 ADAMs in receptor signaling cascades

ADAMs also participate in proteolytic cascades of evolutionarily conserved

signal transduction pathways in eukaryotes. Examples include Notch and beta-catenin

activation in normal homeostasis and beta-amyloid processing in neurodegenerative

pathologies such as Alzheimer‟s disease. All these pathways utilize ADAMs as a-

secretases which perform ectodomain shedding of the target receptor. The cytoplasmic

domain is subsequently cleaved by -secretase complexes (e.g. presenilin). The regulation

of ADAM activity and signaling consequences of ADAM function remain a challenge for

the field and are the two areas of focus for this thesis.

Page 41: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

21

1.1.4.3 ADAM10

The metalloproteinase ADAM10 was initially identified as a critical contributor to

Drosophila neurogenesis [119]. In the study, Rooke et al named the gene Kuzbanian and

showed that it belonged to the ADAM family of metalloproteinase enzymes. Subsequent

investigations showed that ADAM10 was a component of the Notch signaling pathway,

and loss-of-function or dominant negative mutations in the gene perturbed Notch-

dependent development of Drosophila imaginal wing discs and neurons. In a key set of

experiments, Pan et al revealed that ADAM10 performed proteolytic processing of Notch

ectodomain [120, 121]. Thus ADAM10 became the prototypic Notch sheddase in

eukaryotic cells. Following this, studies of in vivo phenotypes of ADAM10 deficiency

primarily focused on Notch signaling. Emerging evidence also suggested its contribution

to EGFR activation by ectodomain shedding of the EGFR ligand HB-EGF, however this

was only demonstrated in vitro using overexpression or dominant-negative ADAM10

constructs [122]. The generation of conditional ADAM10 deficiency (Adam10-floxed

mice) allowed a targeted study of this gene in mammalian systems. To date, mouse

models of tissue-specific ADAM10 deficiency have shown it to be required for Notch-

mediated differentiation of vascular endothelial cells, neuronal cortex, thymopoiesis,

marginal zone B-cells and keratinocytes [123-128]. Thus in a developmental context,

ADAM10 emerges as a required Notch sheddase.

Page 42: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

22

1.1.4.4 ADAM17

The regulation of Tumor Necrosis Factor (TNF) signaling continues to be an

intensively studied event in biology owing to its significance in development, adult tissue

homeostasis, tumorigenesis and inflammation [129, 130]. ADAM17 was identified as the

first metalloproteinase with the capacity to cleave the TNF-alpha precursor from cell

surfaces [3, 131]. Following this initial finding, systemic bioavailability of TNF in vivo

has been attributed to ADAM17 function, however additional investigation using models

of ADAM17 deficiency have suggested that ADAM10 can also shed TNF from the cell

surface . The generation of ADAM17 deficient murine systems (e.g. Adam17Zn/Zn

mice

lacking the Zn2+

binding domain; Adam17-floxed mice; Adam17ex/ex

hypomorphic mice)

has revealed that ADAM17 is a key regulator of EGFR signaling, since mice lacking

Adam17 phenocopy TGF or EGFR knockout mice [39, 132-137].

A central goal in metalloproteinase biology is understanding the physiological

context where specific metalloproteinases can process the same substrate. An example is

Notch activation, which has been introduced above to as a target of ADAM10. Recent

studies have shown that ADAM17 can indeed shed Notch in a ligand-independent

manner, whereas ADAM10 is required for ligand-dependent Notch activation [138, 139].

As a corollary, combined deficiency of ADAM10 and ADAM17 results in accelerated

embryonic lethality and reveals a redundancy between these metalloproteinases [140].

Given the increasing numbers of substrates processed by ADAM10 and ADAM17, it is

important to acknowledge that they can substitute for each other in experimental and

physiological systems.

Page 43: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

23

1.2 Ectodomain shedding in physiological processes

1.2.1 Cell proliferation

TIMP1 and TIMP2 were initially identified as important factors in erythropoeisis

due to their erythroid potentiating activity [141, 142]. Surprisingly, an antisense RNA-

mediated downregulation of TIMP1 transformed murine 3T3 fibroblasts into tumorigenic

cells [11]. Current research investigating the roles played by TIMPs in regulating

proliferation of both normal and malignant cells demonstrate that the cellular context

within which each TIMP is expressed may dictate its effect on proliferation [36, 143-

147]. Table 1.3 shows that each TIMP can exert opposing effects on proliferation, either

enhancing or inhibiting the process depending on the cell type involved. Additionally, the

mechanisms by which these effects are propagated differ between each study, involving

pathways such as ERK [148], EGFR [146], VEGF [85], HGF [54], FGF, NFB [149],

and cyclin D1. However, most of these responses have not been studied in-depth to

identify the direct connection between these signaling effectors and the ligands under

TIMP regulation. It is also important to note that not all of the mentioned pathways are

investigated in each system, and doing so would reveal common signals influenced by

each TIMP regardless of the cell type.

Page 44: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

24

Table 1.3 - TIMP effects on apoptosis and proliferation in vitro

Method Apoptosis Reference

TIMP-1 rTIMP1 Reduce caspase 3 activity, enhance Bcl-2 expression in hepatic stellate cells (HSC) [150]

rTIMP1 increase activity of PI-3K/AKT, JAK2 tyrosine and Bad phosphorylation, maintained Bcl-XL

expression [151]

rTIMP1 Inhibit TNF-induced apoptosis, activation of PI-3K/AKT in endothelial cells [152]

rTIMP1 Protect MCF10 cells from TRAIL-induced cell death, caspase (3,8,9) activity, FAK and PI-3K

activation [153]

TIMP-2 rTIMP2 Increase apoptosis in activated T lymphocytes and Tsup or Jurkat lymphoma cell lines [154]

TIMP2 vector Increase apoptosis in HCC tumor [155]

TIMP3 Ad-TIMP3 Induction of caspase 8/9 activation and cleavage of PPAR and FAK, mitochondrial acitvation [156]

rTIMP3, Ad-TIMP3 Stabilization of death receptors (TNF-R, FAS, TRAIL-RI), caspase 8 activation [157]

Ad-TIMP3 reverses anti-apopotic effect of TNF-α on Fas-induced apoptosis, inhibit NF-B activation [158]

AdTIMP3 Increased apoptosis of rat aortic smooth muscle cells [143]

TIMP4 rTIMP4 Decrease apoptosis in MDA-MB-435 derived tumors, Increase expression of Bcl-2 and Bcl-XL [159]

Purified TIMP4 Induce apoptosis in transformed cardiac fibroblast but not in normal fibroblastst [160]

Method Cell proliferation Reference

TIMP1 purified TIMP1 Stimulate erythroid burst-forming units [145]

rTIMP1 Decrease proliferation of mammary ductal epithelial cells [36]

rTIMP1 Increase proliferation of MDA-MB-435 and activation of ERK and p38 pathways [161]

TIMP-2 rTIMP2 Suppression of TYK growth factor induced proliferation, disrupt EGFR phosphorylation/Grb-2

association [146]

rTIMP2 Inhibit HMVEC proliferation in response to FGF/VEGF, decrease SHP1-integrin association [85]

rTIMP2 Increase proliferation of A549 lung epithelial, cyclin D1 upregulation, NF-B activation, IkB decrease [149]

TIMP-3 AdTIMP3 increase proliferation of cardiac fibroblasts [162]

AdTIMP3 Decrease proliferation of RA-synovial fibroblasts [147]

TIMP4 AdTIMP4 Increase proliferation of cardiac fibroblasts [162]

rTIMP4 Decrease proliferation of G401 Wilm‟s tumor cells [144]

Page 45: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

25

1.2.2 Apoptosis

As shown in Table 1.3, TIMPs are able to trigger apoptosis in a variety of cell

types including fibroblasts, endothelial, epithelial and hematopoietic cells. Resistance to

apoptosis is an important early trait in cellular transformation. While the intracellular

signaling pathways involved in apoptosis take center stage, the direct link between

TIMPs and cell death are not well defined. This is partly because of their target

repertoire, many of which act as triggers of apoptosis. Hepatic stellate cells

overexpressing Timp1 exhibit an anti-apoptotic phenotype in vitro, and Murphy et al

[150] demonstrate that the anti-apoptotic effect is dependent on its MMP inhibitory

function, as a specific loss-of-function mutation at the MMP-inhibitory region enhances

stellate cells susceptibility to various apoptotic stimuli. Studies on a variety of cell lines

such as the erythroleukemia cell line UT-7, endothelial cells and breast epithelial cells by

independent groups show that TIMP1 inhibits apoptosis via a PI(3)K dependent manner

and modulation of the Bcl family of proteins [151-153]. In contrast to TIMP1, adenoviral

Timp3 overexpression leads to enhanced apoptosis in several cell types [143]. Given the

specificity of TIMP3 for inhibiting ADAM17, and ADAM17-mediated shedding of TNF

and its receptors, Timp3 overexpression in vitro results in stabilization of the death

receptors Fas and TNFR1, thereby sensitizing cells to receptor mediated apoptosis via a

caspase-dependent mechanism [156, 157, 163]. Consistent with these roles of TIMP3,

clinical studies have demonstrated that epigenetic silencing of TIMP3 via methylation

occurs in several types of human cancers such as melanoma, breast, pancreatic, prostate,

colon and cervical cancers, suggesting its role as a tumor suppressor [164-172].

Page 46: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

26

Compared to TIMP1 and TIMP3, less is known of the effects of TIMP2 and 4 on

apoptosis. Current literature suggests that the MMP inhibitory function of TIMP2

protects macrophages from apoptosis in an in vitro overexpression model [173] while

enhancing apoptosis in T lymphocytes [154].

The in vitro studies that demonstrate elevated Timp3 expression results in

enhanced apoptosis in normal and transformed cells [156, 157, 174, 175] are in conflict

with the in vivo study where a lack of TIMP3 leads to accelerated mammary epithelial

apoptosis during mammary involution. The level of Timp overexpression achieved in

these in vitro systems is well above physiological thresholds. Regardless of the manner of

TIMP3 manipulation, the cell death response remains the same, suggesting that the

balance between metalloproteinases and their inhibitors, rather than each component per

se, may dictate cell death.

1.2.3 Ectodomain shedding in immunity

Proteolytic processing at the cell surface and in the pericellular environment by

metalloproteinases is an intriguing point of control for the immune system. Immune

function depends on a series of events that activate and amplify the reaction targeted

towards the antigen, and proteolytic systems are central to all of these events. Recent

studies have firmly established the importance of proteolytic regulation of processes

associated with both innate immunity, which provides the first line of defense against

invading pathogens, as well as with adaptive immunity which affords long-term

protection via the generation of memory against specific foreign antigens. Two important

Page 47: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

27

families involved are the matrix metalloproteinases (MMPs) and the disintegrin

metalloproteinases (ADAMs). Metalloproteinase involvement in aspects of immune cell

migration [176, 177], membrane protein ectodomain shedding [178] and cytokine and

chemokine function [14] have been recently reviewed. This section focuses on the role of

ADAMs and MMPs as regulatory triggers of lymphocyte maturation, clonal expansion,

migration and effector functions.

Metalloproteinases located on the plasma membrane, either as transmembrane

proteins or anchored to cell surface molecules serve as activators or inhibitors of immune

mediators. As illustrated in Figure 1.2, this is achieved through several types of

proteolytic processing, including clipping of low molecular weight chemokines or by

„sheddase‟ activity that releases an intact ectodomain of the target cell surface molecule.

Sheddases can also act in combination with intracellular -secretases to perform RIPping

of the remaining membrane associated. In the context of immunity, the cell surface

localization of proteolysis is ideal for the rapid processing and delivery of already

synthesized instructive signals such as cytokines or chemokines. Proteolytic cleavage is

also essential for generating soluble stimuli to initiate a systemic immune response,

although subsequent receptor shedding from the cell surface through metalloproteinase

activity is also a mode for dampening the initiated response. Chemokine gradients are a

critical early step in the immune response [14] and extracellular proteases shed

membrane chemokines or modify (clip) soluble chemokines to alter their localization

patterns and activity in order to direct immune cell migration to the site of injury [179].

Page 48: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

28

In addition, proteolytic processing plays an important role in activating signal

transduction pathways essential for lymphocyte development, such as T cells [180, 181].

Metalloproteinases are kept in check by tissue inhibitors of metalloproteinases, or

TIMPs in the extracellular milieu. While little is known about the immunological

consequences of altered TIMP activity, deficiency of specific murine TIMPs impacts

inflammation in several models. For instance, shedding of Tumor necrosis factor (TNF)

by ADAM17 is an early step in both systemic and local inflammation: serum TNF levels

are known to regulate components of the adaptive immune system as well as affect target

tissues. TIMP3 is the physiological inhibitor of ADAM17, and Timp3

mice exhibit

elevated soluble TNF levels accompanied by an enhanced potential for inflammation in

several tissues [69, 76, 81]. Thus, many examples are continually emerging where

metalloproteinases and their specific inhibitors actively participate in various aspects of

immunity.

1.2.3.1 Metalloproteinases facilitate lymphocyte development and proliferation

Lymphopoiesis begins in the bone marrow, where B and T lymphocyte precursors

are generated. T cell progenitors migrate to the thymus and a cascade of interactions

between immature thymocytes and the thymic stroma produces mature T lymphocytes

which then migrate to peripheral lymphoid tissues [182, 183]. In the spleen or lymph

nodes, mature T lymphocytes encounter foreign antigen-MHC complexes on the surface

of antigen presenting cells (dendritic cells, macrophages, activated B cells) and an

Page 49: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

29

Figure 1.2The processes of clipping, shedding and RIPping

Clipping Shedding RIPping

The proteolytic activation or

inactivation of chemokines and other

chemoattractants by the removal of

short N- or C-terminal peptides.

MMPs are important players [179,

184-186], however amino and

carboxypeptidases and proteinases of

other mechanistic classes are key

activities in some cell types [184].

The proteolysis of the ecto-domain

of cell surface proteins, including

chemokines, cytokines, growth

factors and receptors, as well as

adhesion molecules [3, 187-190].

Shedding can occur through

autocrine, juxtracrine or paracrine

mechanisms via soluble or

membrane-bound protease activity

[191, 192]. The process of shedding

may initiate or dampen a signaling

response depending on the

stimulatory or inhibitory role(s) of

the target being cleaved [137, 193].

ADAM17 mediated TNF shedding

can result in enhanced TNF

signaling, however since ADAM17

also cleaves TNFR1 and 2, the

shedding of these receptors dampens

the initial enhancement of systemic

TNF activity [131, 132, 194].

Regulated Intramembrane Proteolysis

is often performed by -secretases

such as the presenilin complex which

are aspartic proteinases [195].

RIPping is widespread in many

cellular responses and is critical to

downstream signaling upon receptor

activation of many pathways. Notch

signaling requires RIPping of the

intracellular domain (ICD) of the

receptor Notch 1, which translocates

to the nucleus and induces a

transcriptional response [196]. This

process has a specific sequence of

proteolysis where ectodomain

shedding of Notch by ADAMs must

occur prior to RIPping of the ICD

[180, 197]. Interestingly the Notch

ligand Delta-like 1 undergoes a

similar sequence of shedding and

RIPping in by ADAMs and -

secretases in a signal-independent

manner [198].

Page 50: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

30

adaptive immune response is generated. During this event, a small activated T cell

population rapidly proliferates to provide sufficient activated T cells for effector function.

The immune response is then downregulated, leaving a small population of antigen

specific memory T cells to expedite a more rapid secondary immune response.

Notch signaling plays a crucial role in early lymphocyte commitment in the bone

marrow. ADAM mediated shedding of the Notch extracellular domain (ECD) is only

possible following binding to a Notch ligand such as Delta-like 1. Of the four mammalian

Notch proteins, extracellular cleavage of Notch1 and its ligand Delta-like 1 is performed

by ADAM17 [139] and potentially by ADAM10 [181]. RIPping of the Notch 1

intracellular domain by the -secretase presenilin 1 results in its translocation to the

nucleus, triggering transcription of Notch 1 target genes (Figure 1.2). ADAM mediated

cleavage of the Notch ECD commits hematopoeitic progenitor cell differentiation to the

T cell lineage and concomitantly inhibits commitment to the B cell lineage [199, 200]. In

the thymus, the requirement for ADAM17 in the transition of double negative (CD4-CD8

-

) thymocytes to a double positive state (CD4+CD8

+) has been identified by studies on

mice lacking functional ADAM17. C57BL/6 Adam17∆Zn/∆Zn

mice live only until 1 day

postnatal [131], or 2-3 weeks under a mixed (C57B/L/6 x 129) background [201], and

exhibit an overall reduction in lymphocyte numbers in addition to impaired lymphocyte

maturation. Li et al have further shown that this protease acts through the stromal

compartment since ADAM17 deficient bone marrow transplanted into rag deficient mice

is able to support T cell development from the double negative to double positive state.

Although this indicates a non-cell autonomous requirement of ADAM17 during

Page 51: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

31

lymphopoiesis, the bone marrow transplanted mice are not fully rescued suggesting the

requirement of additional factors, and thus the cell autonomous role of ADAM17 cannot

be completely ruled out.

Another important concept in early adaptive immunity is that of clonal expansion

[202], and shedding at the cell surface influences lymphocyte proliferation by modulating

ligand receptor interactions. LAG3 (Lymphocyte Activation Gene 3) inhibits T cell

proliferation, and its cleavage by ADAM10 and ADAM17 alters clonal expansion

(Figure 1.3A). Notably, ADAM10 cleaves LAG3 constitutively whereas ADAM17

activity requires induction by phorbol ester or activation through CD3. Li et al clearly

demonstrate that ADAM10 provides a means for basal T cell proliferation while

ADAM17 enhances this response following the activation of the T cell receptor pathway

in a PKC-dependent manner [203].

The final critical step in immune regulation is the elimination of activated T

lymphocytes after successful clearance of antigen. The death inducing ligands, such as

those belonging to the TNF superfamily, play roles in such regulation. Fas ligand (FasL)

is a classic example of a factor engaged in both cell-mediated and soluble cytotoxicity.

The former involves the membrane bound form of FasL, whereas the latter requires

shedding by ADAM10 or MMP7 to generate a soluble FasL fragment, which can act

systemically [204]. Specific inhibition of ADAM10 results in significantly reduced FasL

shedding in primary human T cell cultures [205]. The resulting elevated surface

expression of FasL on cytotoxic T cells is associated with an increase in their killing

capacity, with more target T cells undergoing activation-induced cell death (AICD) [206]

Page 52: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

32

Figure 1.3 - Cell surface sheddases influence adaptive immunity.

(A) ADAM17 sheds TNF from activated macrophages to enhance systemic toxicity and inflammation.

However ADAM17 can also shed TNF receptors 1 and 2 and dampen TNF function. Membrane-bound

TNF activates effector T cells, and thus ADAM17 may regulate T cell cytotoxicity by altering levels of cell

surface TNF and its receptors. Shedding of the inhibitory molecule LAG3 by ADAM10 and 17 on the

surface of activated T cells promotes clonal expansion required for a successful immune response. (B)

Hosts with MMP8 deficient PMNs exhibit elevated serum IL-13, IL-4 and IgE levels owing to enhanced

Helper T (TH2) cell activity. Consequently the humoral allergic response is also enhanced via elevated

CD23 signaling and antibody production. ADAM10 and 33 shed cell surface CD23 resulting in

macrophage activation and inflammation. It is unknown whether soluble CD23 directly activates

macrophages (dashed line). (C) T lymphocytes express FasL to kill activated effector T cells, termed

activation induced cell death (AICD). Tumor cells also produce FasL as a mechanism of immune evasion.

The T cell receptor NKG2D reduces Fas expression while enhancing FasL production in a cell-autonomous

manner. Tumor cells counteract this via enhanced activity of ERp5 (a disulphide isomerase-like protease),

shedding MICA from their cell surface, causing a decrease in NKG2D expression which sensitizes effector

T cells to Fas mediated apoptosis.

Page 53: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

33

(Figure 1.3C). Members of the TNF superfamily require trimerization for activity, and it

is possible that an increased surface level of these factors promotes clustering, which may

also contribute to the increased functional capacity observed by Schulte et al [206].

1.2.3.2 Myeloid and lymphoid cell migration is dependent on metalloproteinase

activity

One of the earliest responses upon acute and chronic inflammation is that of

leukocyte recruitment to the site of infection. Polymorphonucleocytes (PMNs) are among

the first immune cells to arrive and set the stage for innate as well as adaptive immunity.

Significant knowledge has accumulated on the importance of generating chemokine and

cytokine gradients for immune cell recruitment, and the clipping performed by

metalloproteinases is an emerging paradigm for this step. Chemokines bind to

proteoglycans such as syndecan-1, providing a “homing” signal to circulating PMNs.

MMP7 secreted by PMNs cleaves syndecan-1 bound to CXCL1 and thus triggers the

generation of a CXCL1 chemokine gradient, which attracts neutrophils to the site of

injury [14]. ADAM10 and ADAM17 also shed transmembrane chemokines such as

CX3CL1, while ADAM10 releases CXCL16 [187-189, 207]. This enhances neutrophil

extravasation through the blood vessel endothelial layer into tissue interstitium. In

concert, metalloproteinase-mediated cleavage of these substrates modulates local immune

responses, providing an additional level of control over early immunity.

The generation of a gradient is often coupled with the modification of cell

adhesion properties of immune and endothelial cells, and is performed by the same

Page 54: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

34

subsets of ADAMs and MMPs. Leukocyte recruitment is also affected by the proteolytic

modulation of cell-cell interaction. The first step of transendothelial migration is

mediated by L-selectin, an adhesion molecule expressed on many inflammatory cells

including monocytes, neutrophils as well as B and T lymphocytes. It mediates the initial

tethering and subsequent rolling of cells along vascular endothelium. Upon leukocyte

activation, L-selectin is rapidly clipped by ADAM17 at a specific extracellular site

proximal to the plasma membrane [178, 208]. Blocking ADAM17 activity by both

synthetic and natural MMP inhibitors causes the arrest of lymphocyte migration across

high endothelial venules [209]. B cell chronic lymphocytic leukemia (B-CLL)

lymphocytes have impaired transendothelial migration compared to normal peripheral

blood lymphocytes [210]. Moreover, transendothelial migration of B-CLLs involves the

universal loss of L-selectin and CD23 from their cell surface. ADAM17 deficient

neutrophils are still able to shed L-selectin from their surface and it has recently been

shown that ADAM8 is associated with L-selectin proteolysis in activated neutrophils

[211]. Activated endothelial cells express the cell adhesion molecules ICAM-1 and

VCAM-1, which direct their interaction with leukocytes. ADAM17 has been shown to

shed these molecules from endothelial cells [212-214]. Some secreted serine proteinases,

such as elastase and cathepsin G, can also shed VCAM-1 from neutrophils. The

generation of soluble chemokines and cytokines, and the cleavage of endothelial adhesion

molecules has also been termed ectodomain shedding, and its functional relevance in the

context of inflammation has been reviewed in detail by Garton et al [176].

Page 55: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

35

MMP8 and MMP9 direct the early neutrophil response to the site of infection

[215, 216]. Several studies have shown that MMP8 or 9 deficiency results in impaired

neutrophil responses highlighted by decreased recruitment. A number of studies have

shown that neutrophil transmigration of the vascular endothelium occurs through either

transcellular or paracellular diapedesis and does not involve proteinases [217]. However,

MMPs are probably involved in the necessary processes of both chemoattractant and

basement membrane proteolysis. Neutrophils store MMP9 in tertiary granules and

release it upon activation by proinflammatory cytokines. Using a model of abdominal

sepsis, Renckens et al demonstrate that MMP9 activity is required for an adequate

neutrophil response to deal with E.coli infection [216]. Like MMP9, MMP8 is produced

by human PMNs in both forms, as a secreted pro-MMP or as an active enzyme on their

cell surface [218]. While soluble MMP8 is inhibited by TIMP1 and synthetic inhibitors,

the cell surface form of MMP8 is not. This opens the possibility of a juxtacrine

mechanism for PMN migration in which cell surface MMP8 activity affects the adjacent

cell behavior. In addition to enhanced MMP8 release from granulocytes, neutrophil

activation leads to transcriptional upregulation of this metalloproteinase. MMP8 deficient

mice have twice as many neutrophils in their alveolar space after lipopolysaccharide

inhalation and exhibit elevated serum levels of IgG1, IgE and IL4 – all markers of a TH2

response (Figure 1.3B) [219]. Work by Lopez-Otin et al proposes an unexpected anti-

inflammatory function for MMP8 in several in vivo models of acute and chronic

inflammation all as a result of altered neutrophil recruitment, demonstrating the role of

MMPs in directing an immune response [215, 219, 220].

Page 56: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

36

1.2.3.3 Shedding of cell surface molecules regulates humoral and cell based

immune effector function

1.2.3.3.1 Humoral immunity

Chemokine and cytokine signalling is central to initiating an adequate humoral

response. TNF is a potent pro-inflammatory cytokine and many of its paracrine and

endocrine functions require shedding of its cell membrane bound form by ADAM17

[131]. Curiously, ADAM17 also regulates the levels of the receptors TNFR1 and TNFR2

by cleaving them from the cell surface to generate soluble antagonists [3, 221] (Figure

1.3A). Bacteria have been found to induce ADAM17, either through direct activation of

the EGFR or through IL-6 induction. EGFR transactivation per se is a process that

requires shedding of the membrane bound forms of the EGFR ligands through the

mobilisation of ADAMs [133]. In fact, ADAM17 deficient mice phenocopy defective

EGF signaling as they lack soluble TGF, an important EGFR ligand. Furthermore,

ADAM17 also causes IL-6R shedding; in a model of trans signaling, epithelial IL-6 and

soluble IL-6Rα first form a complex and subsequently interact with a membrane-

associated gp130 homodimer to activate CCL-2 expression and inhibiting CXCL8

production [222]. Thus, ADAM17 is involved with multiple facets of the humoral

response, beginning with its production and subsequently by influencing cytokine

signaling.

Once cleaved, soluble TNF has significant and immediate proinflammatory

consequences if left unchecked. This is evident during septic shock, where increased

systemic levels of TNF leads to non-specific inflammation causing vasodilation,

Page 57: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

37

hypotension and ultimately multiple organ failure. Given that ADAM17 is the primary

TNF sheddase, its inhibitor TIMP3 becomes an important negative regulator of

inflammation. This control is lost in Timp3

mice, which exhibit increased sensitivity to

septic shock owing to uncontrolled TNF activity [69]. Ablating TNFR1 in parallel to

TIMP3 deficiency rescues the mouse from septic shock, thus demonstrating the central

role played by metalloproteinase-mediated regulation of TNF bioavailability in acute

inflammation.

Allergic immune responses are primarily mediated by the antibody IgE, which is

unique in that it is mostly located in tissues as opposed to lymph nodes or other primary

immune organs. IgE associates with the high affinity receptor FcRI on mast cells to

initiate the immune response via multiple activating signals [223]. A second low affinity

IgE receptor called CD23 or FcRII has been shown to exist on several lymphoid (e.g. B

cells, activated T cells, dendritic cells) and myeloid (monocytes, eosinophils, platelets)

immune cells, and even thymic epithelial cells [224]. FcRI activity is considered a non-

specific allergic response, whereas CD23 is involved in enhancing antibody response to

specific antigen in the presence of IgE. The shedding of CD23 has been shown to

increase macrophage activation via TNF release by peripheral blood mononuclear cells

that induces IFN production and release, thereby propogating inflammation [225].

Indicative of its role in exacerbating inflammation, increased soluble CD23 can be

detected in inflamed joints of rheumatoid arthritis patients [226, 227]. It is now known

that ADAM10 is the principal CD23 sheddase while ADAM8, ADAM9, ADAM12,

ADAM15, ADAM17, and ADAM19 do not participate in this event (Figure 1.3B) [227,

Page 58: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

38

228]. The potential for functional redundancy between ADAM10 and ADAM33 has also

been addressed by gain-of-function experiments [227]. ADAM10 emerges as a more

specific and required sheddase, as B cells expressing only ADAM10 successfully shed

CD23. Even though specific inhibition of ADAM10 decreased both constitutive and

enhanced CD23 cleavage, it does not eliminate shedding completely suggesting the

presence of background ADAM33 activity.

1.2.3.3.2 Cell-mediated immunity

Adaptive immunity is mediated by activated T cells or by the antibody response

orchestrated by CD4+ and CD8

+ helper T cells and B cells. Once again, many aspects of

these events involve proteolytic modification of the status of cell membrane proteins as

part of a rapid response. HLA (human leukocyte antigen)-A2 is an MHC Class I protein

with primary functions in T cell development and initiation of immune response by

antigen presentation. It was recently shown in cell culture systems that ADAM10

processing of HLA-A2 subsequently leads to -secretase activity to release the

cytoplasmic domain which is then subjected to proteasomal degradation [229]. The

molecular contacts at the interface between T cells and antigen presenting cells define the

nature of the response against a particular antigen. As the T cell receptor engages a

peptide MHC complex on antigen presenting cells, a specialized supra-molecular

structure termed the immunological synapse assembles. This contains co-stimulatory and

adhesion molecules, many of which are regulated by proteolytic mechanisms. In addition

to endocytosis and lysosomal degradation, shedding can also occur, producing soluble

Page 59: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

39

forms which may act as antagonists of the immune response. Shedding could play a key

role in dictating the components in the synapse, hence defining its nature [230]. CD28,

CD40, CD80 and CD86 are all detected as soluble forms in patients with autoimmune

diseases and could be the result of excessive or aberrant proteolytic processing [231].

Evidence that ADAM17 is a CD40 sheddase has been presented by Contin et al [232].

Similarly, proteolytic shedding of B7 molecules on antigen presenting cells during

CD4+T cell interaction has been documented as a dynamic contributor to the modulation

of costimulatory signals for cytokine production [233]. Although B7 cleavage was

blocked by the synthetic metalloproteinase inhibitor GM6001, the identity of the specific

enzyme(s) is not yet known.

Shedding and clipping have also been implicated in tumor immunity, both in the

regulation of tumor cell survival and in immune evasion through the process of

„immunoediting‟. Effector T cells can arrest tumor growth by elevating FasL activity

triggering apoptosis of tumor cells via the Fas pathway [234]. However, the mechanism

by which T cells regulate their own survival and avoid Fas-mediated apoptosis in this

scenario has only recently been uncovered. The Spies group has identified NKG2D, a

costimulatory T cell receptor as an important regulator of T cell survival but promoter of

target cell apoptosis. NKG2D+ T cells exhibit enhanced FasL production and shedding -

possibly via MMP7 or ADAM10 - to generate soluble FasL which promotes apoptosis.

Expression of NKG2D simultaneously decreases production of the receptor Fas in an

autocrine manner, conferring resistance to NKG2D+ T cells and thereby prolonging their

survival [235]. Many tumors counter FasL cytotoxicity by cell surface shedding of the

Page 60: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

40

MHC class I related ligand (MICA) via a disulphide-isomerase-like protein, ERp5. In an

example of immunoediting, proteolytic cleavage of MICA from tumor cells results in its

binding to NKG2D followed by internalization by T cells [236] (Figure 1.3C). These

studies highlight how sheddases impact immune regulation of tumors as well as tumor

editing of an immune response.

1.2.4 Summary

Proteolytic processing through clipping, shedding and RIPping affects a diversity

of substrates that are critical throughout immunity. These encompass cytokines and their

receptors, chemokines and their adaptors, extracellular components of cell-mediated and

humoral immunity, matrix proteins, cell adhesion molecules as well as those linked to

tumor immunity. Since proteases are involved in all aspects of an immune response -

from development, expansion, effector function, to termination - regulation of their

activity is important in maintaining immune homeostasis. These proteases, which

function extracellularly or on the cell surface, are themselves tightly regulated by

trafficking, activation and by natural inhibitors. Furthermore, interactions with

intracellular proteolytic systems such as the γ-secretases provide another layer of

complexity to the immune response. TIMPs, the biological inhibitors of MMP and

ADAM activity confer an essential point of control during normal immune response, and

synthetic inhibitors of these proteases may prove to be novel therapeutic agents in

treating autoimmune disorders or other inflammatory conditions. However caution must

be exercised here as complete specificity to particular ADAMs and MMPs has yet to be

Page 61: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

41

achieved by small molecule or synthetic metalloproteinase inhibitors, and utilization of

currently available inhibitors would possibly result in unforeseen effects on immune cell

function in both innate and adaptive immunity.

Although only a few ADAMs, MMPs and TIMPs have been explored for their

function in immunology, the emerging data has opened a new awareness for this novel

system of regulation (Figure 1.4). Multiple members exist within these protease families

in humans (23 MMPs; 13 ADAMs), with each being able to process several diverse

substrates. This complexity of MMP or ADAM processing parallels the immune

response, and can be seen to operate at the tissue, cellular and molecular levels.

Additionally, mechanisms of controlling an immune response are highly specific and

depend on proteolysis for the release of antagonists, downregulation of pro-inflammatory

signaling, and even death of activated immune effectors. Despite the subtle biochemical

processing where proteases functionally modify rather than degrade their substrates, the

physiological consequences to the organism are significant.

Page 62: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

42

Figure 1.4 – Localization of metalloproteinases and their substrates impacting immunity.

The metalloproteinases and their substrates discussed in this section are listed above, representing a small

fraction of the ADAMs and MMPs that influence the immune response. The shaded boxes indicate the cell

type(s) associated with each substrate. Note: CD23 is expressed on a variety of myeloid (monocytes,

eosinophils, platelets) and lymphoid (B cells, activated T cells, dendritic cells) lineages [224, 228].

Page 63: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

43

1.3 Thesis outline

1.3.1 Study Rationale

We propose that regulated ectodomain shedding plays regulatory roles in

cytokine bioavailability and signaling. As TIMPs operate in the extra-cellular milieu,

they can potentially act as signaling switches at tissue interfaces, controlling the spectrum

of cytokine signaling that occurs during an immune response. In this manner, we

hypothesize that the TIMP3/ADAM17 axis can direct epithelial:immune crosstalk and

tissue homeostasis during acute and chronic inflammation. In vitro investigations of

TIMP function suggest paradoxical roles of these genes in different cellular processes

depending on the experimental system utilized (discussed in section 1.2), and establishing

the relevance of these findings in vivo is key to understanding which cellular processes

are indeed controlled by endogenous TIMPs.

Chapter 2 of this thesis identifies that TIMP3 simultaneously orchestrates TNFR1

and EGFR signaling in the liver during sterile inflammation caused by the Fas agonist Jo-

2. Chapter 3 establishes the contribution of non-hematopoietic TIMP3 as a stromal factor

that restricts undesired NKT and CD4+ T cell influx into the liver microenvironment,

thereby providing protection against T-cell mediated autoimmune hepatitis induced by

the lectin concanavalin A. Chapter 4 details the critical function of ADAM17 in barrier

immunity; we determine that in the skin of adult mice, keratinocyte-specific ADAM17 is

required for tonic Notch activation which in turn checks uncontrolled transcription of

epithelial cytokines (specifically TSLP and G-CSF) by the transcription factor c-Fos/AP-

Page 64: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

44

1. This basal regulation of AP-1 activity is required to prevent atopic dermatitis and

myeloproliferative disease, thereby maintaining barrier homeostasis in vivo.

1.3.2 Thesis Objectives

Investigating the role of TIMP3 in orchestrating pro- and anti-apoptotic signals

during death receptor activation in the liver

Identifying the relevant tissue compartment of TIMP3 production and its

physiological relevance in hepatic lymphocyte function

Delineating the ADAM17-dependent cellular signaling mechanism(s) which

regulate barrier immunity in the epidermis

Page 65: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

45

2 CHAPTER 2

Ectodomain shedding of EGFR ligands and TNFR1 dictates hepatocyte apoptosis

during fulminant hepatitis in mice

A version of this chapter is a published manuscript:

Murthy, A.., Defamie, V., Smookler, D.S., Di Grappa, M.A., Horiuchi, K., Federici, M.,

Sibilia, M., Blobel, C.P., Khokha, R. Ectodomain shedding of EGFR ligands and TNFR1

dictates hepatocyte apoptosis during fulminant hepatitis in mice. J Clin Invest, 2010.

120(8): p. 2731-44.

Author contributions:

Murthy, A. – designed and performed majority of the experiments, wrote manuscript

Defamie, V. – primary hepatocyte culture

Smookler, D.S & Di Grappa, M.A. – technical assistance with Jo-2 injections

Horiuchi, K. & Blobel C.P. – providing Adam17fl/fl

mice and Adam17-/-

MEFs

Federici, M. – experimental advice on acetaminophen toxicity model

Sibilia, M. – treatment of Egfrfl/fl

and Alb-Cre;Egfrfl/fl

hepatocytes with Jo-2

Page 66: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

46

2.1 Abstract

Fulminant hepatitis by Fas recruits extrinsic apoptotic and hepatoprotective

signals, but mechanisms for their integration are unknown. Tissue inhibitor of

metalloproteinases 3 (TIMP3) controls the critical sheddase a disintegrin and

metalloproteinase 17 (ADAM17), and may dictate stress signaling. Using mice or cells

lacking Timp3, Tnf, Tnfr1, Egfr, and Adam17, we show that ectodomain shedding of TNF

receptors and EGF family of ligands controls JNK, NF-B, caspase and ERK1/2

activation in Fas-induced hepatitis. We first demonstrate that TNF signaling promotes

hepatotoxicity, while excessive TNFR1 shedding in Timp3−/−

mice is protective.

Compound deletions of Timp3/Tnf and Timp3/Tnfr1 conferred complete resistance to Fas-

induced toxicity, uncovering parallel EGFR activation as a significant survival signal.

Loss of TIMP3 enhanced metalloproteinase-dependent EGFR signaling due to increased

release of EGFR ligands TGF, amphiregulin and HB-EGF, while depletion of shed

amphiregulin re-sensitized Timp3−/−

hepatocytes to apoptosis. These findings

demonstrate that TIMP3 and ADAM17 cooperatively dictate cytokine signaling during

death receptor activation. Finally, adenoviral delivery of ADAM17 prevented

acetaminophen-induced liver failure in a clinically relevant model of Fas-dependent

fulminant hepatitis. Thus regulated metalloproteinase activity integrates survival and

death signals during acute stress.

Page 67: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

47

2.2 Introduction

Hepatocytes are highly sensitive to death receptor-mediated apoptosis.

Engagement of this pathway elicits multiple and complex extrinsic signals, some

apoptotic while others hepatoprotective. The cumulative tissue response to these signals

determines survival of the organism; however our understanding of the mechanisms that

coordinate them remains incomplete. Mouse models of acute and chronic hepatotoxicity

have revealed processes downstream of TNF superfamily receptors such as TNF receptor

1 (TNFR1), Fas/CD95, and death receptors 4 and 5 (DR4, DR5) that direct context-

dependent responses. For example, lipopolysaccharide- (LPS) and concanavalin A-driven

hepatitis require TNF signaling. In the LPS model, soluble TNF secreted by components

of the immune system, primarily macrophages and neutrophils, drives the destruction of

hepatocytes and results in fatal hepatitis, whereas hepatitis caused by concanavalin A is

promoted by membrane-bound TNF on T lymphocytes [237]. Furthermore, membrane-

bound TNF on T cells is protective during listeria and mycobacteria infection [238, 239].

Such studies have shown that TNF Receptor 1 signals through c-Jun N-terminal Kinase

(JNK), NF-B and caspases during a stress response, each with significantly different

outcome on survival [240-242]. In addition to the fundamental role that TNF signaling

plays in the above systems, TNF may be a contributing factor in pathologies where it is

not the primary stimulus. TRAIL and its receptor DR4 are known to promote fulminant

hepatitis, but there is a lack of conclusive evidence for the function of TNF in Fas-

mediated hepatic failure [243].

Page 68: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

48

Even less is known about the role of EGFR signaling during acute hepatic stress.

Of the four ERBB family members, EGFR/ERBB1 provides critical mitogenic signals

through ERK1/2 phosphorylation and mice lacking EGFR do not survive postnatally

[244]. Studies have also suggested a protective role for EGFR signaling in models of

hepatocyte regeneration and cellular stress induced by reactive oxygen species or DNA

damage [117, 245]. However controversy exists regarding the role of EGFR activation in

Fas-mediated hepatocyte apoptosis, as some groups have proposed that EGFR interaction

with Fas is required for death-receptor tyrosine phosphorylation and DISC formation

while others report that EGFR ligands EGF, HB-EGF and amphiregulin are protective in

this model [245-249]. Genetic manipulation of EGFR and its modulators is required to

establish its function in hepatocyte apoptosis.

TNF, its receptors, and several EGFR ligands are cleaved from the cell surface by

a disintegrin and metalloproteinase 17 (ADAM17/TNF Alpha Converting Enzyme) in a

process termed ectodomain shedding [3, 131]. ADAM17-mediated release of EGFR

ligands from the cell surface via ectodomain shedding is considered essential for EGFR

activation. Binding of soluble ligand exposes a dimerization loop via a conformational

change in the EGFR monomer, which is then followed by homodimerization and receptor

tyrosine kinase activity. Membrane bound or uncleavable ligand can therefore impede

EGFR activation by preventing dimerization [133, 137]. Mice that lack ADAM17

phenocopy Egfr−/−

mice, pointing to the requirement of this metalloproteinase in EGFR

signaling [135]. ADAM17 activity is blocked by TIMP3, a stromal inhibitor of several

matrix metalloproteinases (MMPs) and ADAMs [104, 250]. During inflammation,

Page 69: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

49

TIMP3 provides an extracellular mode of controlling TNF-mediated stress response. We

have previously reported that TIMP3 checks TNF release and activity during liver

regeneration and endotoxin-mediated septic shock [69, 76]. Together the

ADAM17/TIMP3 axis may simultaneously affect the opposing signals that determine

survival following death receptor activation.

This study investigates stromal control of ectodomain shedding as an integrator of

TNF and EGF signaling during Fas-mediated hepatotoxicity. We provide the first in vivo

evidence that TNF sensitizes hepatocytes to Fas-mediated death and that increased

TNFR1 shedding in TIMP3 deficient mice protects from fulminant hepatitis by

dampening TNF activation of JNK, NF-B and caspases. We also show that TIMP3 is a

negative regulator of EGFR-mediated ERK1/2 phosphorylation, which provide

hepatoprotection. Further, this concept applies to a clinically relevant model of drug

overdose-induced acute liver failure requiring Fas. TIMP3-regulation of ectodomain

shedding is therefore necessary for the apoptotic response during Fas-mediated

hepatotoxicity.

2.3 Results

2.3.1 TNF signaling sensitizes hepatocytes to Fas-mediated apoptosis

The liver is highly sensitive to inflammatory and apoptotic stimuli of TNF

superfamily members such as Fas, TNF, and TRAIL [237, 243], however the exact role

of TNF signaling in mouse models of Fas-mediated hepatotoxicity has yet to be defined.

Primary murine hepatocytes were tested for their sensitivity to Fas-mediated apoptosis in

Page 70: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

50

the presence of a low level of TNF (1ng/ml). TNF enhanced apoptosis in wildtype

hepatocyte cultures treated with the Fas agonist Jo-2, as measured by caspase 3/7 activity.

However TNF on its own was not cytotoxic at this low concentration (Figure 2.1A). This

highlights the potency of TNF to promote Fas-mediated apoptosis.

While mice lacking TNFR1 have previously demonstrated sensitivity to Fas-

mediated hepatotoxicity [251], the kinetics of their survival has not been explored. Here

we utilized a clone of Jo-2 (listed in Methods) that allowed us to dissect the kinetics of

survival after induction of hepatotoxicity. Wildtype, Tnfsf2 deficient (Tnf−/−

) and

Tnfrsf1a deficient (Tnfr1−/−

) mice received a lethal dose of Jo-2 (0.65µg/g i.p.). Both

knockouts showed a significant delay in fulminant hepatic failure with a time to

morbidity of approximately 10 hours compared to 5 hours for wildtype controls (Figure

2.1B). An in vitro timecourse of hepatocyte apoptosis over 24 hours using 1ng/ml TNF +

10ng/ml Jo-2 revealed comparable caspase 3/7 activation between Tnf−/−

and wildtype

cells as expected when exogenous TNF was added to culture, whereas Tnfr1−/−

hepatocytes exhibited decreased caspase 3/7 activation (Figure 2.2A). Histological

examination of livers 3 hours after administration of PBS (vehicle) or Jo-2 revealed

extensive tissue damage and hemorrhaging only in the wildtype mice, however all

genotyped eventually succumbed to fulminant hepatic failure with similar liver damage

(Figure 2.1C, Figure 2.3). Immunohistochemical staining revealed activation of caspase 3

only in wildtype livers after Jo-2 administration (Figure 2.1C). Additional markers of

hepatotoxicity such as serum alanine transaminase (ALT) were significantly elevated in

wildtype mice 3 hours after Jo-2 administration (Figure 2.1D). Further, processing of

Page 71: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

51

Figure 2.1 Ablation of TNF signaling delays Fas-mediated hepatotoxicity.

(A) Apoptosis of wildtype primary hepatocytes treated for 12 hours with indicated doses of Jo-2 in

presence or absence of 1ng/ml TNF. Caspase 3/7 activity was measured by rate of cleavage of fluorgenic

peptide Ac-DEVD-AMC and depicted relative to untreated samples. *P<0.03. Data are mean ± S.D. (n=3).

(B) Survival curve of wildtype (WT), Tnf −/−

and Tnfr1−/−

mice injected intraperitoneally (i.p.) with

0.65μg/g Jo-2. P<0.005 vs. WT (log-rank test). (C-E) WT, Tnf −/−

and Tnfr1 −/−

mice were injected i.p. with

0.65μg/g Jo-2 or PBS. Mice were sacrificed and tissue obtained 3 hours after injection. (C) Liver histology

(H & E, active caspase 3 immunohistochemistry) depicting absence of tissue damage and caspase 3

activation in Tnf −/−

and Tnfr1−/−

mice after treatment with Jo-2. Sections are representative of at least 4

mice per genotype and treatment. Bar = 100μm (D) Hepatic toxicity measured by serum ALT levels.

*P≤0.04. Data are mean ± S.D. (n=5). (E) Immunoblots assaying the processing of caspase 8 and 3 in liver

lysates. All data are representative of at least 2 independent experiments.

Page 72: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

52

caspases 8 and 3 were seen in wildtype liver lysates, depicting the onset of apoptosis in

these mice (Figure 2.1E). In contrast, Tnf−/−

and Tnfr1−/−

mice exhibited lower serum

ALT and a lack of caspase activation, revealing that TNF signaling sensitizes to

fulminant hepatitis in vivo.

2.3.2 Delay of Fas-induced apoptosis in Timp3−/−

livers

A hallmark of fulminant hepatic failure, often lethal in the clinical setting, is the

rapid induction of hepatocyte apoptosis [252]. Metalloproteinases and their inhibitors

may orchestrate the onset of acute tissue damage by regulating cytokine bioavailability in

the microenvironment. Thus hepatic expression of selected TIMP, ADAM and MMP

genes was assayed using qRT-PCR following administration of Jo-2 to wildtype mice.

Timp1 and Timp3, but not Timp2 or Timp4, were significantly up-regulated subsequent to

Jo-2 injection (Figure 2.4A). Of the Adam (Adam9, 10, 12, 17) and Mmp (Mmp 2, 9, Mt1-

mmp) genes evaluated, Adam12 and Mmp2 expression changed upon Jo-2 administration,

but the rest remained comparable between PBS and Jo-2 treated mice (Figure 2.2B).

Timp3−/−

mice have increased TNF bioactivity in other models of hepatic inflammation

[69, 76, 253], we thus hypothesized that they would also display enhanced susceptibility

to Jo-2. Surprisingly, Timp3−/−

mice exhibited a similar resistance to that observed in

Tnf−/−

and Tnfr1−/−

mice and survived significantly longer than wildtype controls after

Jo-2 administration (Figure 2B, P=0.005, log rank test). Negligent serum ALT and the

absence of tissue damage (H & E) 3 hours after Jo-2 administration confirmed

diminished hepatotoxicity in Timp3−/−

mice (Figure 1.4C, D).

Page 73: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

53

Figure 2.2 – Caspase 3/7 activation in TNF and TNFR1 deficient hepatocytes, hepatic

Adam and Mmp expression following Jo-2 administration, and decreased caspase

activation in Timp3

mice.

(A) Caspase 3/7 activation as a measure of apoptosis in WT, Tnf

−/− and Tnfr

−/− hepatocytes treated with

1ng/ml TNF + 10ng/ml Jo-2 over 24 hours. *P<0.05 vs. WT. Data are mean ± S.D. (n=3). (B) qRT-PCR

analysis of indicated Adam and Mmp mRNA in livers of wildtype mice treated with PBS or 0.65μg/g Jo-2.

Mice were sacrificed 3 hours after treatment and liver mRNA was obtained. *P<0.05 ± S.D. (n=3). (C)

Caspase activity measured by cleavage of fluorogenic substrates specific to Caspase 3/7 (Ac-DEVD-AMC)

and Caspase 9 (Ac-LEHD-AMC). WT and T3−/−

mice were treated with PBS or 0.65μg/g Jo-2 and

sacrificed 3 hours later.

Page 74: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

54

Figure 2.3 Histology of liver damage upon Jo-2 treatment across all genotypes.

H & E sections of livers obtained from mice injected with PBS of Jo-2 for the indicated timepoints.

Endpoint = Liver tissue from survival timecourse of all genotypes in Figure 2.9A. Images are representative

of at least 3 mice per genotype and condition. T3 −/−

/Tnf −/−

and T3 −/−

/Tnfr1 −/−

compound knockout mice

exhibit drastically less liver damage compared to all other genotypes at endpoint, as the experiment was

terminated at 24h. Bar = 100 μm.

Page 75: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

55

Hepatocytes utilize the intrinsic pathway of apoptosis, requiring a loss of

mitochondrial membrane potential induced by pro-apoptotic members of the Bcl-2 family

prior to activation of downstream caspases 9 and 3 [237, 241]. Immunohistochemical

staining revealed widespread caspase 3 activation in wildtype but not Timp3−/−

livers at

this timepoint (Figure 1.4E). Processing of caspases 8 and 9 was also found to be absent

in Timp3−/−

livers (Figure 1.4F), and additionally the elevation of Bim supported

activation of intrinsic apoptosis only in wildtype livers (Figure 2.4F). Fluorogenic

caspase 3 and 9 activity assays confirmed their functional activation in wildtype mice

(Figure 2.2C). A potential mechanism explaining the dampened pro-apoptotic response in

Timp3−/−

livers could be altered expression and shedding of Fas itself. Immunoblots of

liver lysates from wildtype and Timp3−/−

mice treated with PBS or Jo-2 revealed no

differences in Fas protein expression (Figure 2.5A). Additionally, analysis of cell surface

expression on untreated hepatocytes using flow cytometry showed comparable levels of

Fas in wildtype and Timp3−/−

mice (Figure 2.5B). These findings indicate that the

hepatoprotection observed in Timp3−/−

mice is not likely due to changes in Fas levels.

Overall, diminished caspase activation and a lack of Bim EL induction demonstrated

delayed hepatocyte apoptosis in Jo-2 treated mice lacking TIMP3.

2.3.3 TNFR1 shedding dampens JNK phosphorylation and NF-B

activation in Timp3−/−

liver

We next asked whether the kinetics of apoptosis and TNF signal transduction

Page 76: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

56

Figure 2.4Delay of Fas-mediated apoptosis and hepatotoxicity in Timp3 −/−

mice.

(A) Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) analysis of Timp1, Timp2,

Timp3 and Timp4 mRNA shows elevated induction of Timp1 and Timp3 in livers of wildtype mice treated

with 0.65μg/g Jo-2. Liver mRNA was obtained 3 hours after injection *P<0.05. Data are mean ± S.E.M.

(n=3). (B) Survival curve of WT and Timp3 −/−

(T3 −/−

) mice treated with 0.65μg/g Jo-2. P<0.005 vs. WT

(log rank test). (C-F) WT and T3 −/−

mice were treated with PBS or 0.65μg/g Jo-2. Three hours after

injection, mice were sacrificed to obtain liver and serum. (C) Hepatotoxicity measured by serum ALT

levels in WT and T3 −/−

mice. *P<0.05. Data are mean ± S.E.M. (n=4). (D) Histology (H & E) of WT (from

Figure 2.1C) and T3−/−

livers 3 hours after treatment with PBS or Jo-2 depicts absence of tissue damage,

hemorrhaging and apoptotic bodies in T3−/−

liver. Bar = 100μm. (E) Immunohistochemical staining of

active caspase 3 (arrows). T3−/−

liver exhibits lack of caspase 3 activation. Sections are representative of at

least 4 mice per genotype and treatment. Bar = 100μm. (F) Immunoblot analysis of caspase 8, 9 and 3

processing and Bim EL induction in liver lysates. All data are representative of at least three independent

experiments.

Page 77: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

57

Figure 2.5 – Comparable Fas expression in WT and T3

liver.

(A) Immunoblot of Fas on liver lysates obtained from WT and T3

−/− mice treated with PBS, 3 hours after

Jo-2 treatment and at survival endpoints after Jo-2 treatment.

(B) Flow cytometry of cell surface Fas on WT and T3 −/−

hepatocytes perfused from untreated mice. Cells

are gated on viable (PI-negative) populations and representative of three mice per genotype.

Page 78: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

58

Figure 2.6 – PARP cleavage in primary hepatocytes, Adam17, Tnfr1 and Tnfr2

expression in liver following Jo-2 treatment.

(A) Immunoblot of caspase 3 target PARP in WT and T3

−/− hepatocytes treated with 1ng/ml TNF + 10

ng/ml Jo-2 over 24 hours. (B) Expression of indicated mRNAs in WT and T3−/−

liver lysates 3 hours after

PBS (-) or 0.65 μg/g Jo-2 treatment (+). *P<0.008. Data are mean ± S.E.M. (n=3), expressed relative to WT

mice injected with PBS, and normalized against Actb (β-actin).

Page 79: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

59

were altered in a TIMP3 deficient state. A 24-hour apoptosis timecourse of wildtype and

Timp3−/−

hepatocyte cultures treated with 1ng/ml TNF + 10ng/ml Jo-2 showed

diminished cleavage of Poly-ADP ribose polymerase (PARP) from 3 to 12 hours in

wildtype hepatocytes compared to Timp3−/−

cells (Figure 2.6A). PARP is a physiological

target of activated caspase 3; and we observed significantly reduced caspase 3/7 activity

in Timp3−/−

cells across all timepoints (Figure 2.7A). Of the two TNF receptors, TNFR1

associates with TRADD and FADD to initiate caspase activation, and we thus asked

whether the transient hepatoprotection observed in the absence of TIMP3 correlated with

TNF receptor shedding. ELISA specific to the extracellular domains of TNFR1 and

TNFR2 in hepatocyte culture media over the 24-hour apoptosis timecourse showed

significantly greater TNFR1 release by Timp3−/−

hepatocytes compared to wildtype, but

only modest release of TNFR2 (Figure 2.7B). Fas and Fas ligand were undetectable in

hepatocyte culture media as well as in mouse serum across all treatments in both

genotypes (data not shown). Activation of the stress-kinase JNK is an important

component of TNFR signaling during stress-response. Phosphorylated JNK promotes

intrinsic apoptosis by activating the pro-apoptotic BH3-only protein Bim and degrading

anti-apoptotic Mcl-1 [237, 243, 254-256]. In accordance with elevated TNFR1 shedding,

early JNK phosphorylation was ablated in Timp3−/−

hepatocytes 3 hours after treatment,

although it was comparable at 12 and 24 hours (Figure 2.7C). JNK phosphorylation is

known to occur in transient and prolonged phases, with each having distinct

consequences on cell survival [257-259]. Here we see that loss of Timp3 ablates the early

Page 80: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

60

Figure 2.7Decreased hepatocyte apoptosis correlates with enhanced TNFR1 shedding

and abrogated TNF signaling upon loss of Timp3.

(A) 24-hour timecourse of apoptosis measured by caspase 3/7 activity in WT and T3

−/− primary

hepatocytes treated with 1ng/ml TNF and 10ng/ml Jo-2. *P<0.03. Data are mean ± S.D. (n=3). (B) ELISA

of TNFR1 and TNFR2 release into hepatocyte culture media from (A). *P<0.002 vs WT. Data are mean ±

S.D. (n=3). (C) Immunoblots of JNK phosphorylation in hepatocyte lysates from (A), n.s. = non-specific.

(D) ELISA of serum TNFR1 levels in WT and T3 −/−

mice before (0 hrs) and after 0.65μg/g Jo-2 treatment.

*P=0.003. Data are mean ± S.E.M. (n=4). (E) Immunoblots of JNK phosphorylation in liver lysates from

WT and T3 −/−

mice 3 hours after treatment with PBS or 0.65μg/g Jo-2. (F) NF-κB binding activity was

assayed in nuclear lysates of WT and T3 −/−

mouse livers from (E) using EMSA. (G) qRT-PCR analysis of

pro-inflammatory cytokine mRNA levels in WT and T3−/−

livers from (E). *P<0.01. Data are mean ±

S.E.M. (n=3). All data are representative of at least two independent experiments.

Page 81: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

61

transient phase while maintaining prolonged activation. Next, in vivo analysis of TNFR1

and 2 shedding and JNK phosphorylation was performed. Constitutively higher serum

levels of TNFR1 (Figure 2.7D) but not TNFR2 (data not shown) were seen in Timp3−/−

mice, along with diminished JNK phosphorylation in Timp3−/−

livers 3 hours after Jo-2

injection (Figure 2.7E). These data indicate that loss of TIMP3 increased TNFR1

shedding and dampened early stress-kinase activation in vitro and in vivo.

In addition to the stress-kinases, NF-B activation downstream of TNFR1 is

observed in numerous models of hepatotoxicity [260, 261]. Electrophoretic mobility shift

assay (EMSA) detected active NF-B in hepatic nuclear fractions of wildtype mice

treated with Jo-2, but not in mice lacking TIMP3 (Figure 2.7F). Consistent with this,

significantly higher induction of pro-inflammatory target genes IL-1, IL-6 and TNF

was observed in wildtype livers compared to Timp3−/−

tissue (Figure 2.7G). We also

measured hepatic TNFR1 and 2 expression, which was comparable across both genotypes

before and after Jo-2 treatment (Figure 2.6B). Basal ADAM17 expression measured by

qRT-PCR was found to be higher in Timp3−/−

liver as we have previously reported [76]

(Figure 2.6B). Together, TIMP3 deficiency ablated TNF signaling in response to hepatic

death-receptor activation, as made apparent by diminished JNK phosphorylation and NF-

B activity. Further, this abrogation was not due to reduced expression of TNF receptors

but arose from increased ectodomain shedding of TNFR1.

Page 82: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

62

2.3.4 Signaling through AKT or AMPK does not contribute to

hepatoprotection in Timp3−/−

mice

Pro-survival signaling through AKT is reported to inhibit Mcl-1 degradation by

GSK3 and JNK1/2 [256, 262]. We therefore assayed AKT phosphorylation upon

treatment with Jo-2. AKT phosphorylation was comparable between wildtype and

Timp3−/−

livers treated with and without Jo-2, implying that AKT activation during Fas-

induced hepatotoxicity does not contribute to the survival of Timp3−/−

mice (Figure

2.8A). Metabolic stress in hepatocytes is known to exacerbate tissue response to pro-

inflammatory or cytotoxic insult [253], however, AMPK phosphorylation was also

comparable between wildtype and Timp3−/−

liver lysates after Jo-2 treatment (Figure

2.8B). Altered lipid metabolism through AMPK signaling probably did not account for

the resistance observed in Timp3−/−

mice.

2.3.5 Compound deletions of Timp3/Tnf or Timp3/Tnfr1 completely prevent

hepatic failure and involve enhanced ERK1/2 phosphorylation

To investigate if decreased TNF signaling was the primary mechanism promoting the

survival of Timp3−/−

mice following Fas-mediated hepatotoxicity, we bred Timp3−/−

mice

with Tnf−/−

and Tnfr1−/−

mice. Remarkably, both strains of double knockout mice were

completely resistant to the lethal dose of Jo-2 (Figure 2.9A); histological analysis showed

that the Timp3−/−

/Tnf−/−

and Timp3−/−

/Tnfr1−/−

compound knockouts exhibited little

evidence of tissue damage even at 24 hours (Figure 2.3). Analysis of serum ALT showed

Page 83: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

63

Figure 2.8Comparable AKT and AMPKβ phosphorylation in WT and T3−/−

livers

upon induction of Fas mediated toxicity.

(A) Immunoblot of phosphorylated AKT in liver lysates of WT and T3

−/− mice 3 hours after treatment with

PBS or Jo-2. (B) Immunoblots assaying AKT and AMPKβ phosphorylation in liver lysates 3 hours after

treatment with PBS or Jo-2. Immunoblots are representative of at least 4 mice per condition.

Page 84: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

64

Figure 2.9Compound loss of Timp3 and Tnf or Tnfr1 completely prevents Fas-

mediated hepatotoxicity and reveals accelerated ERK1/2 phosphorylation.

(A) Survival curve of WT, T3

−/−/Tnf

−/− and T3

−/−/Tnfr1

−/− compound knockout mice (dashed lines)

treated with 0.65μg/g Jo-2, superimposed on survival curves of T3 −/−

, Tnf −/−

and Tnfr1 −/−

mice from

Figure 1B and 2C. (B) Hepatotoxicity measured by serum ALT levels in mice of indicated genotypes 3

hours after treatment with PBS or Jo-2. *P<0.05. Data are mean ± S.E.M. (n=3). (C) Immunoblots of JNK,

and ERK1/2 and phosphorylation in liver lysates of indicated genotypes 3 hours after treatment with PBS

or Jo-2. n.s. = non-specific. (D) ELISA of amphiregulin, HB-EGF and TGFα released by WT and T3 −/−

hepatocytes treated with 1ng/ml TNF + 10ng/ml Jo-2 over 24 hours. *P=0.02 vs WT. Data are mean ± S.D.

(n=3). (E) Immunoblots of ERK1/2 phosphorylation in hepatocyte lysates from (C). B, D and E are

representative of at least two independent experiments.

Page 85: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

65

a lack of hepatotoxicity in Timp3−/−

/Tnf−/−

and Timp3−/−

/Tnfr1−/−

compound knockouts 3

hours after Jo-2 treatment when compared to wildtype mice (Figure 2.9B). This implied

the involvement of additional, parallel signals relevant to hepatocyte survival that may be

regulated by TIMP3.

Given the known activation of MAPKs during cytokine-induced stress response,

we assessed JNK, p38 and ERK1/2 phosphorylation in livers of all 6 genotypes with or

without Jo-2 injection. Loss of either TNF or TNFR1 per se eliminated JNK and p38

phosphorylation in response to Jo-2, confirming the importance of TNF signaling via

stress-activated protein kinases in Fas-mediated hepatotoxicity (Figure 2.9C). TIMP3

deficiency did not significantly affect p38 phosphorylation, ruling out a role for this

MAPK in resistance to Fas-mediated hepatotoxicity in Timp3−/−

mice (Figure 2.9C). In

contrast, constitutive ERK1/2 phosphorylation was markedly elevated in untreated livers

of Timp3−/−

mice compared to other genotypes (Figure 2.9C). After Jo-2 treatment,

ERK1/2 phosphorylation was greater in the double knockout livers compared to other

genotypes. Collectively, these data indicate that TIMP3 deficiency activates pathways

upstream of ERK1/2, potentially providing hepatoprotection.

In addition to TNF and its receptors, EGFR ligands (HB-EGF, TGF,

amphiregulin) are critical substrates of ADAM17, which is inhibited by TIMP3 [137,

263, 264]. Adam17−/−

mice phenocopy Egfr−/−

mice, and hepatic deletion of EGFR

results in failed liver regeneration indicating its requirement as a mitogenic stimulus for

hepatocyte division [265]. We therefore analyzed EGFR signaling in wildtype and

Timp3−/−

primary hepatocytes. ELISA was used to measure the shedding of the EGFR

Page 86: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

66

ligands amphiregulin, HB-EGF, and TGF into media over a 24-hour timecourse of TNF

+ Jo-2 treatment. Significantly elevated concentrations of all 3 ligands were found in

Timp3−/−

hepatocyte culture media (Figure 2.9D). Consistent with higher EGFR ligand

release, Timp3−/−

hepatocytes showed accelerated ERK1/2 phosphorylation (Figure

2.9E). These data suggest that enhanced release of multiple EGFR ligands was

responsible for increased EGFR phosphorylation, which in turn activated survival signals

through ERK1/2, contributing to the observed resistance of Timp3−/−

mice to Fas-

mediated hepatotoxicity.

2.3.6 TIMP3 inhibits metalloproteinase-dependent EGFR signaling

Of the 4 Timp genes, only TIMP3 physiologically inhibits ADAM17 activity [76,

264], and we reasoned that EGFR signaling would be checked by TIMP3. We utilized a

previously established assay to measure GPCR-mediated EGFR signaling in a

metalloproteinase-dependent manner [116, 117, 133, 134] as shown in the schematic

(Figure 2.10A). Mouse embryonic fibroblasts (MEFs) were stimulated with Oleoyl-L-α-

lysophosphatidic acid sodium salt (LPA; a GPCR agonist) and the response was assayed

by immunoblotting for phosphorylated EGFR and ERK1/2. Wildtype MEFs showed

increasing ERK1/2 phosphorylation in a dose-dependent manner, while MEFs lacking

ADAM17 showed diminished ERK1/2 phosphorylation under the same treatment

conditions. On the other hand, loss of Timp3 resulted in markedly enhanced ERK1/2

phosphorylation (Figure 2.10B). Phosphorylated ERK1/2 was also seen in the absence of

LPA in TIMP3 deficient MEFs. As controls in this series of experiments, pre-treatment

Page 87: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

67

of all MEFs with the small molecule ADAM inhibitor TAPI-1 abolished ERK1/2

phosphorylation at the highest dose of LPA, while soluble EGF bypassed the requirement

of metalloproteinase activity. Sustained EGFR phosphorylation in Timp3−/−

MEFs

confirmed enhanced signaling owing to elevated metalloproteinase activity (Figure

2.11A). We next tested whether shed EGFR ligands are relevant to receptor activation

through antibody-mediated depletion in culture media. Given that commercial availability

of a murine-specific neutralizing antibody was limited to amphiregulin, Timp3−/−

MEFs

were stimulated with LPA in the presence of neutralizing amphiregulin antibody (AR).

Indeed, pre-treatment with this antibody significantly diminished ERK1/2

phosphorylation indicating that soluble amphiregulin contributed to EGFR signaling

(Figure 2.10C). Thus, TIMP3 is a specific negative regulator of metalloproteinase-

dependent EGFR signaling.

2.3.7 Increased EGFR ligand shedding is hepatoprotective

We next asked whether enhanced hepatic EGFR signaling due to increased ligand

bioavailability was directly protective against Fas-induced cell death. Conditioned media

was collected from wildtype, Adam17−/−

and Timp3−/−

MEFs stimulated with LPA and

subsequently added to wildtype hepatocytes treated with Jo-2 + TNF (Figure 2.10D). As

independent controls, hepatocytes were pretreated with EGF for protection or Erlotinib

(an EGFR receptor tyrosine kinase inhibitor) for sensitization to apoptosis (Figure 2.10D,

white bars). Conditioned media from Timp3−/−

MEFs provided hepatoprotection, whereas

other genotypes had no effect. Since we had observed that shed amphiregulin plays a

Page 88: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

68

Figure 2.10Loss of Timp3 enhances hepatoprotective EGFR ligand shedding and

downstream ERK1/2 phosphorylation.

(A) Schematic of lysophosphatidic acid (LPA)-induced EGFR signaling via the mechanism of “Triple

Membrane Pass Signal”. (B) Immunoblots assaying ERK1/2 phosphorylation in MEFs of the indicated

genotypes treated with listed doses of LPA. TAPI-1 is an ADAM inhibitor; soluble EGF was used as a

positive control. WT and T3 −/−

immunoblots are representative of at least two independently derived

primary MEF lines from different embryos. (C) T3 −/−

MEFs were pre-treated with 5.0μg/ml IgG or

amphiregulin neutralizing antibodies (αAR) 2 hours prior to treatment with 1.0μM LPA for 5 min, and

ERK1/2 phosphorylation was subsequently assayed. (D) Wildtype hepatocytes were pre-treated for 3 hours

with conditioned media from LPA-treated MEFs of the indicated genotypes (colored bars), then treated

with 1ng/ml TNF + 10ng/ml Jo-2 for an additional 3 hours. As controls, hepatocytes were left untreated,

pre-treated with EGF, or with Erlotinib (white bars). *P<0.01 vs. TNF + Jo-2 treated hepatocytes that were

not pre-treated, **P=0.004. Data are mean ± S.D. (n=3). (E) T3 −/−

hepatocytes were pre-treated with

5.0μg/ml IgG or αAR for 3 hours prior to treatment with TNF + Jo-2 as in (D). *P<0.001 vs. no pre-

treatment. Data are mean ± S.D. (n=3). All data are representative of at least two independent experiments.

Page 89: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

69

functional role in EGFR signaling, we tested whether neutralizing soluble amphiregulin

would re-sensitize Timp3−/−

hepatocytes to apoptosis. We observed far greater caspase3/7

activation upon addition of AR during TNF + Jo-2 treatment of Timp3−/−

hepatocytes

(Figure 2.10E). A physiological consequence of EGFR ligand availability during acute

stress is induction of EGFR signaling and subsequent transcription of anti-apoptotic

genes or suppression of pro-apoptotic signals. Bcl family members Mcl-1 and Bcl-xL,

along with xIAP and cIAP1/2 are putative EGFR targets [266-268], and we observe that

loss of TIMP3 results in elevated transcription of these pro-survival genes upon Jo-2

treatment (Figure 2.11B). Additionally, MAP kinase activation downstream of EGFR is

known to antagonize Bim transcription. Consistently, we observe that Timp3−/−

livers

exhibit decreased BimEL levels upon Jo-2 treatment (Figure 2.4F).Therefore, TIMP3

deficiency enhances ectodomain shedding of EGFR ligands, among which amphiregulin

is a significant contributor to hepatoprotection against death receptor activation.

2.3.8 Hepatocyte-specific loss of ADAM17 or EGFR promotes Fas-induced

killing

In order to directly determine the role of ADAM17 and EGFR in Fas-mediated

hepatocyte apoptosis, we generated mice that lack ADAM17 or EGFR in hepatocytes by

crossing Adam17fl/fl

or Egfrfl/f

mice with those expressing Cre recombinase under control

of the Albumin promoter. Primary hepatocyte cultures were generated from control

(Adam17fl/fl

, Egfrfl/f

) and hepatocyte-specific knockout mice (Adam17hep

, Egfrhep

respectively). Significantly elevated caspase 3/7 activation was observed in Adam17hep

Page 90: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

70

hepatocytes at 1 and 3 hours after TNF + Jo-2 treatment (Figure 2.12A). Additionally

TNFR1, amphiregulin, HB-EGF or TGF levels were undetectable over the timecourse

in Adam17hep

culture media (data not shown). As with Adam17hep

cells, Egfrhep

hepatocytes exhibited increased caspase 3/7 activity at early timepoints compared to their

Egfrfl/fl

controls (Figure 2.12B). To confirm the protective role of ADAM17 in vivo,

Adam17hep

and Adam17fl/fl

control mice were injected with a lower dose of Jo-2

(0.33g/g) and monitored for survival over 3 days after treatment. A greater fraction of

Adam17hep

mice died within 3 days compared to Adam17fl/fl

mice (5/7 versus 2/5). Cre-

mediated deletion of ADAM17 or EGFR in hepatocytes was validated via

immunoblotting (Figure 2.12C, D respectively). We conclude that ADAM17 and EGFR

individually provide hepatoprotection against Fas-induced killing.

2.3.9 Inhibitors of MAPK, EGFR or ADAM17 reverse the Timp3−/−

resistance to Fas

To confirm that TIMP3 is functionally upstream of JNK and ERK1/2 signal

transduction pathways, we first examined the in vivo consequence of MAPK inhibition

on hepatotoxicity. Pretreatment of wildtype mice with a JNK inhibitor (SP600125) led to

decreased serum ALT and hepatic caspase 3 activation versus those treated with vehicle

(Figure 2.13A, B). On the other hand, Timp3−/−

mice pretreated with an EGFR inhibitor

(Erlotinib) exhibited elevated serum ALT and hepatic caspase 3 activation upon Jo-2

injection compared to those pretreated with vehicle (Figure 2.13C, D). Next, small

molecule inhibitors of ADAM17 (TAPI-1), JNK (SP600125), ERK1/2 kinase MEKK

Page 91: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

71

Figure 2.11 – Enhanced EGFR phosphorylation in T3

MEFS, increased EGFR target

gene expression in T3

livers.

(A) LPA enhances metalloproteinase-dependent EGFR phosphorylation in mouse embryonic fibroblasts

(MEFs) lacking TIMP3. Immunoblot of phosphorylated EGFR (p-EGFR, Tyr1173) in WT and T3 −/−

MEF

lysates from Figure 5B. (B) qRT-PCR analysis of putative EGFR target genes involved in regulation of

apoptosis. T3−/−

livers exhibit elevated expression of antiapoptotic genes Mcl1, Birc3 (Ciap1/2), Bcl2l1

(Bcl-xl) and Xiap upon Jo-2 treatment. All results are relative to untreated WT liver cDNA, normalized to

Rn18s (18S) levels. *P<0.05, S.E.M. (n=3).

Page 92: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

72

Figure 2.12ADAM17 and EGFR are individually hepatoprotective against Fas-

mediated hepatotoxicity.

(A) Apoptosis timecourse of control (fl/fl) or Adam17 deficient (Δhep) hepatocytes treated with 1ng/ml

TNF + 10ng/ml Jo-2, measured by caspase 3/7 activation. (B) Control (fl/fl) or Egfr deficient (Δhep)

hepatocytes were treated as in (A) and apoptosis measured by caspase 3/7 activation. *P<0.04. Data are

mean ± S.D. (n=3). Apoptosis timecourse of both mutants were repeated in an independent experiment with

consistent results. (C, D) Confirmation of Adam17 and Egfr deletion by immunoblot.

Page 93: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

73

Figure 2.13TIMP3 is an upstream regulator of EGFR and MAPK activity in Fas-

induced hepatotoxocity.

(A) Serum ALT measurements of WT mice pre-treated with the JNK inhibitor SP600125 (hatched) or

vehicle (PBS + 10% DMSO, white) 45 minutes prior to Jo-2 or PBS treatment. *P<0.05. Data are mean ±

S.E.M. (n=4). (B) Immunohistochemistry depicting active caspase 3 only in WT livers of mice pretreated

with DMSO prior to Jo-2 administration. (C) Serum ALT measurements of T3 −/−

mice pre-treated with

Erlotinib (hatched) or vehicle (black) 1 hour prior to Jo-2 or PBS treatment. *P<0.05. Data are mean ±

S.E.M. (n=4). (D) Immunohistochemistry depicting active caspase 3 in T3 −/−

livers pre-treated with EGFR

inhibitor Erlotinib. (E) Hepatocytes were treated with vehicle only (DMSO), inhibitors of ADAM17

(TAPI-1), JNK (SP600125), ERK1/2 kinase MEKK (U0126), EGFR (Erlotinib) for 1 hr followed by

addition of TNF + Jo-2 for 3 hours. Apoptosis was measured as caspase 3/7 activity. *P=0.0002 and

**P<0.002 vs. DMSO. Data are mean ± S.D. (n=3). Data are representative of at least two independent

experiments.

Page 94: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

74

(U0126), and EGFR (Erlotinib) were added to wildtype and Timp3−/−

hepatocyte cultures

treated with TNF + Jo-2 for 3 hours. The inhibition of ADAM17, ERK1/2 or EGFR

activity in Timp3−/−

hepatocytes significantly elevated caspase3/7 activity, while JNK

inhibition in wildtype hepatocytes was clearly protective (Figure 2.13E). Thus, each

small molecule inhibitor was able to reverse the sensitivity to Fas-induced apoptosis in

wildtype and Timp3−/−

genotypes. The data presented here along with Figures 4 and 5

establish TIMP3 as a regulator of MAPK activity during death receptor activation.

Specifically, the loss of TIMP3 shifts the profile of the MAPK cascade, simultaneously

inhibiting JNK activation while enhancing ERK1/2 phosphorylation to ultimately

promote survival.

2.3.10 Adenoviral ADAM17 prevents acute liver failure in drug-induced

toxicity

Hepatotoxicity driven by acetaminophen (APAP) overdose is the most common

cause of death by acute liver failure in patients upon hospitalization [269-272]. Fas

signaling plays a key role in this clinical setting [273-275]. We therefore investigated the

hepatoprotective potential of ADAM17 in wildtype mice treated with a lethal dose of

APAP (600mg/kg, i.p.). Adenoviruses encoding GFP (Ad-GFP) or ADAM17 (Ad-

ADAM17) were administered at 1x109 pfu/ml 96 hours prior to APAP treatment.

Successful expression of ADAM17 was assayed via immunoblotting (Figure 2.14E).

While a significant proportion of Ad-GFP expressing mice (6 of 9) succumbed to APAP-

Page 95: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

75

Figure 2.14 - Ectopic ADAM17 delivery protects against acetaminophen-driven

fulminant hepatitis.

(A) Survival of wildtype mice expressing hepatic Adam17 or Gfp after treatment with a lethal dose of

APAP. Adenoviral ADAM17 (Ad-ADAM17) or GFP (Ad-GFP) was injected intravenously into wildtype

mice 96 hours prior to treatment with 600mg/kg APAP. Survival was followed for the next 72 hours.

*P=0.01 vs. Ad-GFP (log-rank test). (B) Hepatotoxicity measured by serum ALT levels from mice treated

as in (A). *P<0.01. Data are mean ± S.E.M. (n=3). (C) Histological analysis of liver damage at the survival

endpoints. Significant necrosis is observed only in Ad-GFP livers (black arrows). Images are representative

of at least 5 mice. Bars = 100μm. (D) Immunohistochemical staining of active caspase 3 (black arrows)

shows induction of apoptosis in Ad-GFP livers after APAP treatment. Images are representative of 3 mice

per group. Bars = 100μm. (E) Immunoblots of ADAM17 in liver lysates confirming delivery of adenoviral

ADAM17 to the liver. Immunoblots for cleaved caspase 8 and 3 depicts onset of APAP-induced apoptosis

in Ad-GFP mice.

Page 96: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

76

induced hepatotoxicity, all mice expressing Ad-ADAM17 (10 of 10) survived APAP

treatment (Figure 2.14A, *P=0.013, log rank test). Measuring hepatotoxicity by serum

ALT and histological examination of the two groups of mice revealed that Ad-ADAM17

provided significant protection against APAP overdose (Figure 2.14B, C). Extensive

necrosis, cellular and structural damage was only observed in the livers of Ad-GFP

expressing mice at survival endpoint (Figure 2.14C). Next induction of apoptosis was

measured via immunohistochemical staining for active caspase3 and immunoblotting for

caspases 8 and 3. We see that delivery of adenoviral ADAM17 protects the liver against

APAP-induced caspase activation (Figure 2.14D, E.) Cumulatively, these findings

strikingly demonstrate that ADAM17 delivery prevents liver failure in clinically relevant

acute hepatitis that relies on Fas signaling.

2.4 Discussion

Induction of stress signaling by Fas causes fulminant hepatitis, where the

combined cytokine and growth factor availability in the microenvironment contributes to

promoting or preventing cell death. This challenge engages multiple pathways and cell

surface proteolysis presents an attractive interface to integrate them. We show that

individually TNF, TNFR1 and TIMP3 sensitize the liver to Fas-mediated hepatotoxicity,

and that ADAM17 and EGFR protect against this insult. TIMP3 is the sole physiological

inhibitor of ADAM17, and Timp3−/−

mice highlight how ADAM17-mediated ectodomain

shedding simultaneously regulates TNF and EGF signaling during a stress response.

Enhanced TNFR1 shedding ablates pro-apoptotic JNK phosphorylation and caspase

Page 97: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

77

cleavage in Timp3−/−

mice. The complete protection observed in compound TIMP3 and

TNF/TNFR1 mutant mice treated with Jo-2 further reveals a significant role for ERK1/2-

mediated survival signaling in hepatoprotection. We identify a novel function of TIMP3

in negative regulation of EGFR signaling, where Timp3−/−

cells exhibit elevated release

of EGFR ligands amphiregulin, TGF and HB-EGF, constitutively enhancing EGFR and

ERK1/2 phosphorylation. Together these data are the first demonstration that ectodomain

shedding serves a powerful function in modulating MAPK cascades during acute hepatic

stress.

2.4.1 Ectodomain shedding of TNF is a crucial step in hepatotoxicity

In vivo hepatotoxicity manifests upon viral infection, chronic inflammation or

fulminant hepatic failure [252, 272]. Given the crucial function of the liver in host

defense, hepatocytes are highly sensitive to cell-extrinsic cues such as TNF release by

activated resident macrophages (Kupffer cells). Recent work by a number of groups has

revealed that activators of NF-B, specifically the IKK/NEMO regulatory subunit can

dictate the basal metabolic state of hepatocytes and consequently their capacity to convert

extracellular cues into cell-intrinsic signal transduction during chronic and acute

inflammation, death receptor activation and infection [276-278]. In addition to the shifted

MAPK activation, we observed significantly blunted NF-B activation and pro-

inflammatory target gene expression in livers of Timp3−/−

mice that were treated with Jo-

2. Consistent with our observations, Beraza et al [276] report that hepatocyte-specific

deletion of NEMO protects mice from Jo-2 induced fulminant hepatitis, corroborated

Page 98: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

78

with decreased stress-kinase activation and pro-inflammatory gene induction.

Independent reports demonstrate that deletion of IKK, NEMO or RelA/p65 promotes

inflammation-driven hepatocarcinogenesis and sensitizes mice to TNF-induced

hepatocyte damage [279, 280]. Along with this current study, our previous findings that

Timp3−/−

mice are more sensitive to LPS-induced septic shock and fail to regenerate liver

mass upon partial hepatectomy support the hypothesis that when left unchecked,

ectodomain shedding deregulates TNF signaling in the liver [69, 76, 263].

2.4.2 TIMP3 regulates pro-survival and pro-apoptotic signaling in

hepatocytes

As modeled in Figure 9, the current study shows that in addition to impacting TNF

signaling, ADAM17 activity in hepatocytes is essential for EGFR ligand release and

promotes their paracrine and/or autocrine survival signaling. Engagement of TNFR1

recruits stress kinases and NF-B, which in turn upregulate pro-apoptotic mediators such

as Bim and caspases [281-283](Figure 9C). Meanwhile, EGFR activation acts in an

opposing manner to promote cell survival by induction of anti-apoptoic genes and

suppression of Bim [266-268, 284, 285] (Figure 9D). Therefore the protective mechanism

of ADAM17 mediated EGFR activation may be to increase EGFR ligand availability,

which in turn induces gene transcription of pro-survival factors. TIMP3 acts to regulate

these opposing pathways by inhibiting the shedding of TNFR1 and EGFR ligands (Figure

9E). The physiological role of ADAM17 in TNF-induced hepatitis was confirmed by

Horiuchi et al [286], where Mx1-Cre driven loss of ADAM17 in both myeloid cells and

Page 99: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

79

hepatocytes or LysM-Cre driven loss in myeloid cells conferred resistance to endotoxin-

shock by abrogating the delivery of TNF. We propose that when TNF acts as a secondary

sensitizer, the availability of TNF receptors and parallel activation of other pathways

dictate the extent of hepatotoxicity. Significantly, blocking EGFR signaling by the

depletion of amphiregulin in fibroblasts and hepatocytes, or small molecule inhibition of

EGFR and ERK1/2 reversed the protection provided by greater ectodomain shedding. In

addition to genetic models, our in vivo experiments conducted with these inhibitors

confirmed that countering JNK phosphorylation alleviates hepatotoxicity, while blocking

EGFR activation re-sensitizes Timp3−/−

mice. Overall the stress response to increased

TNF superfamily ligands may depend on the repertoire of cell surface receptors available

to transmit these signals and additional factors in the tissue microenvironment that

promote or counteract death receptor activation.

TRAIL is also reported to promote Fas-mediated hepatotoxicity through JNK and

Bim in primary hepatocytes and the liver [243]. We provide physiological evidence here

that TNF itself operates in a similar manner in vivo. Since TNF and TRAIL can activate

opposing signal transduction through FADD and NF-B, the parallel induction of yet

other pathways can tip the cellular response to survival or death. The pro-survival role of

AKT has been well documented, and its capacity to antagonize JNK may contribute to

protection [256, 287]. Our probing of AKT activity led us to conclude that Timp3−/−

cells

do not utilize this pathway during hepatotoxicity since no increase in phosphorylated

AKT was observed in Timp3 deficient tissues subjected to apoptotic stress. Our results

provide insight into EGFR activation and ERK1/2 signal transduction in a TIMP3

Page 100: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

80

Figure 2.15Inhibition of cell surface ADAM17 impacts MAP kinases during stress

responses and provides stromal regulation of cell survival.

(A) Fas-activation triggers intrinsic apoptosis. (B) Macrophage (Kupffer cell) activation results in TNF

shedding, which acts as a secondary sensitizer to Fas-mediated toxicity. (C) TNF/TNFR1 binding activates

both stress kinases (JNK) and NF-κB. ADAM17 shedding of TNFR1 dampens pro-apoptotic signaling. (D)

ADAM17 shedding of EGFR ligands provides protection from apoptosis through ERK1/2 signaling. (E)

The hepatic microenvironment harbors TIMP3, which checks ADAM17 activity.

Page 101: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

81

deficient state, which commits hepatocytes to survival following Fas-mediated toxicity.

In support of this interpretation, the protective role of the EGFR pathway is demonstrated

in models of stress induced by UV and ROS [117, 288], and a pro-apoptotic role of

TIMP3 in Fas-mediated apoptosis has been suggested in neuronal and synovial fibroblast

models in vitro [158, 289]. Our finding that dampened TNFR1 activation coupled with

enhanced EGFR signaling is protective against acute hepatic failure is consistent with

observations that individually, ablating JNK signaling or elevating EGFR activation can

protect, at least in part, against receptor-mediated apoptosis [243, 245]. While pro-

apoptotic and pro-survival kinases can operate in isolation, it is unlikely that this is the

case during physiological stress. The reported intracellular crosstalk between JNK and

ERK1/2 is another level at which the apoptotic response can be shifted [290, 291]. Our

observations are within the context of death-receptor activation during acute stress,

however the coordination of opposing signal transduction pathways applies to numerous

aspects of hepatic homeostasis which are out of the scope of the current study. A recent

review by Michalopoulos discusses the unknowns of coordinating survival and death

signals during liver regeneration and importantly liver failure upon loss of this regulation

[292].

Fulminant hepatitis is a significant cause of drug-induced liver failure and also

contributes to the hepatotoxicity observed in steatosis and end stage liver disease. Our

findings show that ectodomain shedding promotes hepatocyte survival in this acute

setting by the dual mechanism of enhanced EGFR-ERK1/2 activation and dampened

Page 102: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

82

TNFR1 signaling through JNK and NF-B. We further demonstrate that adenoviral

delivery of ADAM17 is capable of protecting mice from acetaminophen-induced liver

failure. Even though unchecked metalloproteinase activity is implicated in chronic

hepatic inflammation, inducing transient ectodomain shedding may be an attractive

strategy to limit toxicity during fulminant liver failure.

Page 103: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

83

2.5 Methods

2.5.1 Mice

All mice used in this study were of C57BL/6 background. Male mice aged 9-12

weeks were used for all the experimental procedures. Timp3−/−

mice have been

previously described [76], and Tnf−/−

or Tnfr1−/−

mice were obtained from the Jackson

Laboratory. These strains were crossed with Timp3−/−

mice to generate Timp3−/−

Tnf−/−

and Timp3−/−

Tnfr1−/−

mice. Hepatocyte-specific knockouts were produced by crossing

mice expressing Cre recombinase under the Albumin promoter (Jackson Labs) with

Adam17fl/fl

or Egfrfl/fl

mice. Genotypes were confirmed via PCR for the respective genes;

primer sequences are available upon request. Mice were fed 5%-fat chow ad libitum,

housed and cared for in accordance with the guidelines approved by the Canadian

Council for Animal Care, and the Animal Care Committee of the Ontario Cancer

Institute.

2.5.2 Fas-induced hepatotoxicity

Mice were injected intraperitoneally with Fas-agonist antibody (CD95, clone Jo-

2, NA/LE) in 100µL sterile PBS to trigger death receptor activation. Since Jo-2 exhibits

lot-to-lot variation in its potency of hepatocyte killing, we tested three lots and identified

BD pharmingen 554254 at 0.65µg/g to cause morbidity within 5-7 hours in wildtype

mice. Control mice were injected with 100µL sterile PBS. For JNK inhibition

experiments, wildtype mice were injected intraperitoneally with 0.25µg/g SP600125

(Calbiochem) 45 minutes prior to Jo-2 injection. For EGFR inhibition experiments,

Page 104: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

84

Timp3−/−

mice were injected intraperitoneally with 0.50µg/g Erlotinib (generously

provided by Dr. Ming-Sound Tsao) 1 hour prior to Jo-2 injection. PBS containing 10%

DMSO was used as a vehicle. Animals were sacrificed by CO2 asphyxiation at designated

time points for analysis. Survival curves were obtained based on the time at which

animals became pre-moribund. Hepatotoxicity was determined by measuring serum ALT

levels.

2.5.3 APAP-induced hepatotoxicity and adenoviral delivery of ADAM17

To obtain an optimal proportion of adenovirus-infected hepatocytes, a dose

titration of Ad-GFP (Vector Biolabs) was performed in wildtype mice at 1x107, 1x10

8

and 1x109 pfu/ml dissolved in 500

9 pfu/ml of Ad-

GFP showed a significantly higher proportion of GFP-positive hepatocytes compared to

1x107 or 1x10

8 pfu/ml titers. Thus 1x10

9 pfu/ml Ad-GFP and Ad-ADAM17 were

dissolved in sterile PBS and delivered intravenously in the tail veins of 12-week old male

mice. 96 hours after injection of virus, mice were starved for 18 hours prior to

acetaminophen injections (APAP; Sigma A5000). APAP was dissolved in sterile PBS

and warmed to 55oC to dissolve. 600mg/kg dosed in 200l PBS was intraperitoneally

injected into mice. Survival curves were obtained based on the pre-moribund status of

animals, at which point they were sacrificed. Liver, kidney and serum were obtained for

subsequent analyses.

2.5.4 Primary hepatocyte culture and apoptosis assays

Page 105: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

85

Primary hepatocytes were prepared by perfusing the portal vein of the liver with

0.02mg/ml Liberase (Roche) and enriched using a 10% Percoll (Sigma) gradient.

Hepatocytes were cultured in William‟s E medium containing 10% fetal bovine serum,

2mM L-glutamine, 0.1U/ml insulin, 1.0mM dexamethasone and antibiotics on 6-well

plates coated with 2.5mg/ml type I rat-tail collagen. Hepatocytes (1x106/well, 6-well

plate) were serum-starved for 24 hours, then treated with 10ng/ml Fas-agonist antibody

(Jo-2, BD Biosciences) in the presence or absence of 1ng/ml recombinant mouse TNF (R

& D Systems) for the indicated timepoints. Media and cell lysate were collected for

analysis. Hepatocytes were co-incubated with small molecule inhibitors to ADAM17

(10µM TAPI-1, Peptides International), JNK (25µM SP600125, Calbiochem), EGFR

(100nM Erlotinib) and MEKK (10µM UO-126, Sigma) where indicated. For conditioned

media (CM) experiments, culture media was obtained from MEFs of indicated genotypes

that were stimulated with 1.0M LPA for 1 hour. Wildtype hepatocytes were incubated

with CM 3 hours prior to addition of 1ng/ml TNF + 10ng/ml Jo-2. CM was maintained in

a 1:1 dilution with hepatocyte media containing TNF + Jo-2 during induction of

apoptosis for another 3 hours, after which cell lysate was collected for analysis. Timp3−/−

hepatocytes were incubated with 5.0g/ml IgG or Amphiregulin neutralizing antibody

(AF989, R & D Systems) 3 hours prior to addition of 1ng/ml TNF + 10ng/ml Jo-2.

Antibodies were maintained in hepatocyte culture media during induction of apoptosis

for another 3 hours. Cell lysate was collected for caspase3/7 activity analysis.

2.5.5 LPA treatment of MEFs

Page 106: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

86

Primary murine embryonic fibroblasts (MEFs) were generated as described [83]

and cultured in Dulbecco‟s Modified Eagle Medium with 10% fetal bovine serum and

antibiotics. MEFs were cultured until 80% confluent and starved for 48 hours prior to

treatment. Cells were incubated for 30 minutes with 0.03M, 0.1M or 1.0M Oleoyl-L-

α-lysophosphatidic acid sodium salt (LPA, Sigma L7260) dissolved in sterile water.

MEFs were incubated with 50ng/ml recombinant human EGF (BD Biosciences) for 15

minutes as positive controls of EGFR phosphorylation and ERK1/2 phosphorylation.

Where indicated, MEFs were pre-incubated with 10µM TNF Alpha Protease Inhibitor-1

(TAPI-1, Peptides International) or 1µM Erlotinib dissolved in DMSO for 1 hour, then

incubated with the indicated concentrations of LPA for 15 minutes in the presence of

inhibitor. Two independent fibroblast lines from wildtype embryos and four lines from

Timp3−/−

embryos were stimulated with LPA as described to measure EGFR signaling.

For amphiregulin neutralization experiments, Timp3−/−

MEFs were pre-treated with

50.0g/ml IgG or amphiregulin neutralizing antibody (R & D Systems) for 2 hours. Cells

were then treated with 1.0M LPA for an additional 5 minutes. Media and cell lysate

were collected for analysis.

2.5.6 Immunoblotting

Total liver protein was extracted by mortar and pestle homogenization of frozen

tissue. RIPA extraction buffer containing 25mM Tris-HCl pH 7.6, 1% Triton X-100,

0.1% SDS, 1% NP-40, 1% sodium deoxycholate, 5mM EDTA, 50mM NaCl, 200µM

Na3VO4, 2mM PMSF, and appropriate dilution of Complete Mini, EDTA-free protease

Page 107: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

87

inhibitor cocktail tablets (Roche) was used to lyse all tissue and cells, and lysate was

stored at -70oC. 15-50g of protein was loaded on SDS-PAGE gels for western blotting.

The following anti-mouse antibodies were used: anti-phosphorylated JNK (p-JNK,

Thr183/Tyr185), anti-JNK , anti-phosphorylated p44/42 MAP kinase (p-ERK1/2,

Thr202/Tyr204), anti-p44/42 MAP kinase (ERK1/2), anti-p38 MAPK, anti-

phosphorylated p38 MAPK (p-p38, Thr180/Tyr182), anti-EGFR, anti-phosphorylated

EGFR (p-EGFR, Tyr1173), anti-AMPK, anti-phosphorylated AMPK (p-AMPK,

Ser108), anti-AKT, anti-phosphorylated AKT (p-AKT, Ser473), anti-Caspase 3, anti-

Caspase 9 (all from Cell Signaling), anti-Caspase 8 (R & D Systems), TACE/ADAM17

specific for the disintegrin domain (Khokha Lab), and anti--Actin (Santa Cruz).

2.5.7 Caspase activity assays

Fluorometric caspase activity assays were performed using 15g of protein and

20M fluorescent substrates (Caspase 3/7: Ac-DEVD-AMC, Caspase 9: Ac-LEHD-

AMC, AnaSpec Inc.). Protein and substrates were incubated for 60 min and fluorescence

emission measured at 352 nm every 5 minutes for 1 hour to obtain kinetic measurement

of enzymatic activity.

2.5.8 Electrophoretic Mobility Shift Assay (EMSA)

NF-B activity in liver tissue was determined by EMSA using a common NF-B

biotinylated nucleic acid probe (Panomics) on nuclear fractions of livers. Briefly, liver

tissue was homogenized in Buffer A (10mM HEPES pH 7.9, 1.5mM MgCl2, 10mM KCl,

Page 108: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

88

protease inhibitors) and centrifuged to isolate nuclear pellet. The pellet was resuspended

in Buffer B (20mM HEPES pH 7.9, 1.5mM MgCl2, 420mM NaCl, 0.2mM EDTA, 25%

v/v glycerol, protease inhibitors). After centrifugation, the supernatant containing nuclear

extract was collected and stored at -70oC. EMSA was performed according to supplier‟s

protocol (Panomics).

2.5.9 Enzyme-Linked ImmunoSorbent Assay (ELISA)

For serum ELISA, blood was collected from mice by cardiac puncture upon

sacrifice at indicated timepoints. Serum was isolated from whole blood by centrifugation

(BD Vaccutainer CPT cell preparation tube). Serum was diluted 1:200 for TNFR1 and

TNFR2 ELISA, and 1:10 for Fas, Fas ligand, and TNF. Undiluted media was used for

assays on cell culture experiments. ELISA was performed as per manufacturers‟

instructions (TNFR1, Fas, Fas ligand, Amphiregulin, HB-EGF, TGF from R & D

Systems; TNFR2, TNF from BD Pharmingen).

2.5.10 Histology & Immunohistochemistry

Liver lobes were fixed in 5% formalin for 24 hours, transferred to PBS and

processed for embedding in paraffin. 5-µm sections were placed on Superfrost/Plus

microscope slides (Fisher Scientific) and processed as previously described [76]. The

following stains were applied for immunohistochemical analysis: Harris‟ hematoxylin

(Electron Microscopy Sciences), Eosin (Fisher Scientific). Anti-Cleaved Caspase 3

antibody (Cell Signaling) was used according to manufacturer‟s specifications.

Page 109: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

89

2.5.11 RNA preparation and quantitative RT-PCR

RNA was prepared from frozen liver tissue using TRIzol reagent (Invitrogen)

according to manufacturer‟s instructions. cDNA was obtained using a first-strand cDNA

synthesis protocol (Promega). Gene expression was measured using SYBR Green reagent

(Quanta) or Taqman primer/probesets (Applied Biosystems) in a 7800HT Real-time PCR

system (Applied Biosciences). All gene expression levels were normalized to -actin

(SYBR Green) or 18S (Taqman) and fold-change measured relative to control wildtype

samples. qPCR of RNA was used as a negative control. Amount of each product was

calculated using the 2-CT

method. Primer sequences and Taqman primer/probeset IDs

are provided in Table 2.1

2.5.12 Statistical Analyses

Data are reported as mean ± S.D. or S.E.M. All calculations were carried out

using GraphPad Prism software (GraphPad Software). Comparisons were made by two-

tailed Student‟s t-test and analysis of variance; comparisons between Kaplan-Meier

survival curves were made by log-rank test.

Table 2.1Primer sequences used for quantitative RT-PCR analysis.

Gene Forward & Reverse Primer Sequence

Timp1 F: 5‟-CATGGAAAGCCTCTGTGGAT-3‟

R: 5‟-CTCAGAGTACGCCAGGGAAC-3‟

Timp2 F: 5‟-GTCATTGCTGCCTTCCTCTC-3‟

R: 5‟-AAAGGGGTGAAGAATGGCTT-3‟

Timp3 F: 5‟-AAACATCTGCCTGGGTTCAG-3‟

R: 5‟-CAAGCTTCCAGCCAAACTTC-3‟

Page 110: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

90

Timp4 F: 5‟-ACCTCCGGAAGGAGTACGTT-3‟

R: 5‟-TTATCTGGCAGCAACACAGC-3‟

Adam9 F: 5‟-TTGCATCCATTGTTGCTCAT-3‟

R: 5‟-CTTCTCAAAGTCCTCCGCAC-3‟

Adam10 F: 5‟-GCAACATCTGGGGACAAACT-3‟

R: 5‟-TTGCACTTGTCACTGTAGCC-3‟

Adam12 F: 5‟-AGAGAAAGGAGGCTGCATCA-3‟

R: 5‟-GCCTGCTTGACCTCTGGTAG-3‟

Adam17 F: 5‟-AGGATGCTTGGGATGTGAAG-3‟

R: 5‟-CTGTTTGCTCTGGGAGAACC-3‟

Mmp2 F: 5‟-ACTGACACTGGTACTGGCCC-3‟

R: 5‟-GTCACGTGGTGTCACTGTCC-3‟

Mmp9 F: 5‟-CCCGCTGTATAGCTACCTCG-3‟

R: 5‟-CTGTGGTTCAGTTGTGGTGG-3‟

Mmp14 F: 5‟-CTGTCCCAGATAAGCCCAAA-3‟

R: 5‟-GCATTGGGTATCCATCCATC-3‟

Il1b F: 5‟-AAGCCTCGTGCTGTCGGACC-3‟

R: 5‟-CCAGCTGCAGGGTGGGTGTG-3‟

Il6 F: 5‟-TAAGCTGGAGTCACAGAAGGAGTGGC-3‟

R: 5‟-AGGCATAACGCACTAGGTTTGCCGA-3‟

Tnf F: 5‟-ATCCGCGACGTGGAACTGGC-3‟

R: 5‟-AGAAGAGCGTGGTGGCCCCT-3‟

Tnfrsf1a (Tnfr1) F: 5‟-AGCATGCTGGAAGCCTGGCG-3‟

R: 5‟-TGGACGAGGGGGCGGATTT-3‟

Tnfrsf1b (Tnfr2) F: 5‟-TTCCCAAGCCAGCGCCACAG-3‟

R: 5‟-GCCAGCTTGGCTGGGCTTCA-3‟

Actb (-actin) F: 5‟-AGGGAAATCGTGCGTGACAT-3‟

R: 5‟-GAACCGCTCGTTGCCAATAG-3‟

Mcl1 Taqman primer/probe set Assay ID: Mm01257351_g1

Birc3 (Ciap1/2) Taqman primer/probe set Assay ID: Mm01168413_m1

Xiap Taqman primer/probe set Assay ID: Mm00776505_m1

Bcl2l1 (Bcl-xL) Taqman primer/probe set Assay ID: Mm00437783_m1

Rn18s (18S) Taqman primer/probe set Assay ID: Hs99999901_s1

Timp3 genotyping

primers

WT: 5‟-AGTTGCAGAAGGCATCCTGGGGATGGCT-3‟

Anchor: 5‟-

CAAGAATCTTCTTCTCCCGCTTCTCCGCTT-3‟

Neo3: 5‟-CCAAATTAAGGGCCAGCTCATTCCTCCCA-

3‟

Page 111: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

91

3 CHAPTER 3

Stromal TIMP3 regulates liver lymphocyte populations and provides protection

against Th1 T-cell driven autoimmune hepatitis

A version of this manuscript has been submitted for peer review:

Aditya Murthy*, Yang Washington Shao

*, Christopher Wedeles, David Smookler, Rama

Khokha. Stromal TIMP3 regulates liver lymphocyte populations and provides protection

in a mouse model of autoimmune hepatitis.

* These authors contributed equally to this work.

Author contributions:

Murthy, A. – designed and performed in vivo experiments, wrote manuscript

Shao, Y.W. – designed and performed flow cytometry experiments, critiqued manuscript

Wedeles, C. – Performed CD4+ T cell culture experiments and RT-PCR analysis

Smookler, D. – Assisted with bone marrow chimera experiments

Page 112: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

92

3.1 Abstract

Lymphocyte infiltration into epithelial tissues and pro-inflammatory cytokine

release are key steps in autoimmune disease. While cell-autonomous roles of

lymphocytes are well understood in autoimmunity, much less is known about the stromal

factors in these tissues that dictate immune cell function. Tissue inhibitor of

metalloproteinases 3 (TIMP3) controls systemic cytokine bioavailability and signaling by

inhibiting the ectodomain shedding of cytokines and their receptors. Here we show that

TIMP3 produced by the hepatic stroma regulates the basal lymphocyte populations in the

liver and the hepatic response to autoimmunity. TIMP3 deficiency led to spontaneous

accumulation and activation of hepatic CD4+, CD8

+ and NK T cells. Treatment with

concanavalin A resulted in a greatly enhanced T helper 1 (Th1) cytokine response and

acute liver failure in a model of autoimmune hepatitis, which mechanistically depended

on TNF signaling. Bone marrow chimeras demonstrated that TIMP3 in the stromal rather

than hematopoietic compartment was responsible for protecting against autoimmunity.

These results uncover metalloproteinase inhibitors as critical stromal factors in regulating

cellular immunity during autoimmune hepatitis.

3.2 Introduction

As a primary site of pathogen encounter, the liver microenvironment has a unique

makeup of local immune cells. The epithelial and stromal cells of the liver such as

hepatocytes, endothelial cells, stellate cells can additionally act as non-classical APCs

and generate exquisite sensitivity to entering pathogens without arming liver dendritic

Page 113: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

93

cells [293]. Autoimmunity involves infiltration of lymphocytes into the liver and T cell

activation, which accelerates disease progression during viral, autoimmune or toxin-

induced hepatitis [293, 294]. This process typically relies on a pro-inflammatory cytokine

milieu that culminates in hepatotoxicity [295, 296]. The lectin concanavalin A (con A)

causes polyclonal T cell activation and is used as an experimental means of inducing

autoimmune hepatitis which recapitulates several aspects of the human disease. The

pathological and protective roles played by macrophages, NK/NKT cells, and the known

T cell subsets are well investigated in this model. Currently, the stromal factors

responsible for regulating peripheral immune cell homeostasis and cytokine milieu in the

liver are largely unknown [239, 297-299].

Tissue inhibitors of metalloproteinases (TIMPs) comprise a well-known family of

soluble factors, in which the 4 Timp genes post-translationally regulate the enzymatic

activity of all metalloproteinases in the mammalian genome. Metalloproteinases perform

ectodomain shedding in which chemokines, cytokines, growth factors and their receptors

are cleaved from the cell surface. This process regulates leukocyte migration and

cytokine signaling during acute and chronic inflammation [300]. Furthermore, tissue

remodeling by the TIMP/metalloproteinase axis plays a key role in dictating immune cell

function and wound healing during liver injury [301]. We and others have previously

demonstrated that TIMP3 functions in a context-dependent manner to direct cytokine

signaling during acute and chronic hepatic stress [69, 76, 253, 302]. However, the

physiological role of TIMPs in cellular immunity remains completely unknown. In this

study, we show that TIMP3 deficiency results in spontaneous lymphocyte infiltration into

Page 114: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

94

the liver. Con A administration to TIMP3 deficient mice results in enhanced Th1

cytokine production and TNF-dependent liver damage. Finally, utilizing bone marrow

chimeras we identify that TIMP3 derived from the stromal sources provides

hepatoprotection against con A-induced autoimmunity.

3.3 Results

3.3.1 A basal increase in CD4+ T cell and NKT cell populations in Timp3

−/−

livers

We investigated the effect of Timp3 deficiency on steady-state composition of

immune cells in the periphery by characterizing the lymphocyte subsets of liver and

spleen from wildtype (WT) and Timp3−/−

mice. Flow cytometry analysis of liver

lymphocytes showed a relative increase in CD4+ T cells and NKT cells (characterized as

CD3+NK1.1

+ cells; Figure 3.1A). Specifically, absolute cell counts of basal CD4

+ T cells

and NKT cells were over two-fold higher and this was reflected in an overall increase in

the number of mononuclear cells (MNCs; Figure 3.1B). On the other hand, splenic

numbers of the same subsets, along with total MNCs were comparable between wildtype

and Timp3−/−

mice in the basal state (Figure 3.1C, D). Given that lymphocyte activation

precedes infiltration into the periphery [297, 299, 303], we next analyzed the activation

marker CD69 on splenocytes and liver lymphocytes by flow cytometry. A higher number

of CD69 expressing CD4+ and CD8

+ T cells as well as NKT cells were observed only in

resting livers of Timp3−/−

mice, while comparable numbers existed in the spleen (PBS

treated groups, Figure. 3.1E, F). Thus loss of Timp3 results in a spontaneous basal

Page 115: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

95

Figure 3.1 Timp3−/−

mice exhibit spontaneous accumulation and activation of hepatic

CD4+ T cells and NKT cells, and enhanced sensitization to con A-induced hepatitis.

(A, B) Flow cytometry analysis and quantification of cell surface CD4/CD8 expression gated on CD3

+

lymphocytes, and CD3/NK1.1 resting lymphocytes from livers of 12-week old wildtype (WT) and

Timp3−/−

mice. (C, D) Flow cytometry analysis and quantification of cell surface CD4/CD8 expression

gated on CD3+ splenocytes, and CD3/NK1.1 resting splenocytes of 12-week old wildtype (WT) and

Timp3−/−

mice. (E, F) Counts of activated liver lymphocyte (E) and splenocyte (D) subsets gated on CD69+

cells. (G) Survival curve of mice treated with 10mg/g con A, then monitored for 48h.(H) Serum

transaminase (ALT) levels in WT and Timp3−/−

mice treated with PBS or Con A. (I) Liver histology (H &

E) of WT and Timp3−/−

mice treated with PBS or con A for 18h. Arrows depict necrosis. *P<0.05, mean ±

S.D. for B, D-F (n=4); *P<0.01, mean ± S.E.M. for H (n=4). Bar = 100m. Flow cytometry plots and

histology are representative of at least 4 mice per condition. Survival curve is based on n=11 mice per

genotype); *P<0.01 (log-rank test).

Page 116: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

96

increase of total liver lymphocytes that are comprised of higher numbers of activated

CD4+, CD8

+ and NK T cells.

3.3.2 TIMP3 deficiency sensitizes mice to T-cell mediated hepatitis induced

by concanavalin A

We have previously found that Timp3−/−

mice have enhanced sensitivity to LPS-

induced hepatitis caused by increased TNF bioactivity but exhibit resistance to Fas-

mediated fulminant hepatitis due to compound alterations in TNFR1 and EGFR signaling

[69, 302]. These models rely on resident macrophage (Kupffer cell) activity and

hepatocyte-intrinsic responses to apoptotic stimuli, but do not address lymphocyte

function in modulating hepatitis. Intravenous administration of con A offers a model of

autoimmune hepatitis induced by lymphocytes [304, 305]; we therefore examined the

susceptibility of Timp3−/−

mice to con A-induced hepatitis. We observed significantly

greater induction of splenomegaly accompanied by an increase in CD69 expressing CD4+

and CD8+ T cells in Timp3

−/− mice. These mice also exhibited higher numbers of

activated liver lymphocytes, specifically CD4+, CD8

+ T cells and NKT cells (con A

treated groups, Figure 3.1E, F). Significantly more Timp3−/−

mice succumbed to a low

dose (10g/g) of con A compared to controls (Figure 3.1G). A greater increase in serum

transaminase (ALT) levels at 6h and 18h indicated enhanced hepatotoxicity (Figure

3.1H). Histological examination showed severe liver damage with extensive necrosis in

Page 117: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

97

Timp3−/−

mice 18h after treatment with con A (Figure 3.2A). Thus, TIMP3 deficiency

exacerbates autoimmune hepatitis in mice.

3.3.3 Enhanced TNF signaling and Th1 cytokine response drives liver

damage in Timp3−/−

mice

TIMP3 is a negative regulator of TNF shedding in vivo, and this cytokine is a key

contributor to con A-induced hepatitis [69, 76, 81, 238, 239, 295, 304]. Immunoblotting

showed phosphorylation of the TNF signaling effector JNK/SAPK at early (6h) and late

(18h) timepoints after con A administration in Timp3−/−

livers (Figure 3.3A). We tested

whether increased TNF bioactivity underlies accelerated induction of autoimmune

hepatitis in Timp3−/−

mice and found that loss of Tnf in wildtype and Timp3−/−

backgrounds protected against hepatotoxicity as indicated by lower serum ALT (Figure

3.3B). Histological analyses confirmed negligible liver damage and hemorrhaging

(Figure 3.3C), and TUNEL staining revealed an absence of hepatocyte apoptosis in

Timp3−/−

Tnf−/−

versus Timp3−/−

mice (Figure 3.3D).

Analysis of early serum cytokine release over 6 hours following con A

administration showed accelerated kinetics of cytokine release in Timp3−/−

mice

compared to all control groups. Tnf−/−

and compound loss of Timp3−/−

/Tnf−/−

resulted in

significant abrogation of cytokine release (Figure 3.3E). Of the Th1 cytokines, TNF

peaked at 2h remaining high until 6h; IFN exhibited a transient 5-fold greater level at

4h; IL-6 continued to rise dramatically over 6h and the Th2 cytokine IL-4 peaked at 2h.

Page 118: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

98

Figure 3.2Enhanced necrosis, lymphocyte infiltration and macrophage activity in

Timp3−/−

livers.

(A) Histology (H & E) of lymphocyte infiltration and enhanced necrosis (arrows) in Timp3

−/− livers 18h

following con A administration. Insets depict lymphocyte infiltration. (B) Immunohistochemical staining

for F4/80 depicting increased macrophage association with areas of necrosis in Timp3−/−

livers 18h

following con A administration. Bars = 100m. Images are representative of at least 3 mice per condition.

Page 119: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

99

Figure 3.3 Loss of Timp3 enhances TNF signaling and Th1 cytokine response during

autoimmune hepatitis.

(A) Immunoblots of JNK phosphorylation in liver lysates from WT and Timp3

−/− mice treated with PBS or

con A. (B) Serum transaminase (ALT) levels 6 hours after treatment with PBS or con A. (C) H & E of

livers treated with PBS or con A. Arrowheads depict necrosis. (D) TUNEL staining of livers 6 hours after

PBS or con A treatment. Brown nuclei depict apoptotic cells (E) Th1 serum cytokine levels over 6 hours

following con A administration. (F) Serum levels of indicated cytokines and chemokines/growth factors

measured 18 hours after PBS or con A administration. Arrowheads depict non-detectable cytokine levels.

*P≤0.04, mean ± S.E.M. (n=4). Bars = 25m. Histology is representative of at least four mice per

condition.

Page 120: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

100

IL-6 plays paradoxical roles in acute hepatic inflammation depending on the duration of

its bioavailability [306, 307], and IL-4 dependent signaling through STAT6 is required

for promoting T-cell mediated hepatitis [305, 308]. Together, these data demonstrate that

TIMP3 deficiency enhances the Th1 cytokine response during autoimmune hepatitis.

Indeed, Timp3−/−

mice displayed sustained increase in Th1 cytokines 18h after con A

treatment. We also identified increases in factors that promote the Th1 response, namely

IL-1, IL-12 and the chemoattractant MCP-1 (Figure 3.3F). Of note, IL-12 release by

activated macrophages promotes T cell activation [299, 309], and consistent with this we

observed greater association of macrophages to areas of necrosis in Timp3−/−

livers

(Figure 3.2B).

We observed increased serum IL-17A and IL-10 in Timp3−/−

Tnf −/−

mice, which

is particularly intriguing since loss of Timp3 alone only modestly affected these cytokines

(Figure 3.4A). The role Th17 cells is still emerging in autoimmune hepatitis and is

primarily associated with hepatoprotective IL-22 production [310]. IL-10 is also

suggested to be protective through its suppression of the Th1 response and its

involvement in immunological tolerance [298]. Additionally we measured decreases in

splenic and hepatic CD4+ T cell numbers, consistent with abrogated liver damage in

Timp3−/−

Tnf −/−

mice (Figure 3.4B). Further studies are needed to elucidate how

metalloproteainse inhibitors impact these immunoregulatory cytokines.

Page 121: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

101

Figure 3.4Timp3−/−

Tnf −/−

compound deletion elevates serum IL-10, IL-17A and

depletes splenic and hepatic CD4+ T cells.

(A) Serum levels of IL-10 and IL-17A following con A treatment in mice of indicated genotypes. (B)

Counts of CD3+CD4

+ T cells in spleen and liver of mice 6 hours after treatment with PBS or con A.

*P≤0.05 vs. WT, mean ± S.E.M. (n≥3).

Page 122: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

102

Figure 3.5 – TIMP3 is dispensable for cell-autonomous activation and proliferation of

CD4+ T cells.

(A) Gene expression of indicated Timps, Adams and Mmps in resting CD4+ T cells isolated from spleens of

wildtype mice. Data are one of three independent analyses. (B) Proliferation of WT and Timp3−/−

CD4+ T

cells following stimulation with con A over 72 hours. (C, D) Cytokine release of (C) IL-2 and (D) TNF

into culture media of CD4+ T cells stimulated with con A. Data are mean ± S.D. (n=3) and representative of

two independent experiments.

Page 123: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

103

3.3.4 Cell-intrinsic TIMP3 is not required for CD4+ T cell activation

Together the 4 TIMPs inhibit metalloproteinase activity of all known 24 MMPs, 9

active ADAMs and 4 active ADAM-TS enzymes, yet the regulation of lymphocyte

function by TIMPs remains uninvestigated [300]. We examined the cell-autonomous role

of TIMP3 in lymphocyte activation in vitro. First, gene expression analysis of Timps and

specific Adam and Mmp genes in resting splenic CD4+ T cells showed low levels of

Timp1, Timp3 and Timp4 but significantly higher expression of Adam10 and Adam17

(Figure 3.5A). Stimulation of cultured wildtype and Timp3−/−

CD4+ T cells (1.0g/ml

con A over 72 hours) showed comparable expansion of CD4+

T cells in both genotypes

when measured by MTS (Figure 3.5B). IL-2 is required for expansion and growth of

antigen-specific T cells subsequent to TCR ligation, and its concentration was

comparable between wildtype and Timp3−/−

CD4+ T cell culture media (Figure 3.5C).

TNF levels also remained comparable across all time points (Supplemental Fig. 4D).

These data demonstrate that TIMP3 is dispensable for cell-autonomous activation,

expansion and effector function of CD4+ T cells.

3.3.5 Stromal TIMP3 protects against con A-induced hepatitis

To evaluate the physiological relevance of hematopoietic and non-hematopoietic

TIMP3 in vivo we generated congenic bone-marrow chimeras as depicted in Figure 3.6A.

Wildtype (CD45.1+) recipients were reconstituted with Timp3

−/− (CD45.2

+) bone marrow

cells and Timp3−/−

recipients with wildtype bone marrow, achieving >90% reconstitution

at 8 weeks post-transplant (bar graphs, Figure 3.6A). We observed that only stromal

Page 124: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

104

Figure 3.6Stromal TIMP3 protects against a Th1 pro-inflammatory cytokine response

and autoimmune hepatitis.

(A) Schematic depicting generation of radiation chimeras. Stacked graphs indicate comparable bone

marrow reconstitution in WT and Timp3−/−

recipients. (B) Counts of total MNCs and subsets of liver

lymphocytes isolated from radiation chimeras 8 weeks after reconstitution. (C) Serum transaminase (ALT)

levels in WT and Timp3−/−

recipients 6 hours after con A treatment. (D, E) Histological analysis depicting

liver damage (D, H & E) and hepatocyte death (E, TUNEL) 6 hours after con A treatment. (F) Serum levels

of Th1 cytokines over 6 hours following con A treatment. (G) Schematic illustrating the regulation of

lymphocyte infiltration and cytokine release by non-hematopoietic TIMP3. Activation and accumulation of

liver lymphocytes generates a positive-feedback loop and predisposes to autoimmune hepatitis. *P<0.01,

mean ± S.D. for A, B (n=4); *P<0.05, mean ± S.E.M. for C, F (n=3). Bars = 25m. Histology is

representative of at least three mice per condition.

Page 125: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

105

TIMP3 deficiency reproduced the spontaneous basal increase in CD3+ liver lymphocytes

(Figure 3.6B) as described in un-irradiated Timp3−/−

mice (Figure 3.1). Treatment with

con A induced serum toxicity and liver damage at 6h in Timp3−/−

recipients harboring

wildtype bone marrow, while wildtype recipients with Timp3 null hematopoietic cells did

not exhibit hepatotoxicity (Figure 3.6C, D). TUNEL staining showed extensive

hepatocyte death only in Timp3−/−

recipients (Figure 3.6E). Further, timecourse analysis

of serum cytokines showed significant elevations in Th1 cytokines IFN, IL-2 and IL-6

over 6h after con A administration (Figure 3.6F) in these mice. Taken together, these data

show that hematopoietic TIMP3 does not impact cytokine release and activation of T

lymphocytes whereas TIMP3 in non-hematopoietic tissues provides protection against

con A-induced hepatitis. Figure 3.6G models the role of Timp genes in autoimmune

hepatitis, demonstrating the requirement of non-hematopoietic TIMP3 in maintaining

hepatic lymphocyte homeostasis and controlling Th1 cytokine response.

3.4 Discussion

Here we demonstrate that the metalloproteinase inhibitor TIMP3 regulates liver

lymphocyte infiltration and pro-inflammatory function during acute hepatitis. Loss of

Timp3 led to spontaneous expansion of liver CD4+ T and NKT cells, and this resulted in

increased hepatotoxicity following con A treatment. Examination of TNF signaling

revealed that elevated serum TNF levels and signaling were causal to enhanced morbidity

in Timp3

mice. We noted that CD4+ T cells isolated from spleen expressed comparably

low levels of Timp genes in wildtype mice; furthermore, wildtype and Timp3

CD4+ T

Page 126: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

106

cells exhibited comparable expansion and cytokine production following in vitro

stimulation with con A. We thus hypothesized that TIMP3 generated by non-

hematopoietic tissue (such as the liver parenchyma) inhibited lymphocyte infiltration and

tested this by the generation of bone marrow chimeras. Indeed, Timp3

recipients of

wildtype bone marrow exhibited spontaneous increase of CD3+ cells and total liver

mononuclear cells. In these chimeric mice, induction of con A-mediated hepatitis

reproduced the enhanced cytokine response and hepatotoxicity. Figure 8E models the role

of Timp genes in autoimmune hepatitis, illustrating the requirement of non-hematopoietic

TIMP3 in maintaining hepatic lymphocyte homeostasis and controlling a Th1 cytokine

response.

The liver has a unique role in maintaining tolerance to food antigens and gut flora

that drain in, while generating protection against exogenous biological insults such as

hepatitis B and C virus, bacterial antigens as well as non-biological agents. Kupffer cells,

sinusoidal endothelial cells and even hepatocytes participate in local antigen presentation

and expression of innate immune receptors such as TLR4; this process is postulated to

generate tolerance to frequently encountered antigen [311]. On the other hand, circulating

dendritic cells can contribute to protective pro-inflammatory responses during infection

by recruiting lymphocytes to the liver microenvironment following encounter with a

pathogen [312, 313]. Our findings introduce the role of metalloproteinase inhibitors in

tolerance versus hepatoprotection. The use of more sophisticated systems (e.g. HBV,

HCV and CMV models of viral infection, Listeria monocytogenes and Mycobacterium

tuberculosis models of bacterial infection) will prove useful in delineating the

Page 127: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

107

contribution of Timp genes to acute CD8+ cytotoxic T lymphocyte (CTL) or CD4

+ T cell

responses against virus [314, 315], and T-helper subset function against bacterial

granulomas [316]. Given the overabundance of CD4+ T cells and NKT cells observed in

Timp3 null mice, we may find enhanced pathogen clearance and protection in an antigen-

specific manner. Potentially, TIMP3 can regulate the sensitization threshold required for

the cellular immune response in the liver.

Several possible mechanisms may be involved in lymphocyte infiltration upon

loss of TIMP3. Disruption of chemokine gradients is a prominent candidate, as increased

hepatic levels of CC- and CXC- chemokines have shown to promote acute liver injury in

murine models and patients alike [317, 318]. Specifically, increases in liver CCL2 (MCP-

1), CCL3 (MIP-1), CCL4 (MIP-1) and CCL5 (RANTES), and CXCR3 ligands

(CXCL9-11) have been reported in human liver biopsies of acute liver pathologies such

as fulminant hepatitis and acute viral hepatitis [319, 320]. Metalloproteinases MMP1, 2, 3

and 14 can cleave CC-chemokines (CCL2, 7, 8, 13), while CXCL11 is shed by numerous

MMPs (MMP1, 2, 3, 9, 13, 14) [14]. Physiologically, chemokines are immobilized on the

extracellular matrix by binding with glycosaminoglycan-associated proteoglycans such as

syndecan. Syndecan-1 harbors „sinks‟ of CXC-chemokines, and its shedding by MMP7 is

an example of the involvement of metalloproteinases in generating CXCL1 gradients. L-

selectin shedding from lymphocyte surfaces is an important early step in their egress

from lymphoid organs and into the periphery [321]. ADAM17/TACE is the only

metalloproteinase known to shed L-selectin in vivo, and TIMP3 is the only endogenous

inhibitor of ADAM17 [300]. In the current study, loss of Timp3 may potentially generate

Page 128: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

108

a permissive environment for L-selectin shedding and uncoordinated lymphocyte egress

into the systemic microenvironment. Finally, TIMP3 is established as a suppressor of

TNF-induced hepatocyte stress signaling during chronic inflammation following partial

hepatectomy or in models of hepatosteatosis and type 2 diabetes [76, 253]. Elevated

production of reactive oxygen species (ROS) is a feature of TNF-mediated stress

signaling and potentially contributes to the immunopathology describe in this study [322,

323].

Immune cell and cytokine composition in the periphery determines the

progression of autoimmunity [324, 325]. While the identification of precise mechanisms

warrants further investigation, this study is the first to reveal a regulatory function for

endogenous metalloproteinase inhibitors in cellular immunity. Taken together, our results

show stromal TIMP3 as a non-cell-autonomous regulator of lymphocyte entry into the

liver microenvironment, implicating its requirement in immunosuppression and

prevention of liver injury during autoimmune hepatitis. Stromal TIMP3 suppresses

undesired lymphocyte infiltration and protects against hepatitis opening avenues to better

elucidate the role of Timps in autoimmune diseases.

Page 129: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

109

3.5 Methods

3.5.1 Mice

All mice used in this study were of C57BL/6 background. Male mice aged 12-15

weeks were used for all the experimental procedures. Timp3−/−

mice have been

previously described [76]. Tnf−/−

mice were obtained from the Jackson Laboratory and

were crossed with Timp3−/−

mice to generate Timp3−/−

Tnf−/−

mice. C57BL/6-Ly5.1

(CD45.1) mice were generously provided by Dr. Norman Iscove (Professor, University of

Toronto, Department of Medical Biophysics, MaRS centre, Toronto, Canada). Mice were

fed 5%-fat chow ad libitum, housed and cared for in accordance with the guidelines

approved by the Canadian Council for Animal Care, and the Animal Care Committee of

the Ontario Cancer Institute. Genotypes were confirmed via PCR for the respective

genes; primer sequences for genotyping Timp3−/−

mice are provided in Table 3.1.

3.5.2 Induction of hepatitis & generation of bone marrow chimeras

10µg/g concanavalin A (Sigma) was injected into the tail vein of 12-week old

male mice dissolved in 200µL sterile PBS to induce hepatitis. Congenic bone marrow

transplants were performed by exposing 12-week old recipient mice to a single dose of

9Gy (900 Rd) ionizing radiation, followed by reconstitution with 4x106 donor bone

marrow cells by injection into the tail vein. Bone marrow and peripheral reconstitution

was allowed for 8 weeks prior to induction of hepatitis.

3.5.3 CD4+ T cell culture

Page 130: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

110

Isolation of spleen CD4+ T cells is detailed in Supplemental Methods. Cells were

cultured in RPMI 1640 medium containing 10% FBS and 50M -mercaptoethanol.

1x105

CD4+ T cells were stimulated with 1.0ng/mL concanavalin A for 24, 48 and 72

hours. Proliferation was measured using a standard MTS assay. Cell culture supernatant

was obtained at indicated timepoints to measure cytokine levels by ELISA following kit

instructions (R & D Systems).

3.5.4 Serum analysis

Serum was obtained prior to and following concanavalin A injection at 2 hour

intervals by bleeding the tail vein. Hepatotoxicity was assayed by measuring serum levels

of alanine transaminase (ALT) and aspartate aminotransferase (AST). Concentrations of

cytokines were measured using a multiplexed cytokine bead array (CBA) on a BD

FACSArray Bioanalyzer system (BD Biosciences) following manufacturer‟s instructions.

3.5.5 Histology & Immunoblotting

Immunohistochemical staining and immunoblotting of liver lobes were performed

as previously described [302]. Liver lobes were fixed in 4% paraformaldehyde for 24

hours, transferred to 70% ethanol and processed for embedding in paraffin. The following

stains were applied: Harris‟ hematoxylin (Electron Microscopy Sciences), Eosin (Fisher

Scientific), TUNEL staining to measure hepatocyte apoptosis, anti-F4/80 antibody to

visualize macrophages/Kupffer cells. All staining was performed by institutional core

facilities.

Page 131: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

111

3.5.6 Flow cytometry

For splenocyte analysis, red blood cells were lysed prior to further processing.

Liver lymphocytes were obtained by density centrifugation using Percoll. Total

splenocytes were isolated as described in Cell Culture above. Liver lymphocytes were

obtained by density centrifugation using Percoll. Briefly, livers were resuspended in 20ml

of 40% Percoll dissolved in William‟s E medium and passed through a 100um nylon

mesh to create single cell suspensions. This was underplayed with 70% Percoll dissolved

in sterile Hank‟s Buffered Salt Solution (HBSS) and centrifuged to generate an interface

containing liver lymphocytes. The interface was removed and processed for flow

cytometry. The following antibodies were used for flow cytometry (from eBiosciences):

anti-CD3 (clone 145-2C11), anti-CD4 (clone GK1.5), anti-CD8 (clone 53-6.7), anti-B220

(clone RA3-6B2), anti-NK1.1 (clone PK136), anti-Gr-1 (clone RB6-8C5), anti-CD11b

(clone M1/70), anti-F4/80 (clone BM8), anti-CD62L (clone MEL-14), anti-CD44 (clone

IM7), and anti-CD69 (clone H1.2F3). Viability was measured by Propidium Iodide (PI)

uptake. Data was obtained on a BD FACSCalibur system and analyzed using FlowJo

software.

3.5.7 Cell culture

CD4+ T cells were isolated from spleens of mice by negative selection using

magnet-assisted cell sorting (MACS, Miltenyi Biotec). Briefly, splenocytes were

obtained by passing whole spleen through a 70m mesh, followed by red blood cell lysis

in AFCS buffer. Cells were resuspended in PBS containing 0.5% BSA and 2mM EDTA.

Page 132: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

112

Dead cells were removed prior to sorting by using the Dead Cell Removal Kit (Miltenyi

Biotec). Splenocytes were incubated with a cocktail of biotinylated antibodies

against CD8a, CD11b, CD11c, CD19, CD45R (B220), NK1.1, DX1, CD105, CD69,

MHC-class II, and Ter-119 (eBiosciences), followed by incubation with magnetically-

labeled microbeads. Magnetic separation was performed using LS columns on a magnetic

field (Miltenyi Biotec). >95% Purity of CD4+ T cell isolation was verified by flow

cytometry.

3.5.8 Immunoblotting

Total liver protein was extracted by mortar and pestle homogenization of frozen

tissue. RIPA extraction buffer containing 25mM Tris-HCl pH 7.6, 1% Triton X-100,

0.1% SDS, 1% NP-40, 1% sodium deoxycholate, 5mM EDTA, 50mM NaCl, 200µM

Na3VO4, 2mM PMSF, and appropriate dilution of Complete Mini, EDTA-free protease

inhibitor cocktail tablets (Roche) was used to lyse all tissue and cells, and lysate was

stored at -70oC. 30g of protein was loaded on SDS-PAGE gels for western blotting. The

following anti-mouse antibodies were used: anti-phosphorylated JNK (p-JNK,

Thr183/Tyr185), anti-JNK (Cell Signaling).

3.5.9 RNA preparation and quantitative RT-PCR

RNA was prepared from sorted splenic CD4+ T cells using TRIzol reagent (Invitrogen),

and cDNA was obtained using a qScript cDNA supermix protocol (Quanta Biosciences).

Gene expression was measured using SYBR Green reagent (Quanta Biosciences) in a

Page 133: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

113

7800HT Real-time PCR system (Applied Biosciences). All gene expression levels were

normalized to Actb (-actin). Relative amount of each product was calculated using the 2-

CT method. Primer sequences are provided in Table 3.1.

3.5.10 Statistical Analyses

Data are reported as mean ± S.D. or S.E.M. Comparisons were made by two-

tailed Student‟s t-test and analysis of variance; comparisons between Kaplan-Meier

survival curves were made by log-rank test.

Table 3.1 – Primer sequences for quantitative RT-PCR analysis and genotyping.

Gene Forward & Reverse Primer Sequence

Timp1 F: 5‟-CATGGAAAGCCTCTGTGGAT-3‟

R: 5‟-CTCAGAGTACGCCAGGGAAC-3‟

Timp2 F: 5‟-GTCATTGCTGCCTTCCTCTC-3‟

R: 5‟-AAAGGGGTGAAGAATGGCTT-3‟

Timp3 F: 5‟-AAACATCTGCCTGGGTTCAG-3‟

R: 5‟-CAAGCTTCCAGCCAAACTTC-3‟

Timp4 F: 5‟-ACCTCCGGAAGGAGTACGTT-3‟

R: 5‟-TTATCTGGCAGCAACACAGC-3‟

Adam9 F: 5‟-TTGCATCCATTGTTGCTCAT-3‟

R: 5‟-CTTCTCAAAGTCCTCCGCAC-3‟

Adam10 F: 5‟-GCAACATCTGGGGACAAACT-3‟

R: 5‟-TTGCACTTGTCACTGTAGCC-3‟

Adam12 F: 5‟-AGAGAAAGGAGGCTGCATCA-3‟

R: 5‟-GCCTGCTTGACCTCTGGTAG-3‟

Adam17 F: 5‟-AGGATGCTTGGGATGTGAAG-3‟

R: 5‟-CTGTTTGCTCTGGGAGAACC-3‟

Mmp2 F: 5‟-ACTGACACTGGTACTGGCCC-3‟

R: 5‟-GTCACGTGGTGTCACTGTCC-3‟

Mmp9 F: 5‟-CCCGCTGTATAGCTACCTCG-3‟

R: 5‟-CTGTGGTTCAGTTGTGGTGG-3‟

Mt1-mmp F: 5‟-CTGTCCCAGATAAGCCCAAA-3‟

R: 5‟-GCATTGGGTATCCATCCATC-3‟

Page 134: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

114

Timp3 genotyping

primers

WT: 5‟-AGTTGCAGAAGGCATCCTGGGGATGGCT-3‟

Anchor: 5‟-

CAAGAATCTTCTTCTCCCGCTTCTCCGCTT-3‟

Neo3: 5‟-CCAAATTAAGGGCCAGCTCATTCCTCCCA-

3‟

Page 135: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

115

4 CHAPTER 4

ADAM17 activation of epidermal Notch regulates atopic barrier immunity and

myeloproliferation by suppressing c-Fos-driven transcription of epithelial cytokines

A version of this chapter is currently under revision at Immunity:

Aditya Murthy, Yang Washington Shao, Swami R. Narala, Sam D. Molyneux, Juan

Carlos Zúñiga-Pflücker, Rama Khokha.

ADAM17 activation of epidermal Notch

regulates atopic barrier immunity and myeloproliferation by suppressing c-Fos-driven

transcription of epithelial cytokines.

Author contributions:

Murthy, A. – designed and performed majority of the experiments, wrote manuscript

Shao, Y.W. – performed flow cytometry experiments

Narala, S.R. – assisted with chromatin immunoprecipitation experiments

Molyneux. S.D. – analyzed human gene expression datasets

Zúñiga-Pflücker J.C. – provided expertise for ligand dependent Notch activation assays

Page 136: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

116

4.1 Abstract

Epithelial cells of mucosal tissues provide a barrier against environmental stress,

and keratinocytes are key decision-makers for immune cell function in the skin.

Currently, epithelial signaling networks that instruct barrier immunity remain

uncharacterized. Here we show that constitutive or postnatal keratinocyte-specific

deletion of a disintegrin and metalloproteinase 17 (Adam17) triggers T helper 2 (Th2)-

driven atopic dermatitis along with myeloproliferative disease. In vivo and in vitro

deficiency of ADAM17 dampened Notch signaling, increasing production of Th2-

polarizing cytokine TSLP and myeloid growth factor G-CSF. Ligand-independent Notch

activation was identified to regulate AP-1 transcriptional activity, with Notch specifically

antagonizing c-Fos recruitment to the promoters of Tslp and Csf3 (G-Csf). Further, local

skin inflammation was rescued and myeloproliferative disease ameliorated by adenoviral

delivery of active Notch to Adam17

epidermis. Our findings uncover an essential role

of ADAM17 in the adult epidermis and show the gatekeeper function of the

ADAM17/Notch/c-Fos triad in barrier immunity.

4.2 Introduction

Inflammatory skin diseases afflict greater than 10% of people in the western

world [326]. The onset of epidermal inflammation represents a collapse of epithelial

homeostasis resulting in the generation of alarm signals by keratinocytes. This in turn

recruits the immune system to eliminate infected and/or damaged cells and restore

homeostasis. Factors produced by epithelial cells that guide immune cell functions are

Page 137: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

117

being recognized as the „epimmunome‟ and it is important to elucidate the mechanisms

that underpin their synthesis. Stress response pathways (e.g. NF-B, AP-1) have emerged

as key junctions in the maintenance of epithelial cell homeostasis [327]. Disruption of

either developmental programming or induction of physiological stress in keratinocytes

leads to copious production of „alarmins‟ including thymic stromal lymphoprotein,

(TSLP), interleukin 33 (IL-33), as well as hematopoietic growth factors such as

granulocyte colony stimulating factor (G-CSF) or granulocyte/macrophage colony

stimulating factor (GM-CSF). These over time generate atopic dermatitis by TSLP-

mediated activation of dermal dendritic cells, which results in a Th2-polarized local

cellular immune response [328, 329]. Additionally, overt production of myeloid growth

factors by keratinocytes has shown to be causal to the development of myeloproliferative

disease (MPD) in mouse models of inflammatory skin disease [330, 331]. Therefore,

keratinocytes in the epidermis direct local immunity and hematopoietic development, yet

epithelial signaling networks which instruct the epimmunome are poorly defined.

The Notch pathway is a master regulator of cell fate decisions and has established

roles in directing differentiation of all cell layers in metazoans. Recent emerging

evidence also supports a role for this pathway in the maintenance of adult tissue

homeostasis. In epithelial tissues such as the skin, Notch signaling sustains the epidermal

barrier by promoting cell-autonomous keratinocyte terminal differentiation [332].

Importantly it also orchestrates communication between the epithelial and immune

compartments of the skin to prevent chronic inflammation. Recent studies of Notch in

adult tissue homeostasis have highlighted its requirement in preventing inflammatory

Page 138: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

118

skin disease [330, 333]; however, the molecular mechanisms by which epithelial Notch

performs this immunoregulatory function remain elusive.

ADAM17/TACE is a transmembrane metalloproteinase that cleaves cell surface

proteins [133, 300]. It is essential for postnatal survival in mice and is implicated in

immune cell development and function [201, 203, 286]. ADAM-mediated shedding of

Notch receptors is a key step in activating Notch signaling, but to date only ADAM10 has

been shown to perform this action in vivo, as demonstrated by its role in marginal zone B

cell development [123]. ADAM10 also performs S2 cleavage of Notch in keratinocytes,

and epidermal ADAM10 deficiency leads to defective barrier formation [128]. However

the relevance of ADAM17 in epithelial:immune crosstalk is unknown. We investigated

this by inactivating Adam17 in the epidermis of adult mice. The main outcomes were

spontaneous onset of Th2-driven atopic dermatitis (AD) and myeloproliferative disease

(MPD). Keratinocyte-specific deletion of Adam17 triggered spontaneous accumulation of

Th2 cells, dermal mast cell infiltration and elevated serum IgE. An increase in Gr-

1+CD11b

+ cells and concomitant loss of B220

+ B lymphocytes was observed in the bone

marrow. Using in vivo and in vitro approaches, we found that ADAM17 is crucial for

basal Notch activation in adult epidermis, and that ectopic Notch activation was sufficient

to rescue local skin inflammation and MPD in mutant mice. We further identified

elevated activator protein-1 (AP-1) transcriptional activity as causal to TSLP and G-CSF

production. Analysis of signal crosstalk between Notch and AP-1 pathways in mouse and

human keratinocytes revealed that Notch antagonized c-Fos recruitment to epithelial

cytokine promoters to keep stress signaling in check. Collectively, our results

Page 139: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

119

demonstrate that ADAM17 permits tonic Notch activation in the adult epidermis to

regulate epithelial cytokine production and maintain barrier immunity.

4.3 Results

4.3.1 Onset of spontaneous atopic dermatitis upon deletion of epidermal

Adam17

Keratinocytes comprise the vast cellular majority of the epidermis. Basal

keratinocytes reside at the epidermal-dermal interface, and migrate outward towards the

skin surface as they undergo terminal differentiation. We induced Adam17 deletion in

basal keratinocytes by breeding Adam17fl/fl

and K14-Cre transgenic mice (Figure 4.1A).

These mice (termed Adam17ep

) exhibited a progressive atopic-dermatitis like

inflammatory skin disease with 100% penetrance as early as 3 weeks of age (Figure

4.1B). We also measured a marked increase in thymic stromal lymphoprotein (TSLP) and

IL-33, both in the epidermis and systemically (Figure 4.1C). H & E staining showed

epidermal thickening, a loss of subcutaneous fat and regions of hyperkeratosis, while

immunohistochemical analysis with Ki67 revealed keratinocyte hyperproliferation

(Figure 4.1D, E). Histological and flow cytometry analysis revealed massive infiltration

by F4/80+ macrophages (Figure 4.1F, G; FACS quantification in scatterplot to the right)

and Toluidine-B+ mast cells in the dermis (arrows, Figure 4.1H; quantification in

scatterplot to the right). Consistent with mast cell influx, serum IgE levels were >100-

Page 140: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

120

Figure 4.1Epidermal ADAM17 deficiency causes atopic dermatitis (AD).

(A) Immunoblot performed on tail epidermis and dermis to confirm tissue-specific Adam17 deletion. (B)

Adam17fl/fl

(fl/fl) and K14-Cre transgenic mice were bred to generate tissue-specific deletion of Adam17 in

basal keratinocytes (ep mice). (C) Serum and tail epidermis analyzed by ELISA for levels of TSLP and

IL-33. (D, E) Histological analysis performed on dorsal skin (D, H&E) and keratinocyte proliferation (E,

Ki67 immunostaining). (F, G) Immunohistochemical and flow cytometry (FACS) quantification for F4/80

to measure macrophage infiltration in dermis. (H) Toluidine-B staining performed to measure mast cell

infiltration (red arrows), quantified on the right. (I) Serum IgE measured by ELISA. (J, K) Bead array

profiling of serum for indicated cytokines and chemokines. *P<0.05 mean ±S.E.M (n≥4). Histology and

immunoblotting data are representative at least six mice. Scale bars = 100m.

Page 141: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

121

fold higher in Adam17ep

mice (Figure 4.1I). These data indicated spontaneous onset of

AD in mice lacking epidermal ADAM17.

The production of Th2 cytokines typifies atopic diseases [334]. We observed

dramatic changes in serum levels of specific factors; most notably the elevation of Th2

(IL-4, IL-15) and reduction in Th1 (IFN, IL-12p40) cytokines in 10-week old mutant

mice (Figure 4.1J). IL-17A, a major contributor to autoimmunity [335] and a marker of

acute AD was also higher (Figure 4.1J). Serum chemokines were deregulated including a

decrease in CXCL10, a factor implicated in the recruitment of regulatory T cells (Tregs) to

counteract inflammation (Figure 4.1K) [336]. This demonstrated a Th2-dominant atopic

immune response upon epidermal Adam17 loss.

4.3.2 Th2-polarized cellular immunity in skin-draining lymph nodes and

myeloproliferative disease in bone marrow of Adam17ep

mice

In order to characterize the nature and origin of the cellular immune response, we

compared skin-draining (dLN) and mesenteric (mLN) lymph nodes. Along with obvious

lymphadenopathy, flow cytometry analysis showed a 10-fold elevation in Gr-1+CD11b

+

monocytes and the presence of CD11c+MHCII

+ antigen presenting cells (APCs) in

Adam17ep

mice (Figure 4.2A-C). Activated CD4+CD69

+ T lymphocytes were observed

only in the dLNs of Adam17ep

mice (Figure 4.2D). Lymphocyte activation follows a

shift from naïve (CD62L+CD44

) to effector (CD62L

CD44

+) T cell phenotypes [337].

Again, Adam17ep

mice had a 4-fold increase in CD4+ effector T cells specifically in dLN

Page 142: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

122

but not in mLN (Figure 4.2E). This confirmed the skin-specific origin of cellular

immunity in mice lacking epidermal ADAM17.

Mice harboring genetic epidermal defects often exhibit myeloproliferative disease

(MPD) [330, 331]. Our investigation of bone marrow homeostasis revealed strikingly

high levels of serum G-CSF along with the presence of splenomegaly in Adam17ep

mice

(Figure 4.2F, G). Interestingly, we noted a severe drop in bone marrow B220+

lymphocyte populations in Adam17ep

and mice, which upon further characterization

were found to be predominantly naïve B lymphocytes expressing high levels of IgM

compared to controls (Figure 4.2H, I). While the total number of bone marrow cells were

comparable (data not shown), Gr-1+CD11b

+ myeloid populations were proportionally

higher in bone marrow and spleen of Adam17ep

mice (Figure 4.2J), fulfilling all of the

hallmarks of MPD [338]. Thus, epidermal ADAM17 deficiency is sufficient to induce

myeloproliferation in mice.

4.3.3 Precocious differentiation of ADAM17 deficient keratinocytes in vivo

and in vitro

ADAMs perform ectodomain shedding of several factors required for keratinocyte

development including EGFR ligands and Notch. For example ADAM17 cleaves a subset

of EGFR ligands whereas ADAM10 is the recognized metalloproteinase for Notch

processing. The epidermal loss of Adam10 was recently reported to impair keratinocyte

differentiation in mice; they failed to establish the spinous layer of the epidermis [128].

We therefore asked whether the observed epidermal inflammation arose

Page 143: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

123

Figure 4.2Skin-specific Th2 lymphocyte activation and myeloproliferative disease

(MPD) in Adam17ep

mice.

(A) Gross images depicting lymphadenopathy of the skin-draining lymph nodes in ep mice. (B) FACS

analysis of macrophage/monocytes (Gr-1+CD11b

+) in skin-draining lymph nodes (dLN) and mesenteric

lymph nodes (mLN). (C) FACS analysis of activated (CD11c+MHCII

+) dendritic cells in dLNs. (D) FACS

analysis of CD8+ vs. CD4

+ T cell activation by CD69 staining in mLNs and dLNs. (E) Induction of CD4

+

effector T cells (CD62L-CD44

+) in dLNs of ep mice measured by FACS. (F) ELISA measurement of

serum G-CSF levels in control (fl/fl) and Adam17ep

(ep) mice aged 10 weeks. (G) Representative image

of spleen depicting splenomegaly in ep mice. (H) FACS analysis of B220+ B lymphocytes in bone

marrow (BM). Analysis of CD19+B220

+ population shows a greater fraction of IgM

+ naïve B lymphocytes

in ep mice. (J) FACS analysis of CD11b+Gr-1

+ macrophage/monocytes in BM and spleen. *P<0.05 mean

±S.E.M (n=6). Data in A-E and G-I are representative of at least twelve mice. Scale bars = 5mm.

Page 144: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

124

due to incomplete keratinocyte differentiation. This was not the case for Adam17ep

mice,

as immunofluorescence staining of the skin showed comparable establishment of terminal

keratinocyte differentiation as measured by basal (Keratin5), early (Keratin10) and late

(Filaggrin) markers (Figure 4.3A). Further, gene expression analysis of keratinocyte

differentiation markers confirmed the presence of basal and spinous (early) lineages

along with increased expression of granular (late) lineage markers in the epidermis of

newborn Adam17ep

mice (Figure 4.3B). We next created cell-autonomous ex vivo

deletion of Adam17 using Adam17fl/fl

keratinocytes transduced with adenoviral Cre

(termed AdCre-Adam17

), and tested calcium-induced differentiation. AdCre

keratinocytes exhibited accelerated differentiation as measured by gene expression of

basal, early (spinous) and late (granular/cornified) markers (Figure 4.3C). Of note,

adenoviral deletion of Adam17 induced precocious keratinocyte differentiation even in

basal conditions (AdCre Low Ca++

group, Figure 4.3C). Consistent with the onset of

terminal differentiation, AdCre keratinocytes proliferated at a slower rate in basal

conditions when compared with control (AdGFP) keratinocytes (Figure 4.3D). This data

indicates that in contrast to ADAM10 deficiency, loss of Adam17 leads to precocious

keratinocyte differentiation in a cell autonomous manner.

4.3.4 Inducible deletion of epidermal Adam17 in adult mice recapitulates

atopic dermatitis and myeloproliferative disease

Beyond keratinocyte differentiation, we reasoned that ADAM17 would be required for

epidermal homeostasis in the adult skin. We thus bred Adam17fl/fl

mice with

Page 145: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

125

Figure 4.3Accelerated differentiation of Adam17ep

keratinocytes.

(A) Histological (Immunofluorescence) analysis of skin at indicated postnatal ages (P) measuring

expression of basal marker Keratin5 (K5, green), early differentiation marker Keratin10 (K10, red), and

late differentiation marker Filaggrin (Fil, red). (B) qPCR analysis of basal, early and late differentiation

markers in newborn (P1). (C) qPCR analysis of keratinocyte differentiation markers in primary

keratinocytes following calcium-induced terminal differentiation. Cells were maintained in basal medium

(Low Ca++

) or treated with 2mM calcium (High Ca++

) for 48 hours. AdGFP (control): Adam17fl/fl

keratinocytes transduced with adenoviral-GFP; AdCre (Adam17 knockout): Adam17fl/fl

keratinocytes

transduced with adenoviral-Cre to induced cell-autonomous Adam17 deletion. (D) Decreased rate of

proliferation in AdCre-Adam17

keratinocytes cultured in basal conditions. *P<0.05, mean ± S.E.M for B,

C, S.D. for D (n=3). Images are representative of at least 4 mice per genotype. Scale bars = 50m.

Page 146: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

126

transgenic mice expressing tamoxifen-inducible Cre-recombinase under control of the

Keratin14 promoter (termed Adam17epERT

mice) and induced Adam17 deletion as

outlined in Figure 4.4A. Mice were analyzed at early (3 month) and late (8 month) time

points post-deletion. Tamoxifen-induced deletion of Adam17 resulted in gross

lymphadenopathy and skin inflammation (Figure 4.4B, C). We observed epidermal

thickening (H & E, Figure 4.4D) and keratinocyte hyperproliferation as measured by

Ki67 staining as early as 3 months (Figure 4.4E), as well as dermal infiltration by

Toluidine B+ mast cells (arrows, Figure 4.4F).

Flow cytometry analysis indicated higher numbers of CD4+CD69

+ activated T

cells in dLNs of Adam17epERT

mice (Figure 4.4G). Although CD8+CD69

+ T cell numbers

were also higher, the proportion of activated CD4+ T cells was 4 times greater than CD8

+

T cells (Figure 4G, quantified in Figure 4.4H). Consistent with this, a significantly higher

proportion of CD4+CD62L

CD44

+ effector T cells was observed in dLNs of

Adam17epERT

mice at all ages (Figure 4.4I, quantified in Figure 4.4J). Finally, these mice

developed MPD as characterized by loss of B220+ B lymphocytes (Figure 4.5A, B) and

proportionally greater populations of Gr-1+CD11b

+ macrophage/monocytes in bone

marrow and spleen (Figure 4.5C, D). These experiments establish that deletion of

epidermal Adam17 in the adult skin compromises barrier immunity.

Page 147: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

127

Page 148: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

128

Figure 4.4Inducible deletion of epidermal Adam17 in adult mice recapitulates atopic

dermatitis and skin-specific Th2 lymphocyte activation.

(A) Schematic depicting protocol for tamoxifen treatment and mouse analysis. (B) Representative images

of dLNs in Adam17fl/fl

(fl/fl) and K14-CreERT

;Adam17fl/fl

(epERT) mice following tamoxifen treatment as

outlined in Figure 4A. (C) Control (fl/fl) and K14-CreERT transgenic mice were bred to generate

tamoxifen-inducible deletion of Adam17 in basal keratinocytes (epERT mice). Images are representative

of four mice at indicated time points after tamoxifen treatment. (D-F) Histological analysis of dorsal skin

by H & E staining (D) Ki67 immunostaining depicting basal keratinocyte proliferation (E) and mast cell

influx (F, arrows) shown by Toluidine B staining. (G, H) FACS plots and quantification of CD8+ and

CD4+

T cell activation in mesenteric lymph nodes (mLN) and skin-draining lymph nodes (dLN) measured

by cell surface CD69. (I, J) FACS plots and quantification of CD8+ and CD4

+ effector T cells identified as

CD62L-CD44

+ populations in mLNs and dLNs of Adam17

fl/fl and Adam17

epERT mice. *P<0.05, mean ±

S.E.M. (n=4).

Page 149: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

129

Figure 4.5 – Onset of MPD upon tamoxifen-induced deletion of epidermal Adam17 in

adult mice.

(A, B) FACS plots showing a loss of B220

+ B lymphocytes and a relative increase in IgM

+ naïve B

lymphocytes in bone marrow of epERT mice 3 and 8 months after tamoxifen treatment. Total B220+ B

lymphocytes in BM are quantified in the graph on the right. (C, D) MPD depicted by FACS analysis of Gr-

1+CD11b

+ macrophage/monocyte populations in bone marrow (BM) and spleen of epERT mice following

tamoxifen treatment. Bars to the right are quantifications of Gr-1+CD11b

+ myeloid cells. Flow cytometry

plots are representative of 3 mice per condition. *P<0.05, mean ± S.E.M. (n=3).

Page 150: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

130

4.3.5 TNFR1 signaling is dispensable for atopic dermatitis and

myeloproliferative disease in Adam17ep

mice

TNF inhibitors have had measurable success in therapy of inflammatory skin

disease in humans [339-341]. Independent genetic models show that psoriasis-like

inflammatory skin disease caused by epidermal loss of Ikkb or c-Jun/JunB is ameliorated

by the deletion of TNF receptor 1 (Tnfr1), however the importance of TNFR1 signaling

in models of atopic dermatitis has not been tested. ADAM17 is the most relevant

sheddase for the cleavage of membrane bound TNF and its receptors, and we have

previously shown that regulation of ADAM17 activity by its endogenous inhibitor

TIMP3 provides a critical axis of control over TNF in models of acute and chronic

hepatic inflammation [69, 76, 98]. We bred Adam17ep

mice into a Tnfr1 deficient

background and surprisingly found that Tnfr1 loss failed to rescue epidermal

inflammation (Figure 4.6A). Analyses of peripheral immunity demonstrated activation of

CD4+ T cells (Figure 4.6B), induction of CD4

+ effector T cells (Figure 4.6C) and

elevated MHCII expression on APCs in dLN of Adam17ep

;Tnfr1mice (Figure 4.6D).

Adam17ep

;Tnfr1

mice also exhibited MPD, with increased proportions of Gr-

1+CD11b

+ myeloid cells and a loss of B220

+ B lymphocytes in the bone marrow (Figure

4.7A, B). Thus, epidermal ADAM17 prevents atopic dermatitis and maintains bone

marrow homeostasis independent of TNFR1 signaling.

Page 151: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

131

Figure 4.6 – Tnfr1 deletion does not rescue AD in Adam17ep

mice.

(A) Representative images depicting the presence of AD in Adam17

ep;Tnfr1

(ep;Tnfr1

) double

knockout mice. (B) FACS analysis analysis and quantification of CD8+ and CD4

+ T cell activation

measured by CD69 levels in dLNs of control (fl/fl), single knockout (ep) and double knockout

(ep;Tnfr1

) mice. Activated lymphocytes are quantified in graphs to the right. (C) FACs analysis and

quantification of CD62L-CD44

+ effector CD8

+ and CD4

+ T cell subsets in dLNs. (D) Mean fluorescence

intensity (MFI) quantification of MHCII expression measured by FACS of B220+ B lymphocytes and

CD11c+ APCs. Flow cytometry plots are representative of 6 mice per condition. *P<0.05, mean ± S.E.M.

(n=6).

Page 152: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

132

Figure 4.7 – MPD persists in Adam17ep

;Tnfr1

double deficient mice.

(A) FACS analysis of bone marrow (BM) monocyte/macrophages shows comparable numbers of Gr-

1+CD11b

+ cells in ep and ep;Tnfr1

mice. Total cell numbers are quantified by graphs on the right. (B)

Similar decrease in B220+CD19

+ bone marrow B lymphocytes in ep and ep;Tnfr1

mice. Bars are

quantifications of total cell numbers in 6 mice per genotype. *P<0.05, mean ± S.E.M. (n=6).

Page 153: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

133

4.3.6 Loss of Adam17 compromises tonic Notch signaling in the epidermis

Remarkably, Adam17ep

mice closely phenocopy the onset of AD and MPD

previously illustrated upon inducible and combined postnatal deletion of epidermal

Notch1 and Notch2 [330]. However, there is no physiological evidence for ADAM17-

mediated Notch activation in mammals. We tested whether ADAM17 deficient skin had

defective Notch signaling and found Notch intracellular domain (NICD) to be absent in

adult Adam17ep

epidermis (Figure 4.8A). Moreover, expression of multiple Notch target

genes was diminished in Adam17ep

epidermis (Figure 4.8B). Typically, Notch activation

follows ligand binding (Delta-like 1, 3, 4, Jagged 1, 2) and involves metalloproteinase

(ADAM)-mediated S2 cleavage of the receptor at the cell surface with subsequent NICD

release via -secretase (S3 cleavage) [342]. Notch activation can occur in a ligand-

dependent or ligand-independent manner, and Bozkulak et al (2009) have shown that

ADAM10, but not ADAM17, is required for ligand-dependent Notch activation in vitro

[123, 138]. To delineate how ADAM17 contributed to Notch activation in keratinocytes,

we examined both possible scenarios for Notch activation using AdCre-Adam17

keratinocytes. In a ligand-independent model, calcium treatment of AdCre-Adam17

keratinocytes failed to produce NICD or induce Notch target gene expression (Figure

4.7C, D). On the other hand, ADAM17 was dispensable for ligand-dependent Notch

signaling when induced by Fc-Delta-like 4 fusion protein (Figure 4.8E). Together, these

data demonstrate that ADAM17 activity is required for Notch signaling in adult skin, and

suggest that ADAM17 activates Notch in a ligand-independent manner in keratinocytes.

Page 154: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

134

Figure 4.8ADAM17 provides ligand-independent Notch signaling and suppresses c-

Fos activity in keratinocytes. (A) Immunoblot depicting loss of NICD production in adult ep epidermis. (B) qPCR analysis of newborn

(P1) epidermis for Notch target genes Hes5, Hey1 and Hey2. (C) Immunoblot depicting a lack of S3

cleavage in AdCre-Adam17

keratinocytes following calcium (Ca++

) treatment. (D) qPCR measurement

of Notch target gene expression (Hes1, Hey2) following calcium treatment. (E) qPCR of Notch target gene

expression in AdGFP-control and AdCre-Adam17

keratinocytes following ligand-dependent Notch

activation by Delta like 4 (hFc-DLL4). (F) Analysis of published microarray gene expression datasets

indicates differential expression of genes involved in Notch signaling in atopic dermatitis and psoriasis

patients compared to normal controls. *P<0.05 mean ± S.E.M (n=4 for B, n=3 for D, E). Data are

representative of 4 mice (A, B), and three independent experiments (C-E).

Page 155: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

135

Figure 4.9Stratification of atopic dermatitis, atopic eczema and psoriasis patients

based on expression analysis of genes involved in Notch signaling.

(A-C) Heatmaps of significantly differentially expressed genes involved in Notch signaling. (D)

Unsupervised hierarchical clustering of atopic dermatitis, psoriasis and normal skin using the Notch

signaling gene set. Gene lists are provided in Table 5.1 (Appendix A).

Page 156: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

136

We sought evidence for compromised Notch signaling in patients with

inflammatory skin disease using published microarray human gene expression datasets

[343-346]. We consistently observed genes in the Notch pathway to be differentially

expressed in patients with AD (Figure 4.8F), atopic eczema (Figure 4.9A) and psoriasis

(Figure 4.9B, C). Specifically, a decrease in Notch2, Notch3, and Presenilin1 in AD was

observed compared to normal skin (Figure 4.8F; gene lists in Table 4.1 provided in

Appendix A). Of note, Adam10 and Adam17 displayed a heterogeneous expression

pattern in AD skin, likely due to the pleiotropic functions of these metalloproteinases. We

also performed unsupervised clustering analysis and observed that genes in the Notch

signaling module stratified AD samples away from psoriasis and normal samples,

suggesting that Notch signaling is more deregulated in AD than in psoriasis (Figure

4.9D). The above data proposes a link between ADAM17 and Notch signaling in human

atopic skin disease.

4.3.7 Active Notch antagonizes c-Fos/AP-1 transcription of Tslp in human

keratinocytes

Our next goal was to identify the mechanism by which ADAM17-driven Notch activation

keeps epithelial inflammation in check. Epithelial cytokines TSLP and IL-33 have

emerged as key „alarmins‟ in barrier immunity [328, 329, 347]. Keratinocytes also

produce significant amounts of hematopoietic growth factors Csf3 (G-CSF) and Csf2

(GM-CSF) in an AP-1 regulated manner [331, 348]. The potential for alternative Notch

signaling exists via its interaction with transcription factors NF-B, and possibly AP-

Page 157: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

137

1[349-351]. The AP-1 family has pleiotropic functions in stress response where specific

members dimerize, bind to target sequences and regulate gene expression [348]. Notch

has been proposed to inhibit AP-1 activity in vitro [349, 351]. To interrogate members of

the AP-1 transcription factor family antagonized by Notch, we used a human keratinocyte

cell line (HaCaT cells). We began by measuring individual AP-1 proteins (c-Jun, JunB,

JunD, c-Fos, Fra1, Fra2) over 6 hours following treatment with the phorbol ester PMA

(an AP-1 agonist). Of these AP-1 members, c-Fos displayed a distinct profile with its

levels peaking at 2 hours and rapidly terminating at 3 hours (Figure 4.10A). c-Fos

typically dimerizes with the Jun proteins (c-Jun, JunB, JunD) [348] and their levels

gradually increased in HaCaT cells following PMA stimulation (Figure 4.10A). We

inhibited either ADAM17 activity or NICD production by using small molecule

inhibitors (TAPI-1 and DAPT, respectively), and both resulted in a sustained production

of c-Fos upon PMA treatment (Figure 5G). TAPI-1 or DAPT treatment also led to the

depletion of NICD as measured by immunoblotting (Figure 4.10B). Notably, c-Fos

stabilization or NICD depletion correlated with elevated Tslp and Csf3 transcription as

measured by qPCR (Figure 4.10C, D).

The promoter regions of Tslp and Csf3 contain TPA regulatory elements (TRE)

that bind AP-1 and promoter polymorphisms that enhance AP-1 binding are associated

with higher susceptibility to bronchial asthma [352]. These promoters also harbor RBP-j

binding sites that recruit the Notch transcriptional complex (schematics shown in Figure

4.10E, F). We assessed c-Fos and NICD recruitment to these promoters by

Page 158: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

138

Figure 4.10ADAM17 activates Notch to suppress c-Fos transcriptional activity on

cytokine promoters.

(A) Immunoblots of individual AP-1 members from whole cell lysates of HaCaT cells treated with 50ng/ml

PMA for the indicated timepoints. Data are representative of two independent experiments. (B)

Immunoblotting for NICD, c-Fos and Fra1 in HaCaT cells pre-treated with ADAM17 inhibitor TAPI-1 or

-secretase inhibitor DAPT, followed by PMA treatment for the indicated time points. (C, D) qPCR

measurement of Tslp and Csf3 (G-Csf) gene expression in HaCaT cells treated as in (A). (E, F) NICD and

c-Fos conversely regulate Tslp and Csf3 transcription by recruitment to promoter regions. HaCaT cells

were pre-treated with DMSO (control) or the ADAM17 inhibitor TAPI-1, followed by PMA treatment for

the indicated times. Chromatin was precipitated using the indicated antibodies. Tslp and Csf3 promoters

were amplified from precipitated DNA. Schematics of Tslp and Csf3 promoters depict AP-1 binding sites

(TRE), Notch binding sites (RBP-j) and primer design (small arrows). *P<0.05 mean ± S.E.M (n=4).

Data are representative of two independent experiments.

Page 159: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

139

chromatin immunoprecipitation (ChIP) assays. NICD was constitutively present on the

Tslp promoter in HaCaT cells while c-Fos was only recruited at 2 hours after PMA

treatment. Inhibition of ADAM17 activity with TAPI-1 depleted NICD production and

abolished its recruitment to the Tslp promoter while concomitantly prolonging c-Fos

binding to this region at 3 hours after PMA treatment (Figure 4.10E). Similar but not

identical antagonism was observed between NICD and c-Fos at the Csf3 promoter

(Figure 4.10F). These results suggest that loss of Notch activation allows for unchecked

c-Fos transcriptional activity.

4.3.8 Ectopic Notch rescues the enhanced AP-1 driven stress response in

ADAM17 deficient keratinocytes

We next determined whether tonic Notch activation by ADAM17 keeps AP-1

dependent cytokine production in check. Adenoviral Cre-mediated Adam17 deletion in

keratinocytes (AdCre-Adam17

) resulted in spontaneous phosphorylation of the stress

kinase JNK1/2 (Figure 4.11A). Treatment with PMA specifically enhanced the levels of

c-Fos and Fra1 in AdCre-Adam17

compared to AdGFP control keratinocytes.

Additionally, the DNA binding activity of AP-1 members was elevated in AdCre-

Adam17

keratinocytes (Figure 4.11B, C). Next, qPCR analysis showed higher Tslp,

Csf2, Csf3, and c-fos expression in AdCre-Adam17keratinocytes after PMA treatment;

this was abrogated by the pan AP-1 inhibitor Tanshinone IIA (TanIIA) (Figure 4.11D;

Figure 4.12A). We noted the specificity of this response since ADAM17 deficiency did

Page 160: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

140

Figure 4.11Diminished Notch activation upon ADAM17 deficiency enhances c-

Fos/AP-1 driven stress signaling in keratinocytes. (A) Immunoblotting detects spontaneous SAPK/JNK phosphorylation in AdCre-Adam17

keratinocytes.

(B) Elevated protein levels of c-Fos and Fra-1 in AdCre-Adam17

keratinocytes following PMA

treatment. (C) Enhanced AP-1 transcription factor binding for specific members of AP-1 family in AdCre-

Adam17

keratinocytes treated with PMA. AdGFP-control and AdCre-Adam17

keratinocytes were

treated with 10ng/ml PMA for 3 hours, followed by measurement of AP-1 target sequence binding by

individual members of the AP-1 family. (D, F) qPCR analysis of Csf3, Tslp and c-Fos gene expression in

keratinocytes 3 hours after PMA treatment. Presence of the pan AP-1 inhibitor Tanshinone IIA (TanIIA, D)

or ectopic expression of active Notch (AdNICD, F) abrogates gene expression. (E) Measurement of AP-1

luciferase reporter activity in MEFS after 8 hours of PMA treatment. Use of a specific c-Fos inhibitor (T-

5224) or ectopic Notch (AdNICD) abolishes AP-1 luciferase activity. *P<0.05, mean ± S.E.M. (n=3). Data

in A-C are representative of two independent experiments. Data in D and E are representative of three

independent experiments.

Page 161: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

141

Figure 4.12AdCre-deletion of Adam17 in keratinocytes enhances Csf2 gene

expression in an AP-1 dependent manner but does not impact c-Jun, JunB, JunD, Fra1 or

Il33 expression.

(A) qPCR analysis shows increased expression of Csf2 in AdCre-Adam17

-/- keratinocytes treated with

PMA. This is abrogated in the presence of AP-1 inhibitor (Tanshinone IIA). Comparable gene expression

of AP-1 members cjun, junb, jund and fra1 in AdGFP-control and AdCre-Adam17

keratinocytes. (B)

Adenoviral induction of ectopic Notch (AdNICD) abolishes Csf1 and Csf2 expression following PMA

treatment. *P<0.05, mean ± S.E.M. (n=3). All data are representative of three independent experiments.

Page 162: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

142

not alter gene expression of either Csf1, Il33 cytokines or the other AP-1 members (c-

jun, junb, jund, fra1, fra2) (Figure 4.12A). These data show that the loss of Adam17

results in increased AP-1 driven synthesis of the above epithelial cytokines.

We then measured AP-1 activity in an independent cell type (mouse embryonic

fibroblasts, MEFs) and observed that Adam17

MEFs had higher basal AP-1 luciferase

reporter activity. This was enhanced after PMA treatment and reduced by a c-Fos/AP-1

inhibitor (T-5224) (Figure 4.11E). More importantly, transduction of active Notch

(AdNICD) in AdCre-Adam17

keratinocytes ablated the expression of Csf2, Csf3 and

Tslp (Figure 4.11F; Figure 4.12B). In addition, AdNICD abolished both basal and PMA-

induced AP-1 luciferase reporter activity in Adam17

MEFs (Figure 4.11E). We

therefore concluded that ADAM17-dependent Notch activation inhibits AP-1

transcriptional activity, with c-Fos being a relevant AP-1 candidate.

4.3.9 Adenoviral delivery of active Notch rescues local Th2 immunity and

myeloproliferation in Adam17ep

mice

Finally, to test whether Notch could protect Adam17ep

mice from AD, we utilized a

protocol to induce ectopic Notch activity in ADAM17 deficient epidermis (schematic in

Figure 4.13A). Adenoviral delivery of active Notch (AdNICD) into Adam17ep

mice

showed a measurable increase in Notch target gene expression by qPCR analysis of

epidermal scrapings (Figure 4.13B). Importantly, CD4+ T cell activation was abrogated in

dorsal dLNs and a loss of effector CD4+ T cells was observed in AdNICD treated mice

(Figure 4.13C, D). Dorsal delivery of AdNICD also partially rescued MPD resulting in a

Page 163: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

143

decrease in bone marrow Gr-1+CD11b

+ cells (Figure 4.13E). More importantly, a

significant replenishment of B220+ lymphocytes was seen in AdNICD treated mice

(2.93% in AdGFP vs. 13.47% in AdNICD, Figure 4.13F). Thus, active Notch was

sufficient to rescue AD as well as MPD in Adam17ep

mice. Altogether these data show

that ADAM17 activates Notch and this in turn interferes with c-Fos recruitment to the

Tslp and Csf3 promoters to keep epithelial cytokine production in check (outlined in

Figure 4.13G).

4.4 Discussion

This study provides the first demonstration, to our knowledge, of the

physiological requirement of ADAM17 in Notch signaling. We show that ADAM17

provides an inherent permissive signal for basal Notch activation throughout adult life. It

triggers Notch signaling to regulate c-Fos activation, thus maintaining epidermal barrier

homeostasis. Loss of this control induces the production of cytokines or „alarmins‟ which

catalyze local and systemic immune responses even in the absence of an overt exogenous

stress, as illustrated in Figure 4.14. Independent studies have shown that the loss of

Notch, deregulation of AP-1 or overproduction of epithelial TSLP result in inflammatory

skin disease [328, 330, 331, 353]. The ADAM17/Notch/c-Fos triad uncovered in our

study reveals a mechanistic underpinning of all these observations.

Page 164: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

144

Figure 4.13Ectopic Notch activity rescues atopic dermatitis and myeloproliferative

disease in Adam17ep

mice.

(A) Experimental protocol to re-introduce active Notch in dorsal skin of ep mice by subcutaneous

delivery of adenoviral vector harboring NICD (AdNICD). Adenoviral GFP (AdGFP) was used as a control.

(B) qPCR analysis of Notch target genes Hes1 and Herp1 in dorsal epidermis at experimental endpoint.

Bars depict mean expression (n=4). (C, D), FACS analysis of dorsal dLNs to measure CD4+ T cell

activation by cell surface CD69 levels (C), and CD4+CD62L

-CD44

+ effector T cells (D) in ep mice

treated with AdGFP or AdNICD. (E, F), Partial rescue of myeloproliferative disease in bone marrow of

ep mice as measured by FACS analysis of Gr-1+CD11b

+ cells (E) and B220

+ cells (F). (G) ADAM17 is

required for ligand-independent Notch activation, which in turn antagonizes c-Fos transcriptional activity at

promoters of epithelial cytokines, thereby checking their production. *P<0.05 mean ± S.E.M, A, B (n=4)

and C-E (n=3). Data in C-E are representative of three independent experiments. Flow cytometry plots

represent 4 mice per condition.

Page 165: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

145

Figure 4.14 – An illustration of epithelial-immune crosstalk as directed by the

ADAM17/Notch/c-Fos triad.

Notch represses pro-inflammatory c-Fos/AP-1 transcriptional activity in keratinocytes to prevent Th2-

driven atopic dermatitis by TSLP and myeloproliferative disease by G-CSF. ADAM17 induces ligand-

independent Notch activation in keratinocytes, which inhibits c-Fos transcriptional activity on promoter

regions of epithelial cytokines to regulate their production. The loss of ADAM17 reduces Notch activation

and results in the c-Fos-driven production of TSLP and G-CSF. Consequently, a Th2-driven local immune

response is generated in the skin and myeloproliferation is induced in the bone marrow. ADAM17

therefore provides barrier immunity by promoting Notch activation in the skin.

Page 166: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

146

4.4.1 Contexts of ADAM10 and ADAM17 function in Notch signaling

To date, studies of Adam10 knockout mice have established its importance of in

Notch activation during development [123, 124, 128, 354]. On the other hand, the large

majority of reported ADAM17-dependent phenotypes revolve around TNF or EGFR

signaling [76, 98, 133, 136, 286, 339]. Recent in vitro studies have brought to light the

requirement of ADAM17 in ligand-independent Notch activation [138, 355]. Our study

proposes that ligand-independent Notch signaling controls barrier immunity but not

keratinocyte development. The developmental context of Notch activation may be

fulfilled by ADAM10, since mice lacking epidermal Adam10 fail to establish a spinous

layer of the epidermis [128], whereas Adam17ep

mice exhibit precocious keratinocyte

differentiation. Epidermal homeostasis balances cell-autonomous keratinocyte

development and non-autonomous communication between epithelial, stromal and

immune compartments in the skin. By generating inducible deletion of epidermal

Adam17 at 8 weeks of age, we demonstrate the significance of ADAM17 in maintaining

proper epithelial:immune crosstalk in the adult skin.

It is now accepted that Notch activation keeps the epidermal stress response in

check in addition to its established role in keratinocyte development and transformation

[356, 357]. Accumulating evidence suggests that Notch promotes NF-B signaling, but

inhibits AP-1 activity; both these pathways are key components of stress signaling in

keratinocytes but the molecular mechanism by which Notch and AP-1 pathways crosstalk

have remained elusive [331, 341, 349, 353, 358]. While we did not observe defects in

NF-B signaling (data not shown) or rescue AD through Tnfr1 deletion, we elucidated

Page 167: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

147

how ADAM17-dependent Notch activation prevents spontaneous AP-1 activation and

Th2-driven immunity. We probed Notch/AP-1 signal crosstalk with gain/loss of function

approaches, biochemical inhibitors, AP-1 reporter and chromatin immunoprecipitation

assays in independent cell types, and found that 1) c-Fos transcribes TSLP, a key

epithelial cytokine in atopic disease; 2) Active Notch suppresses c-Fos activity by

interfering with its recruitment to cytokine promoters; 3) ADAM17 allows tonic Notch

activation in the skin; and 4) Exogenous Notch ameliorates Th2 immunity caused by

ADAM17 deficiency. Further, the data mining performed in this study indicated a clear

deregulation of the Notch pathway in human atopic dermatitis. Supporting our findings,

polymorphisms have been reported in patients where enhanced AP-1 recruitment to Tslp

and Il4 promoters occurs in several human atopic diseases [352, 359, 360].

Epithelial cells are now recognized as orchestrators of the peripheral immune

response, and several recent studies have identified epithelial mutations causal to

undesired immune cell activation in barrier tissues [327, 361-363]. We demonstrate a

gatekeeper function of ADAM17 in the skin, which may apply to other mucosal tissues

including gut, lung and vaginal epithelium. The epimmunome continues to grow and

understanding the mechanisms that instruct pro- or anti-inflammatory epithelial cytokine

synthesis will be of significant value in the treatment of diseases afflicting barrier tissues.

Page 168: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

148

4.5 Methods

4.5.1 Mice

Adam17fl/fl

mice were generated by Dr. Carl P Blobel. K14-Cre transgenic mice

(Tg(Krt14-Cre)1Amc/J) and Tnfr1-/-

mice (C57BL/6-Tnfrsf1atm1Imx

/J) were originally

obtained from Jackson laboratories. Mice were fed 5%-fat chow ad libitum, housed and

cared for in accordance with protocols approved by the Canadian Council for Animal

Care and the Animal Care Committee of the Ontario Cancer Institute.

4.5.2 Microarray Gene Expression Analysis

Gene expression datasets were obtained from the National Center for

Biotechnology Information Gene Expression Omnibus (GEO) database under the

accessions GSE16161, GSE13355, GSE6012 and GSE6710. Data was utilized in the

processed form from its original study. A gene set comprising members of the Notch

pathway was constructed using information from additional references provided in

Supplemental Data and literature cited in main text. Probes mapping to these genes were

identified on the Affymetrix HGU133A (GSE6710 and GSE6012, 84 probes – Table S1)

and HGU133 Plus2 platforms (GSE13355 and GSE16161, 142 probes –Table S1).

Differential expression for genes within the Notch set was examined using a welch t-test,

and the False Discovery Rate [364] was employed to control for multiple hypotheses,

using a cutoff of 5.0% (Table 5.1, Appendix A). In each dataset we compared: atopic

dermatitis vs. normal (GSE16161), atopic eczema vs. normal (GSE6012) or psoriasis vs

normal (GSE13355 and GSE6710). Significantly differentially expressed genes were

Page 169: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

149

visualized using matrix2png [365], after collapsing probes to genes at their median value,

and scaling the data to N(0,1). Samples in GSE16161 were subjected to hierarchical

clustering (uncentered correlation and average linkage) for the complete Notch signaling

gene set in Cluster 3.0 [366] and the resulting dendogram was visualized using Java

TreeView [367]. The following references were cited in addition to those listed in the

main text: [196, 342, 368-374].

4.5.3 In vivo delivery of active Notch to rescue epidermal inflammation

Mice aged 14 days were treated with a dorsal subcutaneous injection of

4x109pfu/ml adenovirus encoding active Notch (AdNICD, kindly provided by Dr. Erwin

Wagner, Centro Nacional de Investigaciones Oncologicas, Madrid, Spain) or eGFP (Ad-

eGFP, Vector Biolabs) dissolved in 50l sterile PBS. This treatment was administered

every 48h at P14, P16, P18 and P20. At P21, mice were sacrificed and tissue harvested

for analysis. Notch signaling was confirmed by qPCR analysis of target genes Hes1 and

Herp1 from dorsal epidermis scrapings. Epidermis was obtained by incubating minced

dorsal skin in Liberase Blendzyme (Roche) for 1h at 37oC with periodic vortexing. Flow

cytometry analysis is detailed below.

4.5.4 Isolation and culture of primary keratinocytes

Mouse keratinocytes were obtained from Adam17fl/fl

pups following established

protocols [375]. Cell autonomous Adam17 deletion was generated by transducing

Adam17fl/fl

keratinocytes with adenoviral Cre (AdCre-IRES-GFP, Vector Biolabs) or

Page 170: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

150

adenoviral GFP as control (Ad-eGFP, Vector Biolabs) at an MOI of 3. Notch

overexpression was induced by transduction with AdNICD. For calcium-induced

differentiation and ligand-independent Notch activation, cells were cultured with 1mM

CaCl2 in calcium-free Eagles‟ MEM supplemented with 16% calcium-free (chelated)

serum. For ligand-dependent Notch activation, cells were incubated with a fusion protein

consisting of mouse Delta Like 4 and human-Fc at 1g/ml cross-linked with 1g/ml anti-

human IgG1 (H+L, Pierce). Anti-human IgG1 alone was used as a control. For PMA

treatments (Phorbol 12-myristate 13-acetate, P8139, Sigma), media was changed to

serum-free, calcium-free eagles MEM. Pan AP-1 activity was inhibited by pre-treating

keratinocytes with 20M Tanshinone IIA (Enzo Life Sciences, TanIIA) 18 hours prior to

PMA treatment. TanIIA was maintained for the duration of PMA treatment.

4.5.5 AP-1 luciferase assay and PMA treatment of mouse embryonic

fibroblasts (MEFs)

MEFs were transduced with lentiviral AP-1 firefly luciferase and Renilla

luciferase as a control 48 hours prior to treatment (Cignal AP1 reporter, SABiosciences).

Cells were treated with 10ng/ml PMA in serum-free DMEM for 18 hours. To inhibit c-

Fos/AP-1, MEFs were pre-treated with 20M T-5224 (kindly provided by Dr. Erwin

Wagner) 3 hours prior to PMA treatment. T-5224 was maintained for the duration of

PMA treatment. AP-1 activity was calculated as a ratio of AP-1 luciferase luminescence

divided by constitutive Renilla luciferase luminescence and depicted as relative

luminescence units (RLU).

Page 171: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

151

4.5.6 Culture and treatment of HaCaT cells and chromatin

immunoprecipitation (ChIP)

Human keratinocyte cell line (HaCaT cells) was kindly provided by Dr. Hitoshi

Okada (Ontario Cancer Institute, Toronto, Canada). Cells were maintained in DMEM +

10% FBS and serum-starved overnight prior to any treatment. To inhibit ADAM17

activity, HaCaT cells were treated with 100M TAPI-1 3 hours prior to stimulation with

PMA. To inhibit NICD production, cells were treated with 20M of the -secretase

inhibitor DAPT. Chromatin was obtained and ChIP assay performed using the EZ-ChIP

kit (Millipore). The following antibodies were used for immunoprecipitation: anti-human

IgG1 (H+L, Pierce), anti-NICD (Val1744), anti-p-c-Fos (Cell Signaling), anti-Histone H3

(Abcam). Primer sequences are provided in Table 4.2.

4.5.7 Flow cytometry

For splenocyte analysis, red blood cell lysis was performed using AFCS buffer

prior to staining. Superficial axillary and brachial lymph nodes were denoted as skin-

draining lymph nodes (dLNs). Mesenteric lymph nodes (mLNs) were isolated as controls.

Bone marrow cells were obtained from one femur per mouse. The following antibodies

were used for analysis: anti-CD3 (clone 17A2), anti-CD4 (clone RM4-5), anti-CD8

(clone 53-6.7), anti-B220 (clone RA3-6B2), anti-NK1.1 (clone PK136), anti-F4/80 (clone

BM8), anti-MHCII (clone M5/114.15), anti-CD11c (clone N418), anti-Gr-1 (clone RB6-

8C5), anti-CD44 (clone IM7), anti-CD11b (clone M1/70), anti-CD69 (clone H1.2F3),

Page 172: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

152

anti-CD62L (clone MEL-14), anti-IgG2a (clone m2a-15F8), anti-CD19 (clone ID3), and

anti-IgM (clone II/41). 7-AAD was used to assay cell viability.

4.5.8 Histology & cytokine analysis

Dorsal skin was obtained and dissected longitudinally along dorsal midline and

processed as in [98]. The following stains were applied for immunohistochemical

analysis: Harris‟ hematoxylin and eosin, Toluidine B, anti-Ki67 (Abcam), anti-F4/80 (BD

Biosciences), anti-Keratin 5, anti-Keratin 10, and anti-Filaggrin (all from Covance). All

staining was performed by institutional core facilities. Serum was separated by

centrifugation of whole blood (BD Vaccutainer CPT Cell Preparation Tube) and stored at

-70oC. Cytokine levels were measured by following manufacturer‟s protocols for a

cytokine bead array (Millipore). TSLP (R&D Systems) and IL-33 (eBioscience) levels

were measured by standard ELISA following manufacturer‟s protocols.

4.5.9 Immunoblotting and AP-1 binding assay

Keratinocytes were treated with 10ng/ml PMA for 3 hours. Protein was obtained

from tail epidermis by mortar and pestle homogenization of frozen tissue. Protein from

keratinocytes was obtained by washing cells with ice cold PBS followed by lysis in RIPA

extraction buffer. 10 to 50 μg protein was loaded onto SDS-PAGE gels for

immunoblotting. Nuclear lysate for AP-1 binding assays was obtained using standard

protocols [98]. AP-1 binding to target sequence (TRE oligonucleotide) was measured by

using AP-1 family EZ-TFA Transcription Factor Assay (Millipore). The AP-1 antibodies

Page 173: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

153

listed below were used instead of those provided in the kit. The following anti-mouse

antibodies were used: anti-ADAM17 (Khokha lab), anti-active Notch1 (NICD, Abcam

for 72kda fragment, Cell Signaling for 120kda fragment), anti–phosphorylated JNK (p-

JNK, Thr183/Tyr185), anti-JNK, anti– phosphorylated p44/42 MAPK (p-ERK1/2,

Thr202/Tyr204), anti–p44/42 MAPK (ERK1/2), anti--Tubulin anti-p-c-Fos (all from

Cell Signaling), anti-c-Jun, anti-JunD, anti-c-Fos, anti-Fra1, anti-Fra2 (all from Santa

Cruz), anti-c-Jun (BD Biosciences).

4.5.10 RNA preparation and quantitative RT-PCR

RNA was prepared from frozen dorsal epidermis or fresh keratinocytes using TRIzol

reagent (Invitrogen) following standard protocols. cDNA was obtained using a first-

strand cDNA synthesis protocol (Quanta). Gene expression was measured using SYBR

Green reagent (Quanta) in a 7800HT real-time PCR system (Applied Biosciences). All

gene expression levels were normalized to Actb (β-actin), and fold change was measured

relative to control wild-type samples. qPCR of RNA was used as a negative control. The

amount of each product was calculated using the 2–ΔCT

method. Primer sequences are

provided in Table 5.2.

4.5.11 Statistical Analyses

Data are reported as mean ± S.D. or S.E.M. All calculations were carried out

using GraphPad Prism software (GraphPad Software). Comparisons were made by 2-

tailed Student‟s t test and ANOVA.

Page 174: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

154

Table 4.1 – Probelists and significance values for differentially expressed genes in all

studies. (See Appendix A online).

Table 4.2Primer sequences used for qRT-PCR and ChIP analysis.

Gene Forward & Reverse Primer Sequence

Adam17 F: 5‟- CGGAGGAAGCAGGCTCTG-3‟

R: 5‟- GTTTCTAAGTGTGTCGCAGACTG-3‟

c-Fos F: 5‟- CAGAGCTGGAGCCCCTGTGT -3‟

R: 5‟- CGGCTGCCTTGCCTTCTCTG -3‟

c-Jun F: 5‟- TTGCCCCAACAGATCCCGGT -3‟

R: 5‟- TGCGGTTCCTCATGCGCTTC -3‟

Csf1 F: 5‟- TGGCCCATGGGAGACCAGGA -3‟

R: 5‟- GGGCTGGGTGCCTTTATGCC -3‟

Csf2 F: 5‟- ACATGTGTGCAGACCCGCCT -3‟

R: 5‟- TCCGTTTCCGGAGTTGGGGG -3‟

Csf3 F: 5‟- TGACCAGGGGAACGGCCTCT -3‟

R: 5‟- GCTGCAGCCCAGATCACCCA -3‟

Filaggrin (Fil) F: 5‟- GTGGCCAACAGAGAATGAG -3‟

R: 5‟- ATGATGCCCAGAACTATGTGAC -3‟

Fra1 F: 5‟- AACGCGGACCCTACCGAACA -3‟

R: 5‟- GGCTGCTGCTGTCGATGCTT -3‟

Hes1 F: 5‟- AAAGCCTATCATGGAGAAGAGGCG -3‟

R: 5‟- GGAATGCCGGGAGCTATCTTTCTT -3‟

Hes5 F: 5‟- AAAGCCTATCATGGAGAAGAGGCG -3‟

R: 5‟- GGAATGCCGGGAGCTATCTTTCTT -3‟

Hey1 F: 5‟- ACACTGCAGGAGGGAAAGGTT -3‟

R: 5‟- CAAACTCCGATAGTCCATAGCCA -3‟

Hey2 F: 5‟- AAGCGCCCTTGTGAGGAAAC -3‟

R: 5‟- GGTAGTTGTCGGTGAATTGGAC -3‟

Human Csf3 F: 5‟- CCCAGAGCCCCATGAAGCTG -3‟

R: 5‟- GGTGGCACACTCACTCACCAG -3‟

Human Tslp F: 5‟- AGAAAGCTCTGGAGCATCAGGGA -3‟

R: 5‟- ACATACGTGGACACCCAATTCCAC -3‟

Il33 F: 5‟- TTTATGAAGCTCCGCTCTGGCC -3‟

R: 5‟- GCTGTTGACAGGCAGCGAGTA -3‟

JunB F: 5‟- ACATGCACCACCCTTTGCGG -3‟

R: 5‟- CAGCCGCTTTCGCTCCACTT -3‟

Page 175: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

155

JunD F: 5‟- AGGTGGCAGCTAGCGAGGAG -3‟

R: 5‟- TCAGGTTGGCGTAGACCGGG -3‟

Keratin 1 (K1) F: 5‟- GACACCACAACCCGGACCCAAAACTTAGAC -

3‟

R: 5‟- ATACTGGGCCTTGACTTCCGAGATGATG -3‟

Keratin 10 (K10) F: 5‟- GCCAGAACGCCGAGTACCAACAAC -3‟

R: 5‟- GTCACCTCCTCAATAATCGTCCTG -3‟

Keratin 14 (K14) F: 5‟- TGCTGGATGTGAAGACAAGG -3‟

R: 5‟- GGATGACTGAGAGCCAGAGG -3‟

Keratin 5 (K5) F: 5‟- TCTCTTCTGGCTACGGAGGA -3‟

R: 5‟- GAAGCTCATGCCTCCTTGAC -3‟

Loricrin (Lor) F: 5‟- TTGCAACGGAGACAACAGAG -3‟

R: 5‟- GCGACTCAATGGCTTCTTCT -3‟

Transglutaminase 1

(Tgm1)

F: 5‟- GCGGAGGGCTGTGGAGAAGG -3‟

R: 5‟- GGGTGCGCAAACGG AA GGTG -3‟

Transglutaminase 3

(Tgm3)

F: 5‟- GCTTGGGGGTTCGCTCTCG -3‟

R: 5‟- CTGCCTACTGCCTTGGTGCTGAT -3‟

Tslp F: 5‟- GAGGACTGTGAGAGCAAGCCAG -3‟

R: 5‟- GGCAGTGGTCATTGAGGGCTT -3‟

-Actin (Actb) F: 5‟- ACGAGGCCCAGAGCAAGAGA -3‟

R: 5‟- GTGAGCAGCACAGGGTGCT -3‟

ChIP: Human Csf3 F: 5‟- TGGCCAGAGCTGGGAGGCAT -3‟

R: 5‟- AGGCACCTGCCTGGCCCTAA -3‟

ChIP: Human Tslp F: 5‟- ACCCTCCATTGGTGTTGCTGGA -3‟

R: 5‟- GCATCGAGATGGCCGGGCTCA -3‟

Page 176: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

156

5 CHAPTER 5

FUTURE DIRECTIONS

Page 177: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

157

This thesis investigated the contributions of the metalloproteinase ADAM17 and

its endogenous inhibitor TIMP3 to acute and chronic inflammation. The insights gained

from the described studies introduce the tissue-specific roles of TIMPs in the epithelial

stress response, immune cell development and function. Proper immunity involves

communication between the afflicted tissue and resident or local immune cells, which

then calls upon context-dependent innate and/or adaptive immune effectors to eliminate

the source of tissue stress. Simultaneously a cellular repair program is initiated in the

damaged tissue, and this is now thought to depend on signals generated by active immune

cells in the local microenvironment. Following eradication or containment of infection,

an immunoregulatory program is elicited to shut down the immune response and prevent

autoimmunity. Cytokines are key messengers in each of the above steps; this thesis

demonstrates that metalloproteinase-dependent regulation of cytokine production and

signaling is a critical axis in the immune response.

A second insight gained from these studies is the observation that epithelial cells

are not bystanders in the immune response. The pervading notion for several years was

that epithelial cells act in a „responsive‟ manner to exogenous insults. Specifically for

mucosal epithelia, dogma maintained that these cells primarily provided physical barriers

to the external environment, or acted as biochemical „effector‟ cells that metabolized

toxin and died off, to be cleared by local immune components or neighboring epithelial

cells. Several lines of evidence in the study of mucosal immunity is largely revising this

notion, establishing the epithelial cell as a „regulatory‟ unit directing local and systemic

immune cell function in a very context-dependent manner [327].

Page 178: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

158

Chapters 2, 3 and 4 show how epithelial metalloproteinases directly impact

cytokine biology and cellular immunity. In chapter 2 we show how control of ADAM17-

mediated ectodomain shedding balances pro-apoptotic signaling via Fas and TNF with

pro-survival signaling by EGFR. Chapter 3 demonstrates that non-hematopoietic tissues

are the relevant source of TIMP3 in vivo, and this metalloproteinase inhibitor plays an

immunosuppressive role in preventing undesired immune cell egress into the periphery.

Focusing on ADAM17, chapter 4 develops a new mouse model of atopic dermatitis-like

skin disease by Adam17 deletion in keratinocytes. An important yet unknown role of

ADAM17 in regulating tonic Notch signaling is revealed, where epithelial Notch

activation by ADAM17 prevents epithelial cytokine transcription by c-Fos/AP-1. Thus

we show how the control of epithelial stress signaling by metalloproteinases is an

upstream regulator of mucosal immunity. Throughout each chapter, specific cytokines are

identified as key effectors in guiding cell death, survival, acute and chronic inflammation.

Armed with this knowledge of epithelial/stromal metalloproteinase function in

immunity, new avenues can be pursued to better understand the unique and redundant

roles of Timp genes in mammalian biology. In addition to Timp3, we observed that Timp1

gene expression was also elevated in wildtype mice following death receptor activation

(Figure 2.4A). Given the previous observation that TIMP1 regulates hepatocyte growth

factor (HGF) signaling during liver regeneration [54], it is relevant to pursue the role of

TIMP1 in hepatocyte apoptosis during fulminant liver failure as described in chapter 2.

To date our investigations have focused on individual TIMP deficiencies, however it

remains unknown whether TIMPs act redundantly in regulating metalloproteinase

Page 179: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

159

activity throughout the mammalian lifespan. The notion that TIMPs may act as stromal

factors (as demonstrated in chapter 4) are readily investigated in models of immune cell

development, specifically the thymic and bone marrow niches. The introduction of

compound knockout mice harboring multiple TIMP deficiencies allows the study of

potential in vivo redundancies during T and B lymphopoiesis and myelopoiesis.

The ability temporally induce epidermal deletion of Adam17 (generated in chapter

5) has led to the development of more sophisticated murine models to test antigen-

specific barrier immunity in the skin, allowing us to investigate how epidermal ADAM17

impacts immune cell mobilization in clinically relevant models of inflammatory skin

disease. The following sections describe future directions which have emerged from this

thesis, focusing on antigen-specific atopic dermatitis, mechanical cutaneous injury and

the investigation of epithelial TIMPs and ADAM17 in thymopoiesis. An overarching

goal of these future directions is to enhance our understanding of regulated ectodomain

shedding in epithelial:immune crosstalk.

5.1 Manipulating ADAM17 and TIMPs in models of adult skin

inflammation

5.1.1 Antigen-specific atopic dermatitis

Our investigation of epidermal ADAM17 in atopic dermatitis revealed that altered Timp3

gene expression is the most significant change in involved skin from whole genome

analysis of human psoriasis patients (unpublished observations). We have also

consistently noted that aged mice lacking both Timp1 and Timp3 develop spontaneous

Page 180: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

160

dermatitis. These observations suggest that regulation of ectodomain shedding by TIMPs

in the skin impacts local inflammation in humans and mice. In chapter 4, we generated an

inducible model of epidermal Adam17 deletion (Adam17epER

mice). These allowed us to

generate ADAM17 deficiency in adult mice and recapitulate a more clinically relevant

phenotype which was not as severe as that observed in A17ep

mice. Moving the genetic

models forward, both Timp1

Timp3

and Adam17epER

mice will be of significant

utility in studying antigen-driven epidermal inflammation. R. Geha and colleagues

illustrate a protocol to induce atopic dermatitis in an antigen-dependent fashion using

repeated topical administration of chicken ovalbumin (OVA) as a protein allergen

(outlined in Figure 5.1A) [376, 377]. This method provides an ideal system to study the

role of TIMP1, TIMP3 and ADAM17 in progressive skin inflammation when compared

to the severe spontaneous disease seen in Adam17ep

mice (discussed in chapter 4). OVA-

mediated induction of atopic dermatitis will allow us to pose the question of how

epidermal TIMP1, TIMP3 and ADAM17 direct lymphocyte infiltration and effector

function, and dendritic cell activation in the skin microenvironment.

In addition to OVA sensitization described above, the use of established T cell

receptor (TCR) transgenic mice will be used to ask whether control of epithelial

ectodomain shedding regulates effector function of “antigen-experienced” CD4+ T cells

in the periphery. In this system, Timp3

, Timp1

Timp3

and Adam17epER

mice are

crossed with OT-II transgenic mice, which are then treated with OVA peptide fragment

(amino acids 323-339). Presumably, the presence of OVA-specific CD4 co-receptor in

Page 181: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

161

these mice will significantly accelerate atopic dermatitis compared to the sensitization

model and generate a more acute form of skin inflammation.

Antigen presentation by skin-resident dendritic cells is a key step in maintaining

tolerance to commensals or non-pathogenic antigens while generating cellular and

humoral immunity against invading pathogens. In chapter 4 we identified that resting

splenic CD4+ T cells expressed low levels of Timp genes and thus stromal tissues were

the biologically relevant source of TIMP3 in T-cell mediated autoimmune hepatitis.

However all subsets of human dendritic cells (DCs) are thought to transcribe Timps in

response to infection [378, 379]. In a physiological context, migration of epidermal DCs

(called Langerhans cells) and dermal DCs from the skin to local draining lymph nodes

(dLNs) is required for antigen presentation and generation of immunity. Previous studies

have shown that cell-autonomous MMP2 and MMP9 are necessary for DC migration

through the skin microenvironment. Furthermore DCs produce TIMP1 and 2 to

antagonize undesired MMP activity in an autoregulatory mechanism of balancing

invasion and tissue residence [380, 381]. The genetic models listed in this section allow

us to test the role played by TIMPs in DC migration to dLNs following antigen

encounter, and importantly investigate the capacity of TIMP or ADAM17 deficient DCs

to induce Th2-polarization of dLN-resident T cells. As outlined in Figure 5.1B, a typical

method would involve tape stripping of shaved dorsal skin followed by FITC painting.

Subsequently dLNs would be probed for FITC+CD11c

+ DCs that can polarize T cells.

These cells would then be isolated and co-cultured with OT-II TCR-OVA CD4+ T cells

in the presence of OVA peptide. The observations that Timp1

Timp3

mice exhibit

Page 182: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

162

Figure 5.1 – Methods of analyzing antigen-specific responses in atopic dermatitis. (A) OVA sensitization protocol over 50 days, followed by analysis of skin inflammation. (B) Mechanical

injury followed by FITC painting is used to study dermal DC activation and polarization of target T cells.

In our model FITC+ DCs are co-cultured with naïve OT-II TCR-OVA CD4

+ T cells to compare the effect

of individual or multiple Timp or Adam17 deletion.

Page 183: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

163

spontaneous dermatitis lead to a hypothesis that DCs lacking Timp1 and Timp3 or

Adam17 would induce significantly greater Th2-polarization of TCR-OVA CD4+ T cells

and result in enhanced cytokine production.

Independent studies have demonstrated that epidermal sensitization to allergens

leads to enhanced airway hyperresponsiveness to the same antigen and generates asthma

[328, 377]. This is consistent with the progression of the human atopic march, where

atopic dermatitis is coupled with bronchial asthma, allergic rhinitis and forms of

gastrointestinal allergic inflammation (e.g. anaphylaxis, ulcerative colitis) [327, 382,

383]. The protective role of ADAM17 in gut epithelium was recently described by two

studies where compromised EGFR activation led to defective epithelial regeneration in

models of colitis [384, 385]. Additionally the in vitro wound healing response of

keratinocytes was also shown to rely on ADAM17-mediated EGFR activation [136]. An

important question in barrier immunity is whether similar mechanisms operate in

different atopic diseases. The models described above have the potential to identify the

contributions of metalloproteinase function in several atopic diseases.

5.1.2 Mechanical cutaneous injury

Repeated tape stripping of the skin recapitulates cutaneous injury that has been

used to study plasmacytoid dendritic cell (pDC) recruitment to the dermis, specifically

Toll-like receptor function and type 1 interferon signaling in response to self-nucleic

acids released by injured keratinocytes [378, 386, 387]. The tape strip test is also used

clinically to diagnose cutaneous lupus, as patients display an exaggerated inflammatory

Page 184: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

164

response to mild skin injury when compared to healthy controls [387-389]. Here, an

antigen-specific response (such as that induced by OVA) is not tested. Rather, the wound

healing response induced by pDC activation is a primary focus. The role of TIMPs in

epithelial migration during wound healing remains enigmatic, with conflicting reports

showing beneficial and harmful consequences of metalloproteinase inhibition in

keratinocyte migration [390, 391]. Mice lacking individual or multiple TIMP genes

would provide valuable insight in the study of cutaneous wound healing through control

of pDC activation, interferon signaling or keratinocyte migration. A typical method

would include repeated tape stripping of shaved dorsal skin of TIMP deficient mice and

controls, followed by analysis of involved skin and local draining lymph nodes for pDC

recruitment, cytokine production and the wound healing response at early (12-48h) and

late (7-21 days) timepoints. Together, these systems can potentially enhance our

knowledge of ectodomain shedding in epithelial:immune crosstalk using genetic and

inducible models of cutaneous inflammation.

5.2 Redundancy of TIMPs in hematopoietic niches

5.2.1 TIMPs in the thymic stroma

5.2.1.1 Cell autonomous and stromal effects of Timps on T lymphopoiesis

Petrie and colleagues have mapped the global gene expression patterns in compartments

of the thymic stroma (cortical, medullary, the cortico-medullary junction) as well as

developing thymocytes; TIMP genes are exclusively relegated to the stromal

compartment, while ADAM and MMP expression is more heterogeneous [392, 393].

Page 185: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

165

Also, quantitative RT-PCR analysis shows that Timp1, 2 and 4 are expressed at higher

levels by the modified bone marrow stromal cell line OP9-DL1 compared to OP9 control

cells (Figure 5.2). OP9-DL1 cells stably express high levels of the Notch ligand Delta-

like 1 (DL1) and preferentially induce T cell differentiation of uncommitted

hematopoietic progenitors in vitro [394]. Preliminary investigations of T cell

development using the fetal thymus organ culture (FTOC, protocol outlined in Figure

5.3A) system show that: i) small-molecule inhibition of MMPs (using GM6001) and

ADAMs (using TAPI-1) delays or arrests T cell development in a dose dependent manner

(Figure 5.4); and ii) additive loss of TIMPs alters the kinetics of T lymphopoiesis (Figure

5.5). These genetic models are new tools unique to our lab, and permit more thorough

investigation of the fundamental role of Timp genes in thymopoiesis.

Focusing on epithelial: immune crosstalk, we have asked whether regulation of stromal

versus hematopoietic MMPs/ADAMs differentially impacts thymopoiesis. Here the

chimeric FTOC system has utility, where either the stromal or hematopoietic

compartment is deleted for genes of interest. Briefly, Timp mutant FTOCs will be treated

with 2-deoxyguanosine to deplete endogenous hematopoietic cells, and then re-seeded

with congenic (CD45.1) wildtype progenitors (lineage depleted cKit+Sca-1

+) obtained

from E14.5 fetal livers (protocol outlined in Figure 5.3B) [395]. This will functionally

test the role of stromal TIMP genes in development of normal T cell progenitors, and T

cell development will be followed over 21 days.

Page 186: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

166

Figure 5.2 Expression of Timp genes in bone marrow stromal cell line OP9-DL1

compared to OP9-GFP. OP9-GFP: blue; OP9-DL1: purple. Data are mean ±S.D. (n=3).

Page 187: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

167

Figure 5.3 Methods of analyzing hematopoietic vs. epithelial contributions of TIMPs

and ADAM17 in thymopoiesis using fetal thymus organ culture (FTOC).

(A) Protocol for ex vivo treatment of fetal thymi with small-molecule metalloproteinase inhibitors. (B)

Protocol for generating chimeric FTOC where ADAM17 or TIMP deficient thymi are treated with 2-

deoxyguanosine (2-dGuo) to eliminate hematopoietic cells. These are subsequently seeded with congenic

(CD45.1+) hematopoietic progenitors and T cell development followed via flow cytometry.

Page 188: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

168

Figure 5.4 – Pharmacological inhibition of ADAM17 activity arrests T cell development

in a dose-dependent manner. E15.5 fetal thymi were cultured ex vivo for 4 days in the presence of TAPI-1 and T cell development

assessed by flow cytometry. 7-AAD: viability marker. TAPI-1: TNF Alpha Protease Inhibitor-1.

Page 189: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

169

Figure 5.5 – Additive loss of Timp genes impacts T cell development with a gene-

dependent decrease in double-negative 1 (DN1) populations.

E15.5 fetal thymi from pups of indicated genotypes were culture ex vivo up to 4 days. CD3

-CD4

-CD8

- T

cells were analyzed for kinetics of T cell development by flow cytometry. Data are representative of 4 fetal

thymi per condition.

Page 190: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

170

5.2.1.2 Mechanisms underlying altered T cell development in Timp or Adam17

null mice

Notch signaling is a key component of T cell development and its ablation

induces ectopic B lymphopoiesis in the thymus, as Notch activity is required to suppress

thymic B cell development while permitting T cell development [182, 396, 397].

Specifically, Notch1 provides survival signals of uncommitted hematopoietic progenitors

and later, signaling through Notch1 and its transcriptional co- activator Mastermind-like

(MAML) is required for pre-TCR expression and conventional T cell development

past the double negative 3 (DN3) stage [398-400]. On the other hand, Notch appears

dispensable for T cell development in the thymus. A fraction of this unconventional T

cell subset matures in epithelial tissues, and the epidermis harbors the largest population

of T cells in the periphery [401, 402].

Both ADAM10 and ADAM17 are implicated in processing Notch via the S2

cleavage, which occurs prior to the release of the intracellular signaling domain required

to activate transcription of target genes. We thus rationalize that deletion of multiple

Timps will lead to a loss of regulation over ADAM10/17 activity and disrupt Notch

signaling during T cell development. To this end, developing T cells can be sorted at

different stages of maturation based on cell surface CD25 and CD44 expression in

CD4/CD8 double negative (DN) populations: (DN1: CD44+CD25

-; DN2: CD44

+CD25

+;

DN3a: CD44+CD25

-CD27

low intracellularTCR

-; DN3b: CD44

+CD25

-CD27

high

intracellularTCR+; DN4:CD44

-CD25

-TCR

+ or TCR

+ [403]) and analyzed for Notch

target gene expression. Notch activity can also be independently verified using CBF1-

Page 191: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

171

eGFP reporter mice generated by Dr. Gaiano‟s group [404]. Thymic progenitors derived

from these mice will be seeded into FTOC (as in Figure 5.3B) with the selected Timp

mutants mentioned above to measure the impact of Timp deficient thymic epithelium on

Notch signaling.

We have observed that K14-Cre;Adam17fl/fl

mice have altered thymic homeostasis. Since

Keratin14 is expressed in the medullary epithelium of the thymus, we asked whether

Adam17 deletion in this compartment influenced T cell number. A ten-fold decrease in

thymocyte numbers was noted in K14-Cre;Adam17fl/fl

mice (Figure 5.6A). We also

observed disruption of the thymic architecture postnatally, specifically a decrease in the

cellularity of the thymic cortex, where the majority of CD4+CD8

+ T cells reside (Figure

5.6B, C).

Importantly, Lck-Cre;Adam17fl/fl

mice mice did not display this phenotype. Thus

in parallel to the study of TIMP function in T cell development, we will test whether

ADAM17 in the thymic epithelium is required for growth factor signaling (such as

EGFR) and/or developmental cues through Notch. Since our initial observations,

independent work by Vignali and colleagues has confirmed that non-hematopoietic

ADAM17 can impact T cell development [201]; however this study does not directly

address the role of ADAM17 in the thymic epithelium in normal T cell development

since comparisons are made between ADAM17 deficient mice and chimeric genotypes

lacking hematopoietic but not epithelial ADAM17. The authors conclude that since

conventional ADAM17 deficient mice exhibit perturbed thymopoiesis compared to those

Page 192: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

172

Figure 5.6 – Decreased thymic cellularity in K14-Cre;Adam17fl/fl

thymus.

(A) Total thymic cell counts of indicated genotypes were obtained from 6-week old mice. (B) Flow

cytometry analysis shows a proportional decrease in CD4+CD8

+ T cells in K14-Cre-Adam17

fl/fl mice. (C)

Histological analysis shows decreased cellularity of the thymic cortex (C) in K14-Cre;Adam17fl/fl

mice. C:

Cortex; M: Medulla. Flow cytometry and histology are representative of 3 mice per condition.

Page 193: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

173

lacking hematopoietic ADAM17, the relevant compartment of ADAM17 production is

the thymic epithelium. Our use of mice lacking ADAM17 in the thymic medulla will

provide fundamental insights into the contribution of epithelial ectodomain shedding in

thymocyte development.

Page 194: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

174

5.3 Concluding Remarks

Investigation of intracellular signal transduction has vastly contributed to our

understanding of cytokine bioactivity, however, much less is known regarding the

extracellular control of cytokine networks. Numerous investigations of intracellular

signal crosstalk have highlighted the control of heterologous receptor activation and of

intracellular signaling nodes in immunity. Such depth of understanding is lacking with

regards to the extracellular microenvironment where discrete signals simultaneously

converge on the cell surface or where multiple cell types communicate with each other.

An adequate immune response depends on a rapid response to infection, and cytokines

are the messengers that drive this response. Proper integration of cytokine networks

maintains tissue homeostasis and prevents chronic inflammation, autoimmunity and

tumorigenesis. In addition to the studies referenced throughout this thesis, work provided

herein has established the importance of proteolytic cleavage by metalloproteinases in

numerous aspects of immunity. It has provided insights on the lateral control of multiple

cytokines by TIMPs during injury and inflammation, and shown that

compartmentalization of ADAM17-dependent ectodomain shedding can regulate

crosstalk between epithelial and immune cells to guide tissue homeostasis. Nonetheless,

the role of TIMPs and metalloproteinases in immunity still remains a largely undeveloped

field. The principles governing ectodomain shedding described throughout this work

potentially apply to numerous examples of immunopathologies, tumor immunity and

normal tissue homeostasis and further our understanding of the immune response.

Page 195: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

175

6 References

1. Jalkanen, M., Biology of cell surface heparan sulfate proteoglycans. Med Biol,

1987. 65(1): p. 41-7.

2. Koda, J.E., A. Rapraeger, and M. Bernfield, Heparan sulfate proteoglycans from

mouse mammary epithelial cells. Cell surface proteoglycan as a receptor for

interstitial collagens. J Biol Chem, 1985. 260(13): p. 8157-62.

3. Peschon, J.J., et al., An essential role for ectodomain shedding in mammalian

development. Science, 1998. 282(5392): p. 1281-4.

4. Khokha, R. and D.T. Denhardt, Matrix metalloproteinases and tissue inhibitor of

metalloproteinases: a review of their role in tumorigenesis and tissue invasion.

Invasion Metastasis, 1989. 9(6): p. 391-405.

5. Maidment, J.M., et al., Matrix metalloproteinase homologues from Arabidopsis

thaliana. Expression and activity. J Biol Chem, 1999. 274(49): p. 34706-10.

6. Murphy, G., et al., Role of TIMPs (tissue inhibitors of metalloproteinases) in

pericellular proteolysis: the specificity is in the detail. Biochem Soc Symp,

2003(70): p. 65-80.

7. Brew, K., D. Dinakarpandian, and H. Nagase, Tissue inhibitors of

metalloproteinases: evolution, structure and function. Biochim Biophys Acta,

2000. 1477(1-2): p. 267-83.

8. Cawston, T.E., et al., Purification of rabbit bone inhibitor of collagenase.

Biochem J, 1981. 195(1): p. 159-65.

9. Docherty, A.J., et al., Sequence of human tissue inhibitor of metalloproteinases

and its identity to erythroid-potentiating activity. Nature, 1985. 318(6041): p. 66-

9.

10. Edwards, D.R., et al., A growth-responsive gene (16C8) in normal mouse

fibroblasts homologous to a human collagenase inhibitor with erythroid-

potentiating activity: evidence for inducible and constitutive transcripts. Nucleic

Acids Res, 1986. 14(22): p. 8863-78.

11. Khokha, R., et al., Antisense RNA-induced reduction in murine TIMP levels

confers oncogenicity on Swiss 3T3 cells. Science, 1989. 243(4893): p. 947-50.

12. Brenner, C.A., et al., Genes for extracellular-matrix-degrading

metalloproteinases and their inhibitor, TIMP, are expressed during early

mammalian development. Genes Dev, 1989. 3(6): p. 848-59.

13. Waterhouse, P., D.T. Denhardt, and R. Khokha, Temporal expression of tissue

inhibitors of metalloproteinases in mouse reproductive tissues during gestation.

Mol Reprod Dev, 1993. 35(3): p. 219-26.

14. Parks, W.C., C.L. Wilson, and Y.S. Lopez-Boado, Matrix metalloproteinases as

modulators of inflammation and innate immunity. Nat Rev Immunol, 2004. 4(8):

p. 617-29.

Page 196: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

176

15. Khokha, R., et al., Suppression of invasion by inducible expression of tissue

inhibitor of metalloproteinase-1 (TIMP-1) in B16-F10 melanoma cells. J Natl

Cancer Inst, 1992. 84(13): p. 1017-22.

16. Apte, S.S., M.G. Mattei, and B.R. Olsen, Cloning of the cDNA encoding human

tissue inhibitor of metalloproteinases-3 (TIMP-3) and mapping of the TIMP3

gene to chromosome 22. Genomics, 1994. 19(1): p. 86-90.

17. Gasson, J.C., et al., Molecular characterization and expression of the gene

encoding human erythroid-potentiating activity. Nature, 1985. 315(6022): p. 768-

71.

18. Greene, J., et al., Molecular cloning and characterization of human tissue

inhibitor of metalloproteinase 4. J Biol Chem, 1996. 271(48): p. 30375-80.

19. Leco, K.J., et al., Murine tissue inhibitor of metalloproteinases-4 (Timp-4): cDNA

isolation and expression in adult mouse tissues. FEBS Lett, 1997. 401(2-3): p.

213-7.

20. Stetler-Stevenson, W.G., H.C. Krutzsch, and L.A. Liotta, Tissue inhibitor of

metalloproteinase (TIMP-2). A new member of the metalloproteinase inhibitor

family. J Biol Chem, 1989. 264(29): p. 17374-8.

21. Dean, G., et al., The human tissue inhibitor of metalloproteinases (TIMP)-1 gene

contains repressive elements within the promoter and intron 1. J Biol Chem,

2000. 275(42): p. 32664-71.

22. Edwards, D.R., et al., Involvement of AP1 and PEA3 binding sites in the

regulation of murine tissue inhibitor of metalloproteinases-1 (TIMP-1)

transcription. Biochim Biophys Acta, 1992. 1171(1): p. 41-55.

23. Leco, K.J., et al., Differential regulation of TIMP-1 and TIMP-2 mRNA

expression in normal and Ha-ras-transformed murine fibroblasts. Gene, 1992.

117(2): p. 209-17.

24. Leco, K.J., et al., Tissue inhibitor of metalloproteinases-3 (TIMP-3) is an

extracellular matrix-associated protein with a distinctive pattern of expression in

mouse cells and tissues. J Biol Chem, 1994. 269(12): p. 9352-60.

25. Wick, M., et al., Structure of the human TIMP-3 gene and its cell cycle-regulated

promoter. Biochem J, 1995. 311 ( Pt 2): p. 549-54.

26. Pohar, N., T.A. Godenschwege, and E. Buchner, Invertebrate tissue inhibitor of

metalloproteinase: structure and nested gene organization within the synapsin

locus is conserved from Drosophila to human. Genomics, 1999. 57(2): p. 293-6.

27. Wei, S., et al., Drosophila TIMP is a potent inhibitor of MMPs and TACE:

similarities in structure and function to TIMP-3. Biochemistry, 2003. 42(42): p.

12200-7.

28. Lee, M.H., S. Atkinson, and G. Murphy, Identification of the extracellular matrix

(ECM) binding motifs of tissue inhibitor of metalloproteinases (TIMP)-3 and

effective transfer to TIMP-1. J Biol Chem, 2007. 282(9): p. 6887-98.

29. Fernandez-Catalan, C., et al., Crystal structure of the complex formed by the

membrane type 1-matrix metalloproteinase with the tissue inhibitor of

metalloproteinases-2, the soluble progelatinase A receptor. Embo J, 1998.

17(17): p. 5238-48.

Page 197: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

177

30. Iyer, S., et al., Crystal structure of the catalytic domain of matrix

metalloproteinase-1 in complex with the inhibitory domain of tissue inhibitor of

metalloproteinase-1. J Biol Chem, 2007. 282(1): p. 364-71.

31. Morgunova, E., et al., Structural insight into the complex formation of latent

matrix metalloproteinase 2 with tissue inhibitor of metalloproteinase 2. Proc Natl

Acad Sci U S A, 2002. 99(11): p. 7414-9.

32. Huxley-Jones, J., et al., The evolution of the vertebrate metzincins; insights from

Ciona intestinalis and Danio rerio. BMC Evol Biol, 2007. 7: p. 63.

33. Yu, W.P., S. Brenner, and B. Venkatesh, Duplication, degeneration and

subfunctionalization of the nested synapsin-Timp genes in Fugu. Trends Genet,

2003. 19(4): p. 180-3.

34. Murphy, G., A. Murthy, and R. Khokha, Clipping, shedding and RIPping keep

immunity on cue. Trends Immunol, 2008. In press.

35. Mohammed, F.F. and R. Khokha, Thinking outside the cell: proteases regulate

hepatocyte division. Trends Cell Biol, 2005. 15(10): p. 555-63.

36. Fata, J.E., et al., Timp-1 is important for epithelial proliferation and branching

morphogenesis during mouse mammary development. Dev Biol, 1999. 211(2): p.

238-54.

37. Nuttall, R.K., et al., Expression analysis of the entire MMP and TIMP gene

families during mouse tissue development. FEBS Lett, 2004. 563(1-3): p. 129-34.

38. Chirco, R., et al., Novel functions of TIMPs in cell signaling. Cancer Metastasis

Rev, 2006. 25(1): p. 99-113.

39. Chalaris, A., et al., Critical role of the disintegrin metalloprotease ADAM17 for

intestinal inflammation and regeneration in mice. J Exp Med, 2010. 207(8): p.

1617-24.

40. Aimes, R.T. and J.P. Quigley, Matrix metalloproteinase-2 is an interstitial

collagenase. Inhibitor-free enzyme catalyzes the cleavage of collagen fibrils and

soluble native type I collagen generating the specific 3/4- and 1/4-length

fragments. J Biol Chem, 1995. 270(11): p. 5872-6.

41. Xu, J., et al., Proteolytic exposure of a cryptic site within collagen type IV is

required for angiogenesis and tumor growth in vivo. J Cell Biol, 2001. 154(5): p.

1069-79.

42. Chen, M., et al., The recombinant expression of full-length type VII collagen and

characterization of molecular mechanisms underlying dystrophic epidermolysis

bullosa. J Biol Chem, 2002. 277(3): p. 2118-24.

43. Imai, K., et al., Degradation of decorin by matrix metalloproteinases:

identification of the cleavage sites, kinetic analyses and transforming growth

factor-beta1 release. Biochem J, 1997. 322 ( Pt 3): p. 809-14.

44. Tsuruga, E., K. Irie, and T. Yajima, Fibrillin-2 degradation by matrix

metalloproteinase-2 in periodontium. J Dent Res, 2007. 86(4): p. 352-6.

45. Shekhar, M.P., et al., Alterations in galectin-3 expression and distribution

correlate with breast cancer progression: functional analysis of galectin-3 in

breast epithelial-endothelial interactions. Am J Pathol, 2004. 165(6): p. 1931-41.

Page 198: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

178

46. Xu, X., et al., Contributions of the MMP-2 collagen binding domain to gelatin

cleavage. Substrate binding via the collagen binding domain is required for

hydrolysis of gelatin but not short peptides. Matrix Biol, 2004. 23(3): p. 171-81.

47. O'Reilly, M.S., et al., Regulation of angiostatin production by matrix

metalloproteinase-2 in a model of concomitant resistance. J Biol Chem, 1999.

274(41): p. 29568-71.

48. Levi, E., et al., Matrix metalloproteinase 2 releases active soluble ectodomain of

fibroblast growth factor receptor 1. Proc Natl Acad Sci U S A, 1996. 93(14): p.

7069-74.

49. Steinberg, Z., et al., FGFR2b signaling regulates ex vivo submandibular gland

epithelial cell proliferation and branching morphogenesis. Development, 2005.

132(6): p. 1223-34.

50. Kim, T.H., et al., Expression and activation of pro-MMP-2 and pro-MMP-9

during rat liver regeneration. Hepatology, 2000. 31(1): p. 75-82.

51. Creemers, E.E., et al., Deficiency of TIMP-1 exacerbates LV remodeling after

myocardial infarction in mice. Am J Physiol Heart Circ Physiol, 2003. 284(1): p.

H364-71.

52. Ikonomidis, J.S., et al., Accelerated LV remodeling after myocardial infarction in

TIMP-1-deficient mice: effects of exogenous MMP inhibition. Am J Physiol Heart

Circ Physiol, 2005. 288(1): p. H149-58.

53. Roten, L., et al., Effects of gene deletion of the tissue inhibitor of the matrix

metalloproteinase-type 1 (TIMP-1) on left ventricular geometry and function in

mice. J Mol Cell Cardiol, 2000. 32(1): p. 109-20.

54. Mohammed, F.F., et al., Metalloproteinase inhibitor TIMP-1 affects hepatocyte

cell cycle via HGF activation in murine liver regeneration. Hepatology, 2005.

41(4): p. 857-67.

55. Lijnen, H.R., et al., Deficiency of tissue inhibitor of matrix metalloproteinase-1

(TIMP-1) impairs nutritionally induced obesity in mice. Thromb Haemost, 2003.

89(2): p. 249-55.

56. Nothnick, W.B., P.D. Soloway, and T.E. Curry, Jr., Pattern of messenger

ribonucleic acid expression of tissue inhibitors of metalloproteinases (TIMPs)

during testicular maturation in male mice lacking a functional TIMP-1 gene. Biol

Reprod, 1998. 59(2): p. 364-70.

57. Lindroos, P.M., R. Zarnegar, and G.K. Michalopoulos, Hepatocyte growth factor

(hepatopoietin A) rapidly increases in plasma before DNA synthesis and liver

regeneration stimulated by partial hepatectomy and carbon tetrachloride

administration. Hepatology, 1991. 13(4): p. 743-50.

58. Schmidt, C., et al., Scatter factor/hepatocyte growth factor is essential for liver

development. Nature, 1995. 373(6516): p. 699-702.

59. Chen, P., et al., Tissue inhibitor of metalloproteinase-1 deficiency abrogates

obliterative airway disease after heterotopic tracheal transplantation. Am J

Respir Cell Mol Biol, 2006. 34(4): p. 464-72.

Page 199: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

179

60. Kim, K.H., et al., Tissue inhibitor of metalloproteinase-1 deficiency amplifies

acute lung injury in bleomycin-exposed mice. Am J Respir Cell Mol Biol, 2005.

33(3): p. 271-9.

61. Nothnick, W.B., X. Zhang, and H.E. Zhou, Steroidal regulation of uterine edema

and tissue inhibitors of metalloproteinase (TIMP)-3 messenger RNA expression is

altered in TIMP-1-deficient mice. Biol Reprod, 2004. 70(2): p. 500-8.

62. Nothnick, W.B., Tissue inhibitor of metalloproteinase-1 (TIMP-1) deficient mice

display reduced serum progesterone levels during corpus luteum development.

Endocrinology, 2003. 144(1): p. 5-8.

63. Jaworski, D.M., et al., Tissue inhibitor of metalloproteinase-2(TIMP-2)-deficient

mice display motor deficits. J Neurobiol, 2006. 66(1): p. 82-94.

64. Lluri, G., et al., Tissue inhibitor of metalloproteinase-2 (TIMP-2) regulates

neuromuscular junction development via a beta1 integrin-mediated mechanism. J

Neurobiol, 2006. 66(12): p. 1365-77.

65. Jaworski, D.M., et al., Prepulse inhibition and fear-potentiated startle are altered

in tissue inhibitor of metalloproteinase-2 (TIMP-2) knockout mice. Brain Res,

2005. 1051(1-2): p. 81-9.

66. Caterina, J.J., et al., Inactivating mutation of the mouse tissue inhibitor of

metalloproteinases-2(Timp-2) gene alters proMMP-2 activation. J Biol Chem,

2000. 275(34): p. 26416-22.

67. Wang, Z., R. Juttermann, and P.D. Soloway, TIMP-2 is required for efficient

activation of proMMP-2 in vivo. J Biol Chem, 2000. 275(34): p. 26411-5.

68. Cruz-Munoz, W., et al., Enhanced metastatic dissemination to multiple organs by

melanoma and lymphoma cells in timp-3-/- mice. Oncogene, 2006. 25(49): p.

6489-96.

69. Smookler, D.S., et al., Tissue inhibitor of metalloproteinase 3 regulates TNF-

dependent systemic inflammation. J Immunol, 2006. 176(2): p. 721-5.

70. Cruz-Munoz, W., I. Kim, and R. Khokha, TIMP-3 deficiency in the host, but not

in the tumor, enhances tumor growth and angiogenesis. Oncogene, 2006. 25(4):

p. 650-5.

71. Kassiri, Z. and R. Khokha, Myocardial extra-cellular matrix and its regulation by

metalloproteinases and their inhibitors. Thromb Haemost, 2005. 93(2): p. 212-9.

72. Martin, E.L., et al., Differential response of TIMP-3 null mice to the lung insults

of sepsis, mechanical ventilation, and hyperoxia. Am J Physiol Lung Cell Mol

Physiol, 2005. 289(2): p. L244-51.

73. Mahmoodi, M., et al., Lack of tissue inhibitor of metalloproteinases-3 results in

an enhanced inflammatory response in antigen-induced arthritis. Am J Pathol,

2005. 166(6): p. 1733-40.

74. Fedak, P.W., et al., TIMP-3 deficiency leads to dilated cardiomyopathy.

Circulation, 2004. 110(16): p. 2401-9.

75. Gill, S.E., et al., A null mutation for tissue inhibitor of metalloproteinases-3

(Timp-3) impairs murine bronchiole branching morphogenesis. Dev Biol, 2003.

261(2): p. 313-23.

Page 200: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

180

76. Mohammed, F.F., et al., Abnormal TNF activity in Timp3-/- mice leads to chronic

hepatic inflammation and failure of liver regeneration. Nat Genet, 2004. 36(9): p.

969-77.

77. Leco, K.J., et al., Spontaneous air space enlargement in the lungs of mice lacking

tissue inhibitor of metalloproteinases-3 (TIMP-3). J Clin Invest, 2001. 108(6): p.

817-29.

78. Fata, J.E., et al., Accelerated apoptosis in the Timp-3-deficient mammary gland. J

Clin Invest, 2001. 108(6): p. 831-41.

79. Kandalam, V., et al., Early activation of matrix metalloproteinases underlies the

exacerbated systolic and diastolic dysfunction in mice lacking TIMP3 following

myocardial infarction. Am J Physiol Heart Circ Physiol, 2010. 299(4): p. H1012-

23.

80. Ebrahem, Q., et al., Increased neovascularization in mice lacking tissue inhibitor

of metalloproteinases-3. Invest Ophthalmol Vis Sci, 2011.

81. Federici, M., et al., Timp3 deficiency in insulin receptor-haploinsufficient mice

promotes diabetes and vascular inflammation via increased TNF-alpha. J Clin

Invest, 2005. 115(12): p. 3494-505.

82. Koskivirta, I., et al., Mice with tissue inhibitor of metalloproteinases 4 (Timp4)

deletion succumb to induced myocardial infarction but not to cardiac pressure

overload. J Biol Chem, 2010. 285(32): p. 24487-93.

83. English, J.L., et al., Individual Timp deficiencies differentially impact pro-MMP-2

activation. J Biol Chem, 2006. 281(15): p. 10337-46.

84. Toth, M., et al., Tissue inhibitor of metalloproteinase (TIMP)-2 acts

synergistically with synthetic matrix metalloproteinase (MMP) inhibitors but not

with TIMP-4 to enhance the (Membrane type 1)-MMP-dependent activation of

pro-MMP-2. J Biol Chem, 2000. 275(52): p. 41415-23.

85. Seo, D.W., et al., TIMP-2 mediated inhibition of angiogenesis: an MMP-

independent mechanism. Cell, 2003. 114(2): p. 171-80.

86. Weber, B.H., et al., Mutations in the tissue inhibitor of metalloproteinases-3

(TIMP3) in patients with Sorsby's fundus dystrophy. Nat Genet, 1994. 8(4): p.

352-6.

87. Weber, B.H., et al., Sorsby's fundus dystrophy is genetically linked to

chromosome 22q13-qter. Nat Genet, 1994. 7(2): p. 158-61.

88. Langton, K.P., et al., A novel tissue inhibitor of metalloproteinases-3 mutation

reveals a common molecular phenotype in Sorsby's fundus dystrophy. J Biol

Chem, 2000. 275(35): p. 27027-31.

89. Kaur, I., S. Rathi, and S. Chakrabarti, Variations in TIMP3 are associated with

age-related macular degeneration. Proc Natl Acad Sci U S A, 2010. 107(28): p.

E112-3.

90. Yu, Y., et al., Common Variants near FRK/COL10A1 and VEGFA are Associated

with Advanced Age-related Macular Degeneration. Hum Mol Genet, 2011.

91. Garofalo, M., et al., miR-221&222 regulate TRAIL resistance and enhance

tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell, 2009.

16(6): p. 498-509.

Page 201: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

181

92. Selaru, F.M., et al., MicroRNA-21 is overexpressed in human

cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor

of metalloproteinase 3. Hepatology, 2009. 49(5): p. 1595-601.

93. Wang, B., et al., TGFbeta-mediated upregulation of hepatic miR-181b promotes

hepatocarcinogenesis by targeting TIMP3. Oncogene, 2010. 29(12): p. 1787-97.

94. Radpour, R., et al., Hypermethylation of tumor suppressor genes involved in

critical regulatory pathways for developing a blood-based test in breast cancer.

PLoS ONE, 2011. 6(1): p. e16080.

95. Sahebjam, S., R. Khokha, and J.S. Mort, Increased collagen and aggrecan

degradation with age in the joints of Timp3(-/-) mice. Arthritis Rheum, 2007.

56(3): p. 905-9.

96. Liu, H., et al., TIMP3: a physiological regulator of adult myogenesis. J Cell Sci,

2010. 123(Pt 17): p. 2914-21.

97. Kassiri, Z., et al., Combination of tumor necrosis factor-alpha ablation and

matrix metalloproteinase inhibition prevents heart failure after pressure overload

in tissue inhibitor of metalloproteinase-3 knock-out mice. Circ Res, 2005. 97(4):

p. 380-90.

98. Murthy, A., et al., Ectodomain shedding of EGFR ligands and TNFR1 dictates

hepatocyte apoptosis during fulminant hepatitis in mice. J Clin Invest, 2010.

120(8): p. 2731-44.

99. Shackel, N.A., The balancing act of hepatocyte apoptosis. Hepatology, 2010.

52(6): p. 2231-3.

100. Koskivirta, I., et al., Tissue inhibitor of metalloproteinases 4 (TIMP4) is involved

in inflammatory processes of human cardiovascular pathology. Histochem Cell

Biol, 2006. 126(3): p. 335-42.

101. Schulze, C.J., et al., Imbalance between tissue inhibitor of metalloproteinase-4

and matrix metalloproteinases during acute myocardial [correction of

myoctardial] ischemia-reperfusion injury. Circulation, 2003. 107(19): p. 2487-92.

102. Felkin, L.E., et al., Expression of extracellular matrix genes during myocardial

recovery from heart failure after left ventricular assist device support. J Heart

Lung Transplant, 2009. 28(2): p. 117-22.

103. Godenschwege, T.A., et al., Inflated wings, tissue autolysis and early death in

tissue inhibitor of metalloproteinases mutants of Drosophila. Eur J Cell Biol,

2000. 79(7): p. 495-501.

104. Nagase, H., R. Visse, and G. Murphy, Structure and function of matrix

metalloproteinases and TIMPs. Cardiovasc Res, 2006. 69(3): p. 562-73.

105. Visse, R. and H. Nagase, Matrix metalloproteinases and tissue inhibitors of

metalloproteinases: structure, function, and biochemistry. Circ Res, 2003. 92(8):

p. 827-39.

106. Lopez-Otin, C. and L.M. Matrisian, Emerging roles of proteases in tumour

suppression. Nat Rev Cancer, 2007. 7(10): p. 800-8.

107. Kenny, H.A., et al., The initial steps of ovarian cancer cell metastasis are

mediated by MMP-2 cleavage of vitronectin and fibronectin. J Clin Invest, 2008.

118(4): p. 1367-79.

Page 202: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

182

108. Balbin, M., et al., Loss of collagenase-2 confers increased skin tumor

susceptibility to male mice. Nat Genet, 2003. 35(3): p. 252-7.

109. Quintero, P.A., et al., Matrix metalloproteinase-8 inactivates macrophage

inflammatory protein-1 alpha to reduce acute lung inflammation and injury in

mice. J Immunol, 2010. 184(3): p. 1575-88.

110. Hotary, K., et al., A cancer cell metalloprotease triad regulates the basement

membrane transmigration program. Genes Dev, 2006. 20(19): p. 2673-86.

111. Hotary, K.B., et al., Membrane type I matrix metalloproteinase usurps tumor

growth control imposed by the three-dimensional extracellular matrix. Cell, 2003.

114(1): p. 33-45.

112. Sabeh, F., et al., Tumor cell traffic through the extracellular matrix is controlled

by the membrane-anchored collagenase MT1-MMP. J Cell Biol, 2004. 167(4): p.

769-81.

113. Zhai, Y., et al., Expression of membrane type 1 matrix metalloproteinase is

associated with cervical carcinoma progression and invasion. Cancer Res, 2005.

65(15): p. 6543-50.

114. Barbolina, M.V. and M.S. Stack, Membrane type 1-matrix metalloproteinase:

Substrate diversity in pericellular proteolysis. Semin Cell Dev Biol, 2007.

115. Nagase, H. and M. Kashiwagi, Aggrecanases and cartilage matrix degradation.

Arthritis Res Ther, 2003. 5(2): p. 94-103.

116. Daub, H., et al., Signal characteristics of G protein-transactivated EGF receptor.

EMBO J, 1997. 16(23): p. 7032-44.

117. Fischer, O.M., et al., Oxidative and osmotic stress signaling in tumor cells is

mediated by ADAM proteases and heparin-binding epidermal growth factor. Mol

Cell Biol, 2004. 24(12): p. 5172-83.

118. Fischer, O.M., et al., EGFR signal transactivation in cancer cells. Biochem Soc

Trans, 2003. 31(Pt 6): p. 1203-8.

119. Rooke, J., et al., KUZ, a conserved metalloprotease-disintegrin protein with two

roles in Drosophila neurogenesis. Science, 1996. 273(5279): p. 1227-31.

120. Pan, D. and G.M. Rubin, Kuzbanian controls proteolytic processing of Notch and

mediates lateral inhibition during Drosophila and vertebrate neurogenesis. Cell,

1997. 90(2): p. 271-80.

121. Sotillos, S., F. Roch, and S. Campuzano, The metalloprotease-disintegrin

Kuzbanian participates in Notch activation during growth and patterning of

Drosophila imaginal discs. Development, 1997. 124(23): p. 4769-79.

122. Yan, Y., K. Shirakabe, and Z. Werb, The metalloprotease Kuzbanian (ADAM10)

mediates the transactivation of EGF receptor by G protein-coupled receptors. J

Cell Biol, 2002. 158(2): p. 221-6.

123. Gibb, D.R., et al., ADAM10 is essential for Notch2-dependent marginal zone B

cell development and CD23 cleavage in vivo. J Exp Med, 2010. 207(3): p. 623-35.

124. Gibb, D.R., et al., ADAM10 overexpression shifts lympho- and myelopoiesis by

dysregulating site 2/site 3 cleavage products of Notch. J Immunol, 2011. 186(7):

p. 4244-52.

Page 203: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

183

125. Glomski, K., et al., Deletion of Adam10 in endothelial cells leads to defects in

organ-specific vascular structures. Blood, 2011. 118(4): p. 1163-74.

126. Jorissen, E., et al., The disintegrin/metalloproteinase ADAM10 is essential for the

establishment of the brain cortex. J Neurosci, 2010. 30(14): p. 4833-44.

127. Tian, L., et al., ADAM10 is essential for proteolytic activation of Notch during

thymocyte development. Int Immunol, 2008. 20(9): p. 1181-7.

128. Weber, S., et al., The disintegrin/metalloproteinase Adam10 is essential for

epidermal integrity and Notch-mediated signaling. Development, 2011. 138(3): p.

495-505.

129. Chen, G. and D.V. Goeddel, TNF-R1 signaling: a beautiful pathway. Science,

2002. 296(5573): p. 1634-5.

130. Grivennikov, S.I., et al., Intracellular signals and events activated by cytokines of

the tumor necrosis factor superfamily: From simple paradigms to complex

mechanisms. Int Rev Cytol, 2006. 252: p. 129-61.

131. Black, R.A., et al., A metalloproteinase disintegrin that releases tumour-necrosis

factor-alpha from cells. Nature, 1997. 385(6618): p. 729-33.

132. Bell, J., et al., Role of ADAM17 in the ectodomain shedding of TNF-{alpha} and

its receptors by neutrophils and macrophages. J Leukoc Biol, 2007.

133. Blobel, C.P., ADAMs: key components in EGFR signalling and development. Nat

Rev Mol Cell Biol, 2005. 6(1): p. 32-43.

134. Gschwind, A., et al., TACE cleavage of proamphiregulin regulates GPCR-

induced proliferation and motility of cancer cells. EMBO J, 2003. 22(10): p.

2411-21.

135. Lee, D.C., et al., TACE/ADAM17 processing of EGFR ligands indicates a role as

a physiological convertase. Ann N Y Acad Sci, 2003. 995: p. 22-38.

136. Maretzky, T., et al., Migration of growth factor-stimulated epithelial and

endothelial cells depends on EGFR transactivation by ADAM17. Nat Commun,

2011. 2: p. 229.

137. Sahin, U., et al., Distinct roles for ADAM10 and ADAM17 in ectodomain

shedding of six EGFR ligands. J Cell Biol, 2004. 164(5): p. 769-79.

138. Bozkulak, E.C. and G. Weinmaster, Selective use of ADAM10 and ADAM17 in

activation of Notch1 signaling. Mol Cell Biol, 2009. 29(21): p. 5679-95.

139. Brou, C., et al., A novel proteolytic cleavage involved in Notch signaling: the role

of the disintegrin-metalloprotease TACE. Mol Cell, 2000. 5(2): p. 207-16.

140. Saftig, P. and K. Reiss, The "A Disintegrin And Metalloproteases" ADAM10 and

ADAM17: novel drug targets with therapeutic potential? Eur J Cell Biol, 2011.

90(6-7): p. 527-35.

141. Murate, T., et al., Erythroid potentiating activity of tissue inhibitor of

metalloproteinases on the differentiation of erythropoietin-responsive mouse

erythroleukemia cell line, ELM-I-1-3, is closely related to its cell growth

potentiating activity. Exp Hematol, 1993. 21(1): p. 169-76.

142. Stetler-Stevenson, W.G., N. Bersch, and D.W. Golde, Tissue inhibitor of

metalloproteinase-2 (TIMP-2) has erythroid-potentiating activity. FEBS Lett,

1992. 296(2): p. 231-4.

Page 204: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

184

143. Baker, A.H., et al., Divergent effects of tissue inhibitor of metalloproteinase-1, -2,

or -3 overexpression on rat vascular smooth muscle cell invasion, proliferation,

and death in vitro. TIMP-3 promotes apoptosis. J Clin Invest, 1998. 101(6): p.

1478-87.

144. Celiker, M.Y., et al., Inhibition of Wilms' tumor growth by intramuscular

administration of tissue inhibitor of metalloproteinases-4 plasmid DNA.

Oncogene, 2001. 20(32): p. 4337-43.

145. Hayakawa, T., et al., Tissue inhibitor of metalloproteinases from human bone

marrow stromal cell line KM 102 has erythroid-potentiating activity, suggesting

its possibly bifunctional role in the hematopoietic microenvironment. FEBS Lett,

1990. 268(1): p. 125-8.

146. Hoegy, S.E., et al., Tissue inhibitor of metalloproteinases-2 (TIMP-2) suppresses

TKR-growth factor signaling independent of metalloproteinase inhibition. J Biol

Chem, 2001. 276(5): p. 3203-14.

147. van der Laan, W.H., et al., Cartilage degradation and invasion by rheumatoid

synovial fibroblasts is inhibited by gene transfer of TIMP-1 and TIMP-3. Gene

Ther, 2003. 10(3): p. 234-42.

148. Petitfrere, E., et al., Involvement of the p38 mitogen-activated protein kinase

pathway in tissue inhibitor of metalloproteinases-1-induced erythroid

differentiation. FEBS Lett, 2000. 485(2-3): p. 117-21.

149. Lizarraga, F., V. Maldonado, and J. Melendez-Zajgla, Tissue inhibitor of

metalloproteinases-2 growth-stimulatory activity is mediated by nuclear factor-

kappa B in A549 lung epithelial cells. Int J Biochem Cell Biol, 2004. 36(8): p.

1655-63.

150. Murphy, F.R., et al., Inhibition of apoptosis of activated hepatic stellate cells by

tissue inhibitor of metalloproteinase-1 is mediated via effects on matrix

metalloproteinase inhibition: implications for reversibility of liver fibrosis. J Biol

Chem, 2002. 277(13): p. 11069-76.

151. Lambert, E., et al., Tissue inhibitor of metalloproteinases-1 signalling pathway

leading to erythroid cell survival. Biochem J, 2003. 372(Pt 3): p. 767-74.

152. Boulday, G., et al., Exogenous tissue inhibitor of metalloproteinase-1 promotes

endothelial cell survival through activation of the phosphatidylinositol 3-

kinase/Akt pathway. Ann N Y Acad Sci, 2004. 1030: p. 28-36.

153. Liu, X.W., et al., Tissue inhibitor of metalloproteinase-1 protects human breast

epithelial cells from extrinsic cell death: a potential oncogenic activity of tissue

inhibitor of metalloproteinase-1. Cancer Res, 2005. 65(3): p. 898-906.

154. Lim, M.S., et al., Tissue inhibitor of metalloproteinase-2 induces apoptosis in

human T lymphocytes. Ann N Y Acad Sci, 1999. 878: p. 522-3.

155. Tran, P.L., et al., Gene therapy for hepatocellular carcinoma using non-viral

vectors composed of bis guanidinium-tren-cholesterol and plasmids encoding the

tissue inhibitors of metalloproteinases TIMP-2 and TIMP-3. Cancer Gene Ther,

2003. 10(6): p. 435-44.

Page 205: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

185

156. Bond, M., et al., Tissue inhibitor of metalloproteinase-3 induces a Fas-associated

death domain-dependent type II apoptotic pathway. J Biol Chem, 2002. 277(16):

p. 13787-95.

157. Ahonen, M., et al., Tissue inhibitor of metalloproteinases-3 induces apoptosis in

melanoma cells by stabilization of death receptors. Oncogene, 2003. 22(14): p.

2121-34.

158. Drynda, A., et al., Gene transfer of tissue inhibitor of metalloproteinases-3

reverses the inhibitory effects of TNF-alpha on Fas-induced apoptosis in

rheumatoid arthritis synovial fibroblasts. J Immunol, 2005. 174(10): p. 6524-31.

159. Jiang, Y., et al., Stimulation of mammary tumorigenesis by systemic tissue

inhibitor of matrix metalloproteinase 4 gene delivery. Cancer Res, 2001. 61(6): p.

2365-70.

160. Tummalapalli, C.M., B.J. Heath, and S.C. Tyagi, Tissue inhibitor of

metalloproteinase-4 instigates apoptosis in transformed cardiac fibroblasts. J

Cell Biochem, 2001. 80(4): p. 512-21.

161. Porter, J.F., S. Shen, and D.T. Denhardt, Tissue inhibitor of metalloproteinase-1

stimulates proliferation of human cancer cells by inhibiting a metalloproteinase.

Br J Cancer, 2004. 90(2): p. 463-70.

162. Lovelock, J.D., et al., Heterogeneous effects of tissue inhibitors of matrix

metalloproteinases on cardiac fibroblasts. Am J Physiol Heart Circ Physiol,

2005. 288(2): p. H461-8.

163. Smith, M.R., et al., TIMP-3 induces cell death by stabilizing TNF-alpha receptors

on the surface of human colon carcinoma cells. Cytokine, 1997. 9(10): p. 770-80.

164. Bai, Y.X., et al., Clinicopathologic significance of BAG1 and TIMP3 expression

in colon carcinoma. World J Gastroenterol, 2007. 13(28): p. 3883-5.

165. Han, X., et al., Expression of TIMP-3 gene by construction of a eukaryotic cell

expression vector and its role in reduction of metastasis in a human breast cancer

cell line. Cell Mol Immunol, 2004. 1(4): p. 308-10.

166. House, M.G., et al., Aberrant hypermethylation of tumor suppressor genes in

pancreatic endocrine neoplasms. Ann Surg, 2003. 238(3): p. 423-31; discussion

431-2.

167. Karan, D., et al., Expression of ADAMs (a disintegrin and metalloproteases) and

TIMP-3 (tissue inhibitor of metalloproteinase-3) in human prostatic

adenocarcinomas. Int J Oncol, 2003. 23(5): p. 1365-71.

168. Kim, Y.H., et al., CpG island methylation of genes accumulates during the

adenoma progression step of the multistep pathogenesis of colorectal cancer.

Genes Chromosomes Cancer, 2006. 45(8): p. 781-9.

169. Lui, E.L., et al., DNA hypermethylation of TIMP3 gene in invasive breast ductal

carcinoma. Biomed Pharmacother, 2005. 59 Suppl 2: p. S363-5.

170. Riddick, A.C., et al., Identification of degradome components associated with

prostate cancer progression by expression analysis of human prostatic tissues. Br

J Cancer, 2005. 92(12): p. 2171-80.

Page 206: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

186

171. van der Velden, P.A., et al., Expression profiling reveals that methylation of

TIMP3 is involved in uveal melanoma development. Int J Cancer, 2003. 106(4): p.

472-9.

172. Widschwendter, A., et al., DNA methylation in serum and tumors of cervical

cancer patients. Clin Cancer Res, 2004. 10(2): p. 565-71.

173. Johnson, J.L., et al., Suppression of atherosclerotic plaque progression and

instability by tissue inhibitor of metalloproteinase-2: involvement of macrophage

migration and apoptosis. Circulation, 2006. 113(20): p. 2435-44.

174. Ahonen, M., A.H. Baker, and V.M. Kahari, Adenovirus-mediated gene delivery of

tissue inhibitor of metalloproteinases-3 inhibits invasion and induces apoptosis in

melanoma cells. Cancer Res, 1998. 58(11): p. 2310-5.

175. Baker, A.H., et al., Inhibition of invasion and induction of apoptotic cell death of

cancer cell lines by overexpression of TIMP-3. Br J Cancer, 1999. 79(9-10): p.

1347-55.

176. Garton, K.J., P.J. Gough, and E.W. Raines, Emerging roles for ectodomain

shedding in the regulation of inflammatory responses. J Leukoc Biol, 2006. 79(6):

p. 1105-16.

177. Reiss, K., A. Ludwig, and P. Saftig, Breaking up the tie: disintegrin-like

metalloproteinases as regulators of cell migration in inflammation and invasion.

Pharmacol Ther, 2006. 111(3): p. 985-1006.

178. Smalley, D.M. and K. Ley, L-selectin: mechanisms and physiological significance

of ectodomain cleavage. J Cell Mol Med, 2005. 9(2): p. 255-66.

179. McQuibban, G.A., et al., Inflammation dampened by gelatinase A cleavage of

monocyte chemoattractant protein-3. Science, 2000. 289(5482): p. 1202-6.

180. Deftos, M.L., et al., Notch1 signaling promotes the maturation of CD4 and CD8

SP thymocytes. Immunity, 2000. 13(1): p. 73-84.

181. Manilay, J.O., et al., Impairment of thymocyte development by dominant-negative

Kuzbanian (ADAM-10) is rescued by the Notch ligand, delta-1. J Immunol, 2005.

174(11): p. 6732-41.

182. Hayday, A.C. and D.J. Pennington, Key factors in the organized chaos of early T

cell development. Nat Immunol, 2007. 8(2): p. 137-44.

183. Takahama, Y., Journey through the thymus: stromal guides for T-cell

development and selection. Nat Rev Immunol, 2006. 6(2): p. 127-35.

184. Proost, P., S. Struyf, and J. Van Damme, Natural post-translational modifications

of chemokines. Biochem Soc Trans, 2006. 34(Pt 6): p. 997-1001.

185. Van den Steen, P.E., et al., Carboxyterminal cleavage of the chemokines MIG and

IP-10 by gelatinase B and neutrophil collagenase. Biochem Biophys Res

Commun, 2003. 310(3): p. 889-96.

186. Van Den Steen, P.E., et al., Gelatinase B/MMP-9 and neutrophil

collagenase/MMP-8 process the chemokines human GCP-2/CXCL6, ENA-

78/CXCL5 and mouse GCP-2/LIX and modulate their physiological activities. Eur

J Biochem, 2003. 270(18): p. 3739-49.

Page 207: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

187

187. Gough, P.J., et al., A disintegrin and metalloproteinase 10-mediated cleavage and

shedding regulates the cell surface expression of CXC chemokine ligand 16. J

Immunol, 2004. 172(6): p. 3678-85.

188. Hundhausen, C., et al., The disintegrin-like metalloproteinase ADAM10 is

involved in constitutive cleavage of CX3CL1 (fractalkine) and regulates CX3CL1-

mediated cell-cell adhesion. Blood, 2003. 102(4): p. 1186-95.

189. Tsou, C.L., C.A. Haskell, and I.F. Charo, Tumor necrosis factor-alpha-converting

enzyme mediates the inducible cleavage of fractalkine. J Biol Chem, 2001.

276(48): p. 44622-6.

190. Schlondorff, J. and C.P. Blobel, Metalloprotease-disintegrins: modular proteins

capable of promoting cell-cell interactions and triggering signals by protein-

ectodomain shedding. J Cell Sci, 1999. 112 ( Pt 21): p. 3603-17.

191. Li, X. and H. Fan, Loss of ectodomain shedding due to mutations in the

metalloprotease and cysteine-rich/disintegrin domains of the tumor necrosis

factor-alpha converting enzyme (TACE). J Biol Chem, 2004. 279(26): p. 27365-

75.

192. Xia, M., et al., Shedding of Fas ectodomain that affects apoptosis of hepatocytes

occurring in regenerative liver. J Gastroenterol, 2002. 37(12): p. 1042-7.

193. Lichtenthaler, S.F., Ectodomain shedding of the amyloid precursor protein:

cellular control mechanisms and novel modifiers. Neurodegener Dis, 2006. 3(4-

5): p. 262-9.

194. Peiretti, F., et al., Proteasome inhibition activates the transport and the

ectodomain shedding of TNF-alpha receptors in human endothelial cells. J Cell

Sci, 2005. 118(Pt 5): p. 1061-70.

195. De Strooper, B., Nicastrin: gatekeeper of the gamma-secretase complex. Cell,

2005. 122(3): p. 318-20.

196. Kramer, H., RIPping notch apart: a new role for endocytosis in signal

transduction? Sci STKE, 2000. 2000(29): p. PE1.

197. Hartmann, D., et al., The disintegrin/metalloprotease ADAM 10 is essential for

Notch signalling but not for alpha-secretase activity in fibroblasts. Hum Mol

Genet, 2002. 11(21): p. 2615-24.

198. Six, E., et al., The Notch ligand Delta1 is sequentially cleaved by an ADAM

protease and gamma-secretase. Proc Natl Acad Sci U S A, 2003. 100(13): p.

7638-43.

199. Maillard, I., T. Fang, and W.S. Pear, Regulation of lymphoid development,

differentiation, and function by the Notch pathway. Annu Rev Immunol, 2005. 23:

p. 945-74.

200. Tan, J.B., et al., Requirement for Notch1 signals at sequential early stages of

intrathymic T cell development. Nat Immunol, 2005. 6(7): p. 671-9.

201. Li, N., et al., Non-cell autonomous expression of TNF-alpha-converting enzyme

ADAM17 is required for normal lymphocyte development. J Immunol, 2007.

178(7): p. 4214-21.

202. Cohn, M., et al., Reflections on the clonal-selection theory. Nat Rev Immunol,

2007. 7(10): p. 823-30.

Page 208: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

188

203. Li, N., et al., Metalloproteases regulate T-cell proliferation and effector function

via LAG-3. EMBO J, 2007. 26(2): p. 494-504.

204. Ethell, D.W., R. Kinloch, and D.R. Green, Metalloproteinase shedding of Fas

ligand regulates beta-amyloid neurotoxicity. Curr Biol, 2002. 12(18): p. 1595-

600.

205. Kirkin, V., et al., The Fas ligand intracellular domain is released by ADAM10

and SPPL2a cleavage in T-cells. Cell Death Differ, 2007. 14(9): p. 1678-87.

206. Schulte, M., et al., ADAM10 regulates FasL cell surface expression and

modulates FasL-induced cytotoxicity and activation-induced cell death. Cell

Death Differ, 2007. 14(5): p. 1040-9.

207. Ludwig, A. and C. Weber, Transmembrane chemokines: versatile 'special agents'

in vascular inflammation. Thromb Haemost, 2007. 97(5): p. 694-703.

208. Li, Y., et al., ADAM17 deficiency by mature neutrophils has differential effects on

L-selectin shedding. Blood, 2006. 108(7): p. 2275-9.

209. Faveeuw, C., G. Preece, and A. Ager, Transendothelial migration of lymphocytes

across high endothelial venules into lymph nodes is affected by

metalloproteinases. Blood, 2001. 98(3): p. 688-95.

210. Chen, J.R., et al., Transendothelial migration of lymphocytes in chronic

lymphocytic leukaemia is impaired and involved down-regulation of both L-

selectin and CD23. Br J Haematol, 1999. 105(1): p. 181-9.

211. Gomez-Gaviro, M., et al., Expression and regulation of the metalloproteinase

ADAM-8 during human neutrophil pathophysiological activation and its catalytic

activity on L-selectin shedding. J Immunol, 2007. 178(12): p. 8053-63.

212. Tsakadze, N.L., et al., Tumor necrosis factor-alpha-converting enzyme

(TACE/ADAM-17) mediates the ectodomain cleavage of intercellular adhesion

molecule-1 (ICAM-1). J Biol Chem, 2006. 281(6): p. 3157-64.

213. Garton, K.J., et al., Stimulated shedding of vascular cell adhesion molecule 1

(VCAM-1) is mediated by tumor necrosis factor-alpha-converting enzyme (ADAM

17). J Biol Chem, 2003. 278(39): p. 37459-64.

214. Singh, R.J., et al., Cytokine stimulated vascular cell adhesion molecule-1 (VCAM-

1) ectodomain release is regulated by TIMP-3. Cardiovasc Res, 2005. 67(1): p.

39-49.

215. Van Lint, P., et al., Resistance of collagenase-2 (matrix metalloproteinase-8)-

deficient mice to TNF-induced lethal hepatitis. J Immunol, 2005. 175(11): p.

7642-9.

216. Renckens, R., et al., Matrix metalloproteinase-9 deficiency impairs host defense

against abdominal sepsis. J Immunol, 2006. 176(6): p. 3735-41.

217. Dejana, E., The transcellular railway: insights into leukocyte diapedesis. Nat Cell

Biol, 2006. 8(2): p. 105-7.

218. Weiss, S.J., et al., Oxidative autoactivation of latent collagenase by human

neutrophils. Science, 1985. 227(4688): p. 747-9.

219. Gueders, M.M., et al., Matrix metalloproteinase-8 deficiency promotes

granulocytic allergen-induced airway inflammation. J Immunol, 2005. 175(4): p.

2589-97.

Page 209: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

189

220. Gutierrez-Fernandez, A., et al., Increased inflammation delays wound healing in

mice deficient in collagenase-2 (MMP-8). Faseb J, 2007.

221. Reddy, P., et al., Functional analysis of the domain structure of tumor necrosis

factor-alpha converting enzyme. J Biol Chem, 2000. 275(19): p. 14608-14.

222. Gomez, M.I., et al., Bacterial induction of TNF-alpha converting enzyme

expression and IL-6 receptor alpha shedding regulates airway inflammatory

signaling. J Immunol, 2005. 175(3): p. 1930-6.

223. Kinet, J.P., The high-affinity IgE receptor (Fc epsilon RI): from physiology to

pathology. Annu Rev Immunol, 1999. 17: p. 931-72.

224. Kijimoto-Ochiai, S., CD23 (the low-affinity IgE receptor) as a C-type lectin: a

multidomain and multifunctional molecule. Cell Mol Life Sci, 2002. 59(4): p.

648-64.

225. Armant, M., et al., Regulation of cytokine production by soluble CD23:

costimulation of interferon gamma secretion and triggering of tumor necrosis

factor alpha release. J Exp Med, 1994. 180(3): p. 1005-11.

226. Ribbens, C., et al., Increased synovial fluid levels of soluble CD23 are associated

with an erosive status in rheumatoid arthritis (RA). Clin Exp Immunol, 2000.

120(1): p. 194-9.

227. Weskamp, G., et al., ADAM10 is a principal 'sheddase' of the low-affinity

immunoglobulin E receptor CD23. Nat Immunol, 2006. 7(12): p. 1293-8.

228. Lemieux, G.A., et al., The low affinity IgE receptor (CD23) is cleaved by the

metalloproteinase ADAM10. J Biol Chem, 2007. 282(20): p. 14836-44.

229. Carey, B.W., D.Y. Kim, and D.M. Kovacs, Presenilin/gamma-secretase and

alpha-secretase-like peptidases cleave human MHC Class I proteins. Biochem J,

2007. 401(1): p. 121-7.

230. Gonzalez, P.A., et al., Modulation of immunological synapse by membrane-bound

and soluble ligands. Cytokine Growth Factor Rev, 2007. 18(1-2): p. 19-31.

231. Contin, C., et al., Potential role of soluble CD40 in the humoral immune response

impairment of uraemic patients. Immunology, 2003. 110(1): p. 131-40.

232. Contin, C., et al., Membrane-anchored CD40 is processed by the tumor necrosis

factor-alpha-converting enzyme. Implications for CD40 signaling. J Biol Chem,

2003. 278(35): p. 32801-9.

233. Logue, E.C., et al., ICOS-induced B7h shedding on B cells is inhibited by TLR7/8

and TLR9. J Immunol, 2006. 177(4): p. 2356-64.

234. Ebert, E.C., Interleukin-12 up-regulates perforin- and Fas-mediated lymphokine-

activated killer activity by intestinal intraepithelial lymphocytes. Clin Exp

Immunol, 2004. 138(2): p. 259-65.

235. Groh, V., et al., Fas-ligand-mediated paracrine T cell regulation by the receptor

NKG2D in tumor immunity. Nat Immunol, 2006. 7(7): p. 755-62.

236. Kaiser, B.K., et al., Disulphide-isomerase-enabled shedding of tumour-associated

NKG2D ligands. Nature, 2007. 447(7143): p. 482-6.

237. Kaufmann, T., et al., Fatal hepatitis mediated by tumor necrosis factor TNFalpha

requires caspase-8 and involves the BH3-only proteins Bid and Bim. Immunity,

2009. 30(1): p. 56-66.

Page 210: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

190

238. Das, M., et al., Induction of hepatitis by JNK-mediated expression of TNF-alpha.

Cell, 2009. 136(2): p. 249-60.

239. Grivennikov, S.I., et al., Distinct and nonredundant in vivo functions of TNF

produced by t cells and macrophages/neutrophils: protective and deleterious

effects. Immunity, 2005. 22(1): p. 93-104.

240. Aggarwal, B.B., Signalling pathways of the TNF superfamily: a double-edged

sword. Nat Rev Immunol, 2003. 3(9): p. 745-56.

241. Chen, X., et al., Bid-independent mitochondrial activation in tumor necrosis

factor alpha-induced apoptosis and liver injury. Mol Cell Biol, 2007. 27(2): p.

541-53.

242. Hughes, P.D., et al., Apoptosis regulators Fas and Bim cooperate in shutdown of

chronic immune responses and prevention of autoimmunity. Immunity, 2008.

28(2): p. 197-205.

243. Corazza, N., et al., TRAIL receptor-mediated JNK activation and Bim

phosphorylation critically regulate Fas-mediated liver damage and lethality. J

Clin Invest, 2006. 116(9): p. 2493-9.

244. Miettinen, P.J., et al., Epithelial immaturity and multiorgan failure in mice

lacking epidermal growth factor receptor. Nature, 1995. 376(6538): p. 337-41.

245. Berasain, C., et al., Novel role for amphiregulin in protection from liver injury. J

Biol Chem, 2005. 280(19): p. 19012-20.

246. Eberle, A., et al., Fluorescence resonance energy transfer analysis of

proapoptotic CD95-EGF receptor interactions in Huh7 cells. Hepatology, 2005.

41(2): p. 315-26.

247. Khai, N.C., et al., In vivo hepatic HB-EGF gene transduction inhibits Fas-

induced liver injury and induces liver regeneration in mice: a comparative study

to HGF. J Hepatol, 2006. 44(6): p. 1046-54.

248. Musallam, L., et al., EGF mediates protection against Fas-induced apoptosis by

depleting and oxidizing intracellular GSH stocks. J Cell Physiol, 2004. 198(1): p.

62-72.

249. Reinehr, R., F. Schliess, and D. Haussinger, Hyperosmolarity and CD95L trigger

CD95/EGF receptor association and tyrosine phosphorylation of CD95 as

prerequisites for CD95 membrane trafficking and DISC formation. FASEB J,

2003. 17(6): p. 731-3.

250. Wisniewska, M., et al., Structural determinants of the ADAM inhibition by TIMP-

3: crystal structure of the TACE-N-TIMP-3 complex. J Mol Biol, 2008. 381(5): p.

1307-19.

251. Tsuji, H., et al., Tumor necrosis factor receptor p55 is essential for intrahepatic

granuloma formation and hepatocellular apoptosis in a murine model of

bacterium-induced fulminant hepatitis. Infect Immun, 1997. 65(5): p. 1892-8.

252. O'Grady, J.G., S.W. Schalm, and R. Williams, Acute liver failure: redefining the

syndromes. Lancet, 1993. 342(8866): p. 273-5.

253. Menghini, R., et al., Tissue inhibitor of metalloproteinase 3 deficiency causes

hepatic steatosis and adipose tissue inflammation in mice. Gastroenterology,

2009. 136(2): p. 663-72 e4.

Page 211: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

191

254. Chang, L., et al., The E3 ubiquitin ligase itch couples JNK activation to

TNFalpha-induced cell death by inducing c-FLIP(L) turnover. Cell, 2006. 124(3):

p. 601-13.

255. Kamata, H., et al., Reactive oxygen species promote TNFalpha-induced death and

sustained JNK activation by inhibiting MAP kinase phosphatases. Cell, 2005.

120(5): p. 649-61.

256. Morel, C., et al., Mcl-1 integrates the opposing actions of signaling pathways that

mediate survival and apoptosis. Mol Cell Biol, 2009. 29(14): p. 3845-52.

257. Javelaud, D. and F. Besancon, NF-kappa B activation results in rapid inactivation

of JNK in TNF alpha-treated Ewing sarcoma cells: a mechanism for the anti-

apoptotic effect of NF-kappa B. Oncogene, 2001. 20(32): p. 4365-72.

258. Liu, J. and A. Lin, Role of JNK activation in apoptosis: a double-edged sword.

Cell Res, 2005. 15(1): p. 36-42.

259. Wicovsky, A., et al., Sustained JNK activation in response to tumor necrosis

factor is mediated by caspases in a cell type-specific manner. J Biol Chem, 2007.

282(4): p. 2174-83.

260. Luo, J.L., et al., Inhibition of NF-kappaB in cancer cells converts inflammation-

induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor

regression. Cancer Cell, 2004. 6(3): p. 297-305.

261. Vallabhapurapu, S. and M. Karin, Regulation and function of NF-kappaB

transcription factors in the immune system. Annu Rev Immunol, 2009. 27: p. 693-

733.

262. Maurer, U., et al., Glycogen synthase kinase-3 regulates mitochondrial outer

membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell,

2006. 21(6): p. 749-60.

263. Black, R.A., TIMP3 checks inflammation. Nat Genet, 2004. 36(9): p. 934-5.

264. Lee, M.H., et al., Full-length and N-TIMP-3 display equal inhibitory activities

toward TNF-alpha convertase. Biochem Biophys Res Commun, 2001. 280(3): p.

945-50.

265. Natarajan, A., B. Wagner, and M. Sibilia, The EGF receptor is required for

efficient liver regeneration. Proc Natl Acad Sci U S A, 2007. 104(43): p. 17081-6.

266. Alfano, D., I. Iaccarino, and M.P. Stoppelli, Urokinase signaling through its

receptor protects against anoikis by increasing BCL-xL expression levels. J Biol

Chem, 2006. 281(26): p. 17758-67.

267. Faber, A.C., et al., Differential induction of apoptosis in HER2 and EGFR

addicted cancers following PI3K inhibition. Proc Natl Acad Sci U S A, 2009.

106(46): p. 19503-8.

268. Leu, C.M., C. Chang, and C. Hu, Epidermal growth factor (EGF) suppresses

staurosporine-induced apoptosis by inducing mcl-1 via the mitogen-activated

protein kinase pathway. Oncogene, 2000. 19(13): p. 1665-75.

269. Henderson, N.C., et al., Critical role of c-jun (NH2) terminal kinase in

paracetamol- induced acute liver failure. Gut, 2007. 56(7): p. 982-90.

270. Imaeda, A.B., et al., Acetaminophen-induced hepatotoxicity in mice is dependent

on Tlr9 and the Nalp3 inflammasome. J Clin Invest, 2009. 119(2): p. 305-14.

Page 212: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

192

271. Lee, W.M., Acetaminophen toxicity: changing perceptions on a social/medical

issue. Hepatology, 2007. 46(4): p. 966-70.

272. Malhi, H., G.J. Gores, and J.J. Lemasters, Apoptosis and necrosis in the liver: a

tale of two deaths? Hepatology, 2006. 43(2 Suppl 1): p. S31-44.

273. Tagami, A., H. Ohnishi, and R.D. Hughes, Increased serum soluble Fas in

patients with acute liver failure due to paracetamol overdose.

Hepatogastroenterology, 2003. 50(51): p. 742-5.

274. Tinel, M., et al., Subliminal Fas stimulation increases the hepatotoxicity of

acetaminophen and bromobenzene in mice. Hepatology, 2004. 39(3): p. 655-66.

275. Zhang, H., et al., Reduction of liver Fas expression by an antisense

oligonucleotide protects mice from fulminant hepatitis. Nat Biotechnol, 2000.

18(8): p. 862-7.

276. Beraza, N., et al., Hepatocyte-specific NEMO deletion promotes NK/NKT cell-

and TRAIL-dependent liver damage. J Exp Med, 2009.

277. Luedde, T., et al., Deletion of NEMO/IKKgamma in liver parenchymal cells

causes steatohepatitis and hepatocellular carcinoma. Cancer Cell, 2007. 11(2): p.

119-32.

278. Sun, B. and M. Karin, NF-kappaB signaling, liver disease and hepatoprotective

agents. Oncogene, 2008. 27(48): p. 6228-44.

279. Geisler, F., et al., Genetic inactivation of RelA/p65 sensitizes adult mouse

hepatocytes to TNF-induced apoptosis in vivo and in vitro. Gastroenterology,

2007. 132(7): p. 2489-503.

280. Maeda, S., et al., IKKbeta couples hepatocyte death to cytokine-driven

compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell,

2005. 121(7): p. 977-90.

281. Inta, I., et al., Bim and Noxa are candidates to mediate the deleterious effect of the

NF-kappa B subunit RelA in cerebral ischemia. J Neurosci, 2006. 26(50): p.

12896-903.

282. Ghosh, S., M.J. May, and E.B. Kopp, NF-kappa B and Rel proteins:

evolutionarily conserved mediators of immune responses. Annu Rev Immunol,

1998. 16: p. 225-60.

283. Song, L., et al., IKKbeta programs to turn on the GADD45alpha-MKK4-JNK

apoptotic cascade specifically via p50 NF-kappaB in arsenite response. J Cell

Biol, 2006. 175(4): p. 607-17.

284. Gong, Y., et al., Induction of BIM is essential for apoptosis triggered by EGFR

kinase inhibitors in mutant EGFR-dependent lung adenocarcinomas. PLoS

medicine, 2007. 4(10): p. e294-e294.

285. Reginato, M.J., et al., Integrins and EGFR coordinately regulate the pro-

apoptotic protein Bim to prevent anoikis. Nat Cell Biol, 2003. 5(8): p. 733-40.

286. Horiuchi, K., et al., Cutting edge: TNF-alpha-converting enzyme

(TACE/ADAM17) inactivation in mouse myeloid cells prevents lethality from

endotoxin shock. J Immunol, 2007. 179(5): p. 2686-9.

287. Kim, A.H., et al., Akt phosphorylates and negatively regulates apoptosis signal-

regulating kinase 1. Mol Cell Biol, 2001. 21(3): p. 893-901.

Page 213: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

193

288. Berasain, C., et al., Amphiregulin: an early trigger of liver regeneration in mice.

Gastroenterology, 2005. 128(2): p. 424-32.

289. Wetzel, M., et al., Tissue inhibitor of metalloproteinases-3 facilitates Fas-

mediated neuronal cell death following mild ischemia. Cell Death Differ, 2008.

15(1): p. 143-51.

290. Junttila, M.R., S.P. Li, and J. Westermarck, Phosphatase-mediated crosstalk

between MAPK signaling pathways in the regulation of cell survival. FASEB J,

2008. 22(4): p. 954-65.

291. Xia, Z., et al., Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis.

Science, 1995. 270(5240): p. 1326-31.

292. Michalopoulos, G.K., Liver regeneration after partial hepatectomy: critical

analysis of mechanistic dilemmas. Am J Pathol, 2010. 176(1): p. 2-13.

293. Crispe, I.N., The liver as a lymphoid organ. Annu Rev Immunol, 2009. 27: p.

147-63.

294. Ichiki, Y., et al., T cell immunity in autoimmune hepatitis. Autoimmun Rev, 2005.

4(5): p. 315-21.

295. Croft, M., The role of TNF superfamily members in T-cell function and diseases.

Nat Rev Immunol, 2009. 9(4): p. 271-85.

296. Medzhitov, R., Origin and physiological roles of inflammation. Nature, 2008.

454(7203): p. 428-35.

297. Bonder, C.S., et al., Rules of recruitment for Th1 and Th2 lymphocytes in

inflamed liver: a role for alpha-4 integrin and vascular adhesion protein-1.

Immunity, 2005. 23(2): p. 153-63.

298. Erhardt, A., et al., IL-10, regulatory T cells, and Kupffer cells mediate tolerance

in concanavalin A-induced liver injury in mice. Hepatology, 2007. 45(2): p. 475-

85.

299. Jiang, W., et al., TLR-9 activation aggravates concanavalin A-induced hepatitis

via promoting accumulation and activation of liver CD4+ NKT cells. J Immunol,

2009. 182(6): p. 3768-74.

300. Murphy, G., A. Murthy, and R. Khokha, Clipping, shedding and RIPping keep

immunity on cue. Trends Immunol, 2008. 29(2): p. 75-82.

301. Consolo, M., et al., Matrix metalloproteinases and their inhibitors as markers of

inflammation and fibrosis in chronic liver disease (Review). Int J Mol Med, 2009.

24(2): p. 143-52.

302. Murthy, A., et al., Ectodomain shedding of EGFR ligands and TNFR1 dictates

hepatocyte apoptosis during fulminant hepatitis in mice. J Clin Invest, 2010.

303. Boisvert, J., et al., Liver-infiltrating lymphocytes in end-stage hepatitis C virus:

subsets, activation status, and chemokine receptor phenotypes. J Hepatol, 2003.

38(1): p. 67-75.

304. Tiegs, G., J. Hentschel, and A. Wendel, A T cell-dependent experimental liver

injury in mice inducible by concanavalin A. J Clin Invest, 1992. 90(1): p. 196-

203.

305. Toyabe, S., et al., Requirement of IL-4 and liver NK1+ T cells for concanavalin

A-induced hepatic injury in mice. J Immunol, 1997. 159(3): p. 1537-42.

Page 214: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

194

306. Jin, X., et al., Paradoxical effects of short- and long-term interleukin-6 exposure

on liver injury and repair. Hepatology, 2006. 43(3): p. 474-84.

307. Klein, C., et al., The IL-6-gp130-STAT3 pathway in hepatocytes triggers liver

protection in T cell-mediated liver injury. J Clin Invest, 2005. 115(4): p. 860-9.

308. Duran, A., et al., Crosstalk between PKCzeta and the IL4/Stat6 pathway during T-

cell-mediated hepatitis. EMBO J, 2004. 23(23): p. 4595-605.

309. Deng, Z.B., et al., Immature myeloid cells induced by a high-fat diet contribute to

liver inflammation. Hepatology, 2009. 50(5): p. 1412-20.

310. Zenewicz, L.A., et al., Interleukin-22 but not interleukin-17 provides protection to

hepatocytes during acute liver inflammation. Immunity, 2007. 27(4): p. 647-59.

311. Crispe, I.N., Hepatic T cells and liver tolerance. Nat Rev Immunol, 2003. 3(1): p.

51-62.

312. Echeverria, I., et al., Enhanced T cell responses against hepatitis C virus by ex

vivo targeting of adenoviral particles to dendritic cells. Hepatology, 2011. 54(1):

p. 28-37.

313. Palucka, K., J. Banchereau, and I. Mellman, Designing vaccines based on biology

of human dendritic cell subsets. Immunity, 2010. 33(4): p. 464-78.

314. Das, A., et al., Functional skewing of the global CD8 T cell population in chronic

hepatitis B virus infection. J Exp Med, 2008. 205(9): p. 2111-24.

315. Raziorrouh, B., et al., Inhibitory Molecules that Regulate Expansion and

Restoration of HCV-Specific CD4+ T Cells in Patients with Chronic Infection.

Gastroenterology, 2011.

316. Egen, J.G., et al., Intravital imaging reveals limited antigen presentation and T

cell effector function in mycobacterial granulomas. Immunity, 2011. 34(5): p.

807-19.

317. Heydtmann, M. and D.H. Adams, Chemokines in the immunopathogenesis of

hepatitis C infection. Hepatology, 2009. 49(2): p. 676-88.

318. Sahin, H., C. Trautwein, and H.E. Wasmuth, Functional role of chemokines in

liver disease models. Nat Rev Gastroenterol Hepatol, 2010. 7(12): p. 682-90.

319. Casrouge, A., et al., Evidence for an antagonist form of the chemokine CXCL10 in

patients chronically infected with HCV. J Clin Invest, 2011. 121(1): p. 308-17.

320. Leifeld, L., et al., Early up-regulation of chemokine expression in fulminant

hepatic failure. J Pathol, 2003. 199(3): p. 335-44.

321. Amend, B., et al., Induction of autoimmunity by expansion of autoreactive

CD4+CD62Llow cells in vivo. J Immunol, 2006. 177(7): p. 4384-90.

322. Cauwels, A., et al., Caspase inhibition causes hyperacute tumor necrosis factor-

induced shock via oxidative stress and phospholipase A2. Nat Immunol, 2003.

4(4): p. 387-93.

323. Hasselblatt, P., et al., Hepatocyte survival in acute hepatitis is due to c-Jun/AP-1-

dependent expression of inducible nitric oxide synthase. Proc Natl Acad Sci U S

A, 2007. 104(43): p. 17105-10.

324. Eksteen, B., et al., Immune-mediated liver injury. Semin Liver Dis, 2007. 27(4):

p. 351-66.

Page 215: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

195

325. Dienes, H.P. and U. Drebber, Pathology of immune-mediated liver injury. Dig

Dis, 2010. 28(1): p. 57-62.

326. Bieber, T., Atopic dermatitis. N Engl J Med, 2008. 358(14): p. 1483-94.

327. Swamy, M., et al., Epithelial decision makers: in search of the 'epimmunome'. Nat

Immunol, 2010. 11(8): p. 656-65.

328. Demehri, S., et al., Skin-derived TSLP triggers progression from epidermal-

barrier defects to asthma. PLoS Biol, 2009. 7(5): p. e1000067.

329. Ziegler, S.F. and D. Artis, Sensing the outside world: TSLP regulates barrier

immunity. Nat Immunol, 2010. 11(4): p. 289-93.

330. Dumortier, A., et al., Atopic dermatitis-like disease and associated lethal

myeloproliferative disorder arise from loss of Notch signaling in the murine skin.

PLoS One, 2010. 5(2): p. e9258.

331. Meixner, A., et al., Epidermal JunB represses G-CSF transcription and affects

haematopoiesis and bone formation. Nat Cell Biol, 2008. 10(8): p. 1003-11.

332. Blanpain, C., et al., Canonical notch signaling functions as a commitment switch

in the epidermal lineage. Genes Dev, 2006. 20(21): p. 3022-35.

333. Demehri, S., et al., Notch-deficient skin induces a lethal systemic B-

lymphoproliferative disorder by secreting TSLP, a sentinel for epidermal

integrity. PLoS Biol, 2008. 6(5): p. e123.

334. Fort, M.M., et al., IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated

pathologies in vivo. Immunity, 2001. 15(6): p. 985-95.

335. Ogura, H., et al., Interleukin-17 promotes autoimmunity by triggering a positive-

feedback loop via interleukin-6 induction. Immunity, 2008. 29(4): p. 628-36.

336. Muller, M., et al., CXCR3 signaling reduces the severity of experimental

autoimmune encephalomyelitis by controlling the parenchymal distribution of

effector and regulatory T cells in the central nervous system. J Immunol, 2007.

179(5): p. 2774-86.

337. Fang, D., et al., Dysregulation of T lymphocyte function in itchy mice: a role for

Itch in TH2 differentiation. Nat Immunol, 2002. 3(3): p. 281-7.

338. Campbell, P.J. and A.R. Green, The myeloproliferative disorders. N Engl J Med,

2006. 355(23): p. 2452-66.

339. Guinea-Viniegra, J., et al., TNFalpha shedding and epidermal inflammation are

controlled by Jun proteins. Genes Dev, 2009. 23(22): p. 2663-74.

340. Leung, D.Y., et al., New insights into atopic dermatitis. J Clin Invest, 2004.

113(5): p. 651-7.

341. Pasparakis, M., et al., TNF-mediated inflammatory skin disease in mice with

epidermis-specific deletion of IKK2. Nature, 2002. 417(6891): p. 861-6.

342. Artavanis-Tsakonas, S., M.D. Rand, and R.J. Lake, Notch signaling: cell fate

control and signal integration in development. Science, 1999. 284(5415): p. 770-

6.

343. Guttman-Yassky, E., et al., Broad defects in epidermal cornification in atopic

dermatitis identified through genomic analysis. J Allergy Clin Immunol, 2009.

124(6): p. 1235-1244 e58.

Page 216: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

196

344. Nair, R.P., et al., Genome-wide scan reveals association of psoriasis with IL-23

and NF-kappaB pathways. Nat Genet, 2009. 41(2): p. 199-204.

345. Olsson, M., et al., Increased expression of aquaporin 3 in atopic eczema. Allergy,

2006. 61(9): p. 1132-7.

346. Reischl, J., et al., Increased expression of Wnt5a in psoriatic plaques. J Invest

Dermatol, 2007. 127(1): p. 163-9.

347. Saenz, S.A., B.C. Taylor, and D. Artis, Welcome to the neighborhood: epithelial

cell-derived cytokines license innate and adaptive immune responses at mucosal

sites. Immunol Rev, 2008. 226: p. 172-90.

348. Eferl, R. and E.F. Wagner, AP-1: a double-edged sword in tumorigenesis. Nat

Rev Cancer, 2003. 3(11): p. 859-68.

349. Chu, J., et al., Repression of activator protein-1-mediated transcriptional

activation by the Notch-1 intracellular domain. J Biol Chem, 2002. 277(9): p.

7587-97.

350. Fukushima, H., et al., The association of Notch2 and NF-kappaB accelerates

RANKL-induced osteoclastogenesis. Mol Cell Biol, 2008. 28(20): p. 6402-12.

351. Talora, C., et al., Specific down-modulation of Notch1 signaling in cervical

cancer cells is required for sustained HPV-E6/E7 expression and late steps of

malignant transformation. Genes Dev, 2002. 16(17): p. 2252-63.

352. Harada, M., et al., TSLP Promoter Polymorphisms are Associated with

Susceptibility to Bronchial Asthma. Am J Respir Cell Mol Biol, 2010.

353. Zenz, R., et al., Psoriasis-like skin disease and arthritis caused by inducible

epidermal deletion of Jun proteins. Nature, 2005. 437(7057): p. 369-75.

354. Esteve, P., et al., SFRPs act as negative modulators of ADAM10 to regulate

retinal neurogenesis. Nat Neurosci, 2011. 14(5): p. 562-9.

355. van Tetering, G., et al., Metalloprotease ADAM10 is required for Notch1 site 2

cleavage. J Biol Chem, 2009. 284(45): p. 31018-27.

356. Nicolas, M., et al., Notch1 functions as a tumor suppressor in mouse skin. Nat

Genet, 2003. 33(3): p. 416-21.

357. Williams, S.E., et al., Asymmetric cell divisions promote Notch-dependent

epidermal differentiation. Nature, 2011. 470(7334): p. 353-8.

358. Chu, J. and E.H. Bresnick, Evidence that C promoter-binding factor 1 binding is

required for Notch-1-mediated repression of activator protein-1. J Biol Chem,

2004. 279(13): p. 12337-45.

359. Pastore, S., et al., Dysregulated activation of activator protein 1 in keratinocytes

of atopic dermatitis patients with enhanced expression of

granulocyte/macrophage-colony stimulating factor. J Invest Dermatol, 2000.

115(6): p. 1134-43.

360. Song, Z., et al., Polymorphic nucleotides within the human IL-4 promoter that

mediate overexpression of the gene. J Immunol, 1996. 156(2): p. 424-9.

361. Islam, S.A., et al., Mouse CCL8, a CCR8 agonist, promotes atopic dermatitis by

recruiting IL-5+ T(H)2 cells. Nat Immunol, 2011. 12(2): p. 167-77.

362. Biton, M., et al., Epithelial microRNAs regulate gut mucosal immunity via

epithelium-T cell crosstalk. Nat Immunol, 2011. 12(3): p. 239-46.

Page 217: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

197

363. Pasparakis, M., Regulation of tissue homeostasis by NF-kappaB signalling:

implications for inflammatory diseases. Nat Rev Immunol, 2009. 9(11): p. 778-

88.

364. Benjamini, Y.a.H., Y., Controlling the false discovery rate: a practical and

powerful approach to multiple testing. Journal of the Royal Statistical Society,

Series B (Methodological), 1995. 57(1): p. 289-300.

365. Pavlidis, P. and W.S. Noble, Matrix2png: a utility for visualizing matrix data.

Bioinformatics, 2003. 19(2): p. 295-6.

366. Eisen, M.B., et al., Cluster analysis and display of genome-wide expression

patterns. Proc Natl Acad Sci U S A, 1998. 95(25): p. 14863-8.

367. Saldanha, A.J., Java Treeview--extensible visualization of microarray data.

Bioinformatics, 2004. 20(17): p. 3246-8.

368. Ehebauer, M., P. Hayward, and A. Martinez-Arias, Notch signaling pathway. Sci

STKE, 2006. 2006(364): p. cm7.

369. Harada, H., et al., Localization of putative stem cells in dental epithelium and

their association with Notch and FGF signaling. J Cell Biol, 1999. 147(1): p.

105-20.

370. Jeffries, S. and A.J. Capobianco, Neoplastic transformation by Notch requires

nuclear localization. Mol Cell Biol, 2000. 20(11): p. 3928-41.

371. Kopan, R., Notch: a membrane-bound transcription factor. J Cell Sci, 2002.

115(Pt 6): p. 1095-7.

372. Liu, Z.J., et al., Regulation of Notch1 and Dll4 by vascular endothelial growth

factor in arterial endothelial cells: implications for modulating arteriogenesis

and angiogenesis. Mol Cell Biol, 2003. 23(1): p. 14-25.

373. Ohishi, K., et al., Monocytes express high amounts of Notch and undergo cytokine

specific apoptosis following interaction with the Notch ligand, Delta-1. Blood,

2000. 95(9): p. 2847-54.

374. Wu, L., et al., Identification of a family of mastermind-like transcriptional

coactivators for mammalian notch receptors. Mol Cell Biol, 2002. 22(21): p.

7688-700.

375. Lichti, U., J. Anders, and S.H. Yuspa, Isolation and short-term culture of primary

keratinocytes, hair follicle populations and dermal cells from newborn mice and

keratinocytes from adult mice for in vitro analysis and for grafting to

immunodeficient mice. Nat Protoc, 2008. 3(5): p. 799-810.

376. Jin, H., et al., Animal models of atopic dermatitis. J Invest Dermatol, 2009.

129(1): p. 31-40.

377. Spergel, J.M., et al., Epicutaneous sensitization with protein antigen induces

localized allergic dermatitis and hyperresponsiveness to methacholine after single

exposure to aerosolized antigen in mice. J Clin Invest, 1998. 101(8): p. 1614-22.

378. Chu, C.C., P. Di Meglio, and F.O. Nestle, Harnessing dendritic cells in

inflammatory skin diseases. Semin Immunol, 2011. 23(1): p. 28-41.

379. Johnson, T.R., et al., Primary human mDC1, mDC2, and pDC dendritic cells are

differentially infected and activated by respiratory syncytial virus. PLoS ONE,

2011. 6(1): p. e16458.

Page 218: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

198

380. Baratelli, F.E., et al., Prostaglandin E2-dependent enhancement of tissue

inhibitors of metalloproteinases-1 production limits dendritic cell migration

through extracellular matrix. J Immunol, 2004. 173(9): p. 5458-66.

381. Ratzinger, G., et al., Matrix metalloproteinases 9 and 2 are necessary for the

migration of Langerhans cells and dermal dendritic cells from human and murine

skin. J Immunol, 2002. 168(9): p. 4361-71.

382. MacDonald, T.T. and A. Di Sabatino, The immunologic basis for gastrointestinal

food allergy. Curr Opin Gastroenterol, 2009. 25(6): p. 521-6.

383. Spergel, J.M., From atopic dermatitis to asthma: the atopic march. Ann Allergy

Asthma Immunol, 2010. 105(2): p. 99-106; quiz 107-9, 117.

384. Brandl, K., et al., MyD88 signaling in nonhematopoietic cells protects mice

against induced colitis by regulating specific EGF receptor ligands. Proc Natl

Acad Sci U S A, 2010. 107(46): p. 19967-72.

385. Chalaris, A., et al., Critical role of the disintegrin metalloprotease ADAM17 for

intestinal inflammation and regeneration in mice. J Exp Med, 2010.

386. Gregorio, J., et al., Plasmacytoid dendritic cells sense skin injury and promote

wound healing through type I interferons. J Exp Med, 2010. 207(13): p. 2921-30.

387. Guiducci, C., et al., Autoimmune skin inflammation is dependent on plasmacytoid

dendritic cell activation by nucleic acids via TLR7 and TLR9. J Exp Med, 2010.

207(13): p. 2931-42.

388. Wenzel, J. and T. Tuting, An IFN-associated cytotoxic cellular immune response

against viral, self-, or tumor antigens is a common pathogenetic feature in

"interface dermatitis". J Invest Dermatol, 2008. 128(10): p. 2392-402.

389. Wenzel, J., et al., The expression pattern of interferon-inducible proteins reflects

the characteristic histological distribution of infiltrating immune cells in different

cutaneous lupus erythematosus subsets. Br J Dermatol, 2007. 157(4): p. 752-7.

390. Gao, L., L. Chao, and J. Chao, A novel signaling pathway of tissue kallikrein in

promoting keratinocyte migration: activation of proteinase-activated receptor 1

and epidermal growth factor receptor. Exp Cell Res, 2010. 316(3): p. 376-89.

391. Miyoshi, H., et al., Beneficial effects of tissue inhibitor of metalloproteinases-2

(TIMP-2) on chronic dermatitis. J Dermatol, 2005. 32(5): p. 346-53.

392. Griffith, A.V., et al., Spatial mapping of thymic stromal microenvironments

reveals unique features influencing T lymphoid differentiation. Immunity, 2009.

31(6): p. 999-1009.

393. Petrie, H.T. and J.C. Zuniga-Pflucker, Zoned out: functional mapping of stromal

signaling microenvironments in the thymus. Annu Rev Immunol, 2007. 25: p.

649-79.

394. Schmitt, T.M. and J.C. Zuniga-Pflucker, Induction of T cell development from

hematopoietic progenitor cells by delta-like-1 in vitro. Immunity, 2002. 17(6): p.

749-56.

395. Ramsdell, F., J.C. Zuniga-Pflucker, and Y. Takahama, In vitro systems for the

study of T cell development: fetal thymus organ culture and OP9-DL1 cell

coculture. Curr Protoc Immunol, 2006. Chapter 3: p. Unit 3 18.

Page 219: Regulation of metalloproteinase-dependent ectodomain ......Regulation of metalloproteinase-dependent ectodomain shedding in cytokine biology and inflammation. Aditya Murthy, Doctor

199

396. Anderson, G. and E.J. Jenkinson, Lymphostromal interactions in thymic

development and function. Nat Rev Immunol, 2001. 1(1): p. 31-40.

397. Yuan, J.S., et al., Functions of notch signaling in the immune system: consensus

and controversies. Annu Rev Immunol, 2010. 28: p. 343-65.

398. Ciofani, M. and J.C. Zuniga-Pflucker, Notch promotes survival of pre-T cells at

the beta-selection checkpoint by regulating cellular metabolism. Nat Immunol,

2005. 6(9): p. 881-8.

399. Guidos, C.J., Synergy between the pre-T cell receptor and Notch: cementing the

alphabeta lineage choice. J Exp Med, 2006. 203(10): p. 2233-7.

400. Maillard, I., et al., The requirement for Notch signaling at the beta-selection

checkpoint in vivo is absolute and independent of the pre-T cell receptor. J Exp

Med, 2006. 203(10): p. 2239-45.

401. Barbee, S.D., et al., Skint-1 is a highly specific, unique selecting component for

epidermal T cells. Proc Natl Acad Sci U S A, 2011. 108(8): p. 3330-5.

402. Hayday, A.C., Gammadelta T cells and the lymphoid stress-surveillance

response. Immunity, 2009. 31(2): p. 184-96.

403. Taghon, T., et al., Developmental and molecular characterization of emerging

beta- and gammadelta-selected pre-T cells in the adult mouse thymus. Immunity,

2006. 24(1): p. 53-64.

404. Duncan, A.W., et al., Integration of Notch and Wnt signaling in hematopoietic

stem cell maintenance. Nat Immunol, 2005. 6(3): p. 314-22.