the interplay of ige and its high affinity receptor fceri

132
Research Collection Doctoral Thesis The interplay of IgE and its high affinity receptor FcεRI Author(s): Pochanke, Veronika Publication Date: 2005 Permanent Link: https://doi.org/10.3929/ethz-a-005062847 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library

Upload: vutu

Post on 31-Jan-2017

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Interplay of IgE and its High Affinity Receptor FceRI

Research Collection

Doctoral Thesis

The interplay of IgE and its high affinity receptor FcεRI

Author(s): Pochanke, Veronika

Publication Date: 2005

Permanent Link: https://doi.org/10.3929/ethz-a-005062847

Rights / License: In Copyright - Non-Commercial Use Permitted

This page was generated automatically upon download from the ETH Zurich Research Collection. For moreinformation please consult the Terms of use.

ETH Library

Page 2: The Interplay of IgE and its High Affinity Receptor FceRI

Diss. ETHNo. 16149

The Interplay of IgE and its High Affinity Receptor FceRI

A dissertation submitted to the

SWISS FEDERAL INSTITUTE OF TECHNOLOGY ZURICH

for the degree of

Doctor of Natural Sciences

presented by

VERONIKA POCHANKE

Dipl. Biol., Université de Lausanne

born 07.09.1975

citizen of Wil (SG), Switzerland

accepted on the recommendation of

Prof. Dr. Hans Hengartner, examiner

Prof. Dr. Rolf M. Zinkernagel, co-examiner

Dr. Kathy D. McCoy, co-examiner

2005

Page 3: The Interplay of IgE and its High Affinity Receptor FceRI
Page 4: The Interplay of IgE and its High Affinity Receptor FceRI

Table of Contents 1

Table of Contents

Summary 3

Zusammenfassung 5

Abbreviations 7

1 Introduction 11

1.1 Immunoglobulin E (IgE) 11

1.2 The high affinity IgE receptor, FcsRI 19

1.3 Allergy 24

1.4 Central Question 30

2 Results Part 1 31

Induction of IgE and allergic-type responses in fur mite-infested mice 31

2.1 Abstract 31

2.2 Introduction 32

2.3 Materials and Methods 34

2.4 Results 36

2.5 Discussion 46

3 Results Part II 51

Generation and characterization of hmFcsRIocß Tg mice 51

3.1 Abstract 51

3.2 Introduction 52

3.3 Materials and Methods 54

3.4 Results 60

3.5 Discussion 76

4 Results Part III 79

Generation of a muFcsRIa-huIgGiFc fusion protein 79

4.1 Abstract 79

4.2 Introduction 80

4.3 Materials and Methods 82

4.4 Results 86

Page 5: The Interplay of IgE and its High Affinity Receptor FceRI

2 Table of Contents

4.5 Discussion 97

5 Discussion 101

5.1 The functions of IgE 101

5.2 The epidemic of allergy 102

5.3 Mechanisms of IgE-mediated immune responses 103

5.4 Remaining questions to be answered 103

5.5 Questions addressed in this thesis 106

5.6 Concluding remarks 108

6 References 109

Acknowledgements 127

Curriculum Vitae 129

Page 6: The Interplay of IgE and its High Affinity Receptor FceRI

Summary 3

Summary

Allergic diseases, such as eczema, rhinitis, asthma, and food allergies, have greatly

increased in the past two decades, mainly in developed countries. Immunoglobulin E (IgE)

is known to play a crucial role in the development of allergies, however, the physiological

function of IgE has most likely evolved to provide host defense mechanisms against

parasite infections. IgE-mediated immune responses involve similar mechanisms during

both allergic reactions and parasite clearance. Exposure to antigens leads to crosslinking of

antigen-specific IgE molecules bound to high affinity IgE receptors, FcsRI, expressed

predominantly on mast cells and basophils. Activation of FcsRI mediates the release of

allergic mediators, which are responsible for symptoms of allergy. Understanding the

mechanisms regulating IgE-dependent responses is therefore essential to provide effective

therapy against allergic disease and to alleviate damage caused by parasite infections.

The goal of the present thesis was to investigate the initial requirements for IgE production

and to elucidate the role of FcsRI expression on different cell subsets. Animal models that

closely resemble the pathology of human allergic conditions and allow measurement of

physiologic parameters are required to efficiently address these questions. We therefore

established and characterized a novel allergy model based on infestation of mice with fur

mites. The advantage of this model lies in the usage of natural murine ectoparasites

allowing physiological exposure to antigens, which strongly resembles the human atopic

sensitization. Fur mites were found to induce high levels of serum IgE in a T cell- and In¬

dependent manner. Moreover, our results strongly suggest that mite antigen entry occurs

via the skin of infested mice leading to T cell activation and B cell isotype switch to IgE in

skin-draining lymph nodes. Furthermore, mite-induced mast cell degranulation in the skin

was detected implying that some of the IgE produced in response to mite infestation was

antigen-specific.

In the second part of this thesis we investigated the functions of FcsRI. An intriguing

difference in the structure and cellular distribution of the human and murine high affinity

IgE receptor was previously found. While murine FcsRI has an obligatory tetrameric

structure and is only expressed on mast cells and basophils, the human receptor can be

additionally expressed as a trimer found on eosinophils, dendritic cells, Langerhans' cells,

monocytes, macrophages, and platelets. To investigate the role of FcsRI expression on

Page 7: The Interplay of IgE and its High Affinity Receptor FceRI

4 Summary

these effector and antigen presenting cells, we generated a murine transgenic system

(hmFcsRIaß Tg mice) that mirrors the human distribution pattern of FcsRI. Although

mRNA expression of the high affinity IgE receptor was successfully demonstrated in the

targeted cells of the hmFcsRIaß Tg mice, FcsRI protein expression could not be found.

Further investigation of the model is therefore required to determine the reason for the

absence of detectable FcsRI protein expression and to evaluate how it may be overcome.

Lastly, to provide an additional tool for the investigation of FcsRI we made an attempt to

generate a specific antibody against the a chain of the murine receptor. Such an antibody is

crucial for specific and efficient analysis of the expression of the murine FcsRI, which was

important for the study of FcsRI transgenic mice. To achieve the production of an anti-

muFcsRIa antibody, a fusion protein composed of the extracellular domain of the murine

FcsRIa and the human IgGi Fc portion was generated and used to immunize FcsRIa-

deficient mice. However, as the first series of immunizations did not result in the synthesis

of anti-muFcsRIa antibodies and because a commercial anti-mouse FcsRIa antibody

became available during the completion of this project, the investigation was interrupted.

Nevertheless, the successfully generated and purified muFcsRIa-huIgGiFc fusion protein

is available in large amounts and can be used in future if required.

In conclusion, the regulation of IgE production and the effector functions induced by IgE

interactions with FcsRI expressed on different cell types are still not fully understood.

Analysis of novel animal models, such as those presented in this thesis, may provide

answers to some of the still open questions and eventually lead to successful therapy of

allergy and parasite-induced disease.

Page 8: The Interplay of IgE and its High Affinity Receptor FceRI

Zusammenfassung 5

Zusammenfassung

Allergische Reaktionen, wie z.B. Ekzem, Heuschnupfen, Asthma und

Nahrungsmittelallergien, sind in den letzten 20 Jahren vor allem in entwickelten Ländern

drastisch gestiegen. Die Entstehung von Allergien ist zum grossen Teil vom

Immunoglobulin E (IgE) abhängig, die physiologische Funktion von IgE dient jedoch zum

Schutz gegen parasitäre Infektionen. Die Immunantwort, die durch IgE ausgelöst wird,

umfasst ähnliche Mechanismen bei allergischen Reaktionen wie auch bei der Abwehr

gegen Parasiten. Kontakt mit Antigenen führt zu einer Kreuzvernetzung von

antigenspezifisehen IgE Molekülen, die an Hochaffinitätsrezeptoren für IgE, FcsRI,

gebunden sind. FcsRI ist vorwiegend auf Mastzellen und Basophilen exprimiert und seine

Aktivierung führt zur Freisetzung von pro-inflammatorischen Substanzen, z.B. Histamin,

welche die klassischen Merkmale von Allergie hervorrufen. Mechanismen der von IgE

ausgelösten Immunantworten zu verstehen, ist daher essentiell um wirksame Mittel gegen

Allergien und parasitäre Krankheiten zu finden.

Das Ziel der vorliegenden Dissertation war es, die Voraussetzungen für die IgE Produktion

zu untersuchen, und die Rolle von FcsRI Expression auf diversen Zelltypen aufzuklären.

Tiermodelle, welche die Pathologie von menschlichen allergischen Erkrankungen

nachahmen und die es ermöglichen, physiologische Parameter zu messen, sind erforderlich

um diese Fragen effizient anzugehen. Wir haben ein neues Allergiemodel etabliert und

charakterisiert, das auf dem Befall von Mäusen durch Fellmilben basiert. Der Vorteil dieses

Models liegt darin, dass natürliche Mausektoparasiten den Kontakt mit Antigenen in

physiologischer Art und Weise ermöglichen, was wiederum der atopischen Sensibilisierung

bei Menschen ähnlich ist. Wir haben gezeigt, dass Milbenantigene durch die Haut von

befallenen Mäusen aufgenommen und über die Lymphe in die dränierenden Lymphknoten

transportiert werden. Dort führt das Antigen zur Aktivierung von T Zellen, die zur

Produktion von IL-4 stimuliert werden. Diese Th2 Antwort führt zum IgE-Klassenwechsel

in B Zellen und schliesslich zu stark erhöhten IgE Serumwerten. Zudem haben wir

milbenabhängige Mastzelldegranulation in der Haut beobachtet, was darauf hindeutet, dass

ein Teil an IgE, das als Antwort auf den Milbenbefall produziert wurde, Antigen-spezifisch

sein muss.

Page 9: The Interplay of IgE and its High Affinity Receptor FceRI

6 Zusammenfassung

Im zweiten Teil dieser Arbeit haben wir die Funktionen des FcsRI Rezeptors untersucht.

Ein überraschender Unterschied in der Struktur und in der zellulären Verteilung zwischen

dem IgE Rezeptor von Mensch und Maus wurde bereits vor einigen Jahren erkannt.

Während FcsRI in der Maus eine ausschliesslich tetramerische Struktur aufweist und nur

auf Mastzellen und Basophilen exprimiert wird, kann FcsRI im Menschen eine trimerische

Form annehmen, die auch auf eosinophilen Granulozyten, dendritischen Zellen,

Langerhans-Zellen, Monozyten, Makrophagen und Thrombozyten exprimiert werden kann.

Um die Funktion von FcsRI Expression auf diesen Zellen zu untersuchen, haben wir ein

transgenes System (hmFcsRIaß Tg Maus) hergestellt, das die menschliche FcsRI

Expression widerspiegelt. Obwohl die mRNA Expression des IgE Rezeptors in den Zellen

der hmFcsRIaß transgenen Mäuse erfolgreich gezeigt wurde, konnten wir keine FcsRI

Protein Expression nachweisen. Das Modell erfordert deshalb weiterer Analyse, um den

Grund für die fehlende FcsRI Protein Expression zu identifizieren, und zu beurteilen wie

diese zu überwinden sei.

Darüber hinaus haben wir versucht, einen spezifischen Antikörper gegen die a Kette des

FcsRI der Maus herzustellen. Ein solcher Antikörper ist erforderlich für eine spezifische

und effiziente Untersuchung der FcsRI Expression, was für die Studie der FcsRI

transgenen Mäuse ausschlaggebend war. Um die Produktion von anti-muFcsRIa

Antikörpern zu erzielen, haben wir ein Fusionsprotein hergestellt, das aus der

extrazellulären Domäne der a Kette von FcsRI und dem Fc-Teil des menschlichen IgGi

Antikörpers bestand. Dieses Fusionsprotein wurde anschliessend zur Immunisierung von

FcsRIa""

Mäusen verwendet. Da aber die ersten Immunisierungsserien nicht zur Synthese

des erwünschten Antikörpers geführt hatten, und weil ein kommerzieller anti-muFcsRIa

Antikörper während der Durchführung dieses Projektes erhältlich wurde, haben wir die

Versuche eingestellt. Dennoch steht das erfolgreich generierte und isolierte Fusionsprotein

für allfällige weitere Studien zur Verfügung.

Schlussfolgernd ist festzuhalten, dass die Regulation der IgE Produktion und die Folgen der

Interaktionen zwischen IgE und FcsRI noch nicht gänzlich geklärt sind. Analyse neuer

Tiermodelle, wie z.B. solcher die in dieser Dissertation präsentiert wurden, könnten

Antworten auf einige der offenen Fragen liefern und schliesslich zur erfolgreichen Therapie

gegen Allergie und parasitäre Infektionen führen.

Page 10: The Interplay of IgE and its High Affinity Receptor FceRI

Abbreviations 7

Abbreviations

Ab Antibody

AD Atopic dermatitis

Ag Antigen

AID Activation-induced cytidine deaminase

Amp Ampicillin

APC Antigen presenting cell

BAL Bronchoalveolar lavage

BMMC Bone marrow-derived mast cell

bp Base pair

BSA Bovine serum albumin

BSS Balanced salt solution

CD Cluster of differentiation

CD40L CD40 ligand

cDNA Complementary DNA

CDR Complementarity-determining region

Cs IgE heavy chain constant region

CFA Complete Freund's adjuvant

CSR Class-switch recombination

DC Dendritic cell

DNA Deoxyribonucleic acid

DNase Deoxyribonuclease

ELISA Enzyme-linked immunosorbent assay

Eos Eosinophil

ER Endoplasmic reticulum

Fab Antigen-binding fragment

FACS Fluorescence activated cell sorting

Fe Crystallizable fragment

FCS Fetal calf serum

FcsRI High affinity IgE receptor

FcsRIa Alpha chain of the high affinity IgE receptor

Page 11: The Interplay of IgE and its High Affinity Receptor FceRI

8 Abbreviations

FcsRIß Beta chain of the high affinity IgE receptor

FcsRIy Gamma chain of the high affinity IgE receptor

FR Framework

GFP Green fluorescent protein

GLT Germline transcript

HBSS Hanks balanced salt solution

HEK Human embryonic kidney cells

Hyg Hygromycin

i.p. Intraperitoneal

i.v. Intravenous

ICAM Intercellular adhesion molecule

IFA Incomplete Freund's adjuvant

IFN Interferon

Ig Immunoglobulin

IL Interleukin

FMDM Iscove's modified Dulbecco's media

ITAM Immunoreceptor tyrosine-based activation motif

Ka Association constant

Kana Kanamycin

kb Kilobase

kDa Kilodalton

k0ff Dissociation rate

LC Langerhans' cell

LFA Lymphocyte function-associated antigen

LN Lymph node

mAb Monoclonal antibody

MACS Magnetic-assisted cell sorting

MAUD Mite-associated ulcerative dermatitis

MC Mast cell

MHC Major histocompatibility complex

MIP Macrophage inflammatory protein

mRNA Messenger RNA

Page 12: The Interplay of IgE and its High Affinity Receptor FceRI

Abbreviations 9

MO Macrophage

iV. brasiliensis Nippostrongylus brasiliensis

NF-kB Nuclear factor-KB

OD Optical density

OVA Ovalbumin

PBMC Peripheral blood mononucleocytes

PBS Phosphate buffered saline

PCR Polymerase chain reaction

PLC Phospholipase C

PMA Phorbol myristate acetate

PTK Protein tyrosine kinase

RNA Ribonucleic acid

rRNA Ribosomal RNA

RT Room temperature

RT-PCR Reverse transcription-PCR

S. mansoni Schistosoma mansoni

s.c. Subcutaneous

s.d. Standard deviation

SCF Stem cell factor

SEM Standard error of the mean

Spl Spleen

STAT Signal transducer and activator oft

T. spiralis Trichinella spiralis

TCR T cell receptor

Tg Transgenic

TGF Transforming growth factor

Th Helper T cell

TLR Toll-like receptor

TNF Tumor necrosis factor

WT Wild type

Page 13: The Interplay of IgE and its High Affinity Receptor FceRI

10 Abbreviations

Page 14: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 11

1 Introduction

1.1 Immunoglobulin E (IgE)

Physiological properties and structure of IgE

The first demonstration to show that a serum factor (originally named "reagin") was

responsible for the development of allergic reactions was performed in 1921 by Carl

Prausnitz and Heinz Küstner (Prausnitz, 1921). They injected serum from a fish-allergic

individual (Küstner himself) into the skin of a non-allergic person (Prausnitz) and observed

a typical weal and erythema reaction at the site of serum transfer after local administration

of fish extract. Only decades later, in 1967, was reagin recognized as a new

immunoglobulin class and was designated IgE due to the observed erythema reaction

(Ishizaka and Ishizaka, 1967). The physiological characteristics of IgE differ from the other

four known immunoglobulins, IgM, IgG, IgD, and IgA. First, IgE is the least abundant

antibody class in serum, with concentrations of about 100 ng/ml in healthy individuals,

compared with 10 mg/ml for IgG (Sutton and Gould, 1993). However, in atopic or

parasitized individuals IgE concentrations can increase by up to 1000-fold (Barbera et al.,

1977; Durmaz et al., 1998). Second, with a serum half-life of only about 2 days, IgE has the

shortest biological half-life of all the immunoglobulins. However, if IgE is cell surface-

bound, this half-life can be extended to 1-2 weeks (Tada et al., 1975). Similar to other

immunoglobulins, IgE consists of heavy (H) and light (L) chains with variable (V) and

constant (C) regions but it differs in that it is composed of s H-chains, which contain four C

domains (Csl-Cs4). Such distribution of C regions is also found in IgM while the H-chains

of IgG, IgD, and IgA are all composed of only three C domains (Gould et al., 2003) (Figure

1). The additional IgE domain (Cs2) replaces the hinge region found in the other antibody

classes and is a critical determinant of the physical properties of IgE (See paragraph "IgE-

FcsRI interactions"). Recently, the crystal structure of the IgE-Fc fragment has been solved

providing new insight into the conformation of IgE and the binding properties of IgE to its

high affinity receptor, FcsRI (Wan et al., 2002).

Page 15: The Interplay of IgE and its High Affinity Receptor FceRI

12 Introduction

B

s-s

B

H

m

a

m

O

u

H chains: a, 7, 8, e, |i

L chains: k. A,

Figure 1. Structures of immunoglobulins. Immunoglobulins are composed of two heavy (H) and

two light (L) chains. There are five types (isotypes) of H chains (a, y, S, e, and ]i) and two types of L

chains (k and X). The amino-terminal (NH3+) regions of each chain contain variable (V) domains

responsible for antigen binding, while the carboxy-terminal (COO) regions contain constant (C)

domains responsible for the biological activity of the antibodies. The a, y, and 5 H chains contain

three C domains (CH1-CH3) and a proline-rich hinge region (A), while the e and ]i H chains, which

lack the hinge region, contain four C domains (CH1-CH4) (B). Immunoglobulins can also be

subdivided into antigen-binding (Fab) and "crystallizable" fragments (Fc) by enzymatic digestion

above the disulfide bonds (S-S) that link the H chains.

Regulation of IgE

The very low levels of serum IgE of healthy individuals suggest that IgE synthesis may be

tightly regulated. This is thought to be important to prevent the potentially lethal

consequences of IgE-dependent immune responses. Thus, multiple factors are involved in

the induction of IgE synthesis.

One of the first steps leading to IgE production is specific binding of antigen to membrane-

bound B cell receptors, which subsequently ensures the specificity of the IgE produced. B

cells have the capacity to internalize, process and present antigenic peptides on MHC class

II molecules (Vercelli et al., 1989). Peptide-MHC class II complexes are recognized by

specific T cell receptors (TCRs) on CD4+ T cells. CD4+ T cells, also known as T helper

cells, are crucial for regulation of both cellular and humoral immune responses by secreting

cytokines and providing signals required for antibody production. Two subtypes of T helper

Page 16: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 13

cells (Thl and Th2) have been described based on their functional properties and the profile

of cytokines they produce (Abbas et al., 1996). Thl cells represent a major source of

interleukin (IL)-2, interferon (IFN)-y, tumor necrosis factor (TNF)-a, and other cytokines

important for cell-mediated immune responses. Thl responses antagonize IgE effector

functions, as IL-12 and IFN-y suppress IgE synthesis through direct effects on B cells

(Coffman and Carty, 1986). On the other hand, Th2 cells secrete IL-4, IL-5, IL-6, IL-10,

and IL-13, which are required for antibody responses, including IgE production (Aebischer

and Stadler, 1996). After antigen-specific interaction of TCRs with peptide-MHC class II

complexes, Th2 cells provide B cells with signals crucial for IgE synthesis. Such signals

involve cytokines secreted by Th2 cells as well as cell-to-cell interactions between Th2

cells and B cells (Figure 2). The most important cytokines for induction of IgE are IL-4 and

IL-13 (Finkelman et al., 1988; Minty et al., 1993; Punnonen et al., 1993). Binding of IL-4

and IL-13 to their receptors expressed on the surface of B cells leads to activation of the

transcription factor signal transducer and activator of transcription 6 (STAT6), which

provides the first signal for isotype switching to IgE (de Vries et al., 1993). Ligation of

CD40 (expressed on B cells) to its ligand CD40L (expressed on Th2 cells) activates the

transcription factor nuclear factor-KB (NF-kB), which induces the second signal leading to

IgE synthesis. NF-kB acts in synergy with IL-4-induced STAT6. On one hand, STAT6

together with NF-kB induce production of Cs germline transcripts (GLTs) by binding to

the Is promoter located upstream of the Cs genes (Iciek et al., 1997). Although Cs GLTs

lack the VDJ region and are "sterile", meaning that they are not translated into protein

because of stop codons in all reading frames, Cs GLT expression is a necessary step for the

subsequent class-switch recombination (CSR) to IgE (Geha et al., 2003). On the other hand,

synergy between STAT6 and NF-kB results in expression of activation-induced cytidine

deaminase (AID) (Chaudhuri et al., 2003). AID has been shown to play a crucial role in

CSR (Muramatsu et al., 2000) by a yet unresolved mechanism involving cytidine

deamination of either RNA (Muramatsu et al., 1999) or single-stranded DNA (Chaudhuri et

al., 2003). Thus, signals through both IL-4 and CD40 are required to induce Cs GLT

expression and to reach a threshold level of AID expression necessary for switching to IgE

(Messner et al., 1997). However, it has been reported that under some circumstances IgE

can be produced independently of T cells (Jabara et al., 1990), IL-4 (Grunewald et al.,

Page 17: The Interplay of IgE and its High Affinity Receptor FceRI

14 Introduction

2001; Morawetz et al., 1996), or CD40 (Litinskiy et al., 2002) implying that regulation of

IgE synthesis is a complex process involving additional molecules and signaling pathways.

Tceli

Antigen (~)

B cell

IL-13

Figure 2. Major T cell - B cell interactions regulating induction of IgE. Specific binding of

antigen by B cells via surface IgM (a) leads to processing and presentation of antigens by MHC

class II molecules and allows antigen-specific interactions with T cells via TCRs (b). Activated CD4+

Th2 cells express high levels of CD40L and produce the cytokines IL-4 and IL-13. Interaction of

CD40L with CD40 expressed by B cells (c) activates NF-kB (d), while binding of IL-4 and IL-13 to

their receptors (e) recruits STAT6 (f). Synergy between NF-kB and STAT6 leads to transcription of

Ce GLT and AID (g), both required for subsequent IgE synthesis (h).

Page 18: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 15

Effector functions of IgE

Type I immediate hypersensitivity

The immune system attempts to protect individuals from pathogens by inducing localized

inflammation. However, an inflammatory response can also have deleterious effects.

Inappropriate immune responses resulting in host damage can derive either from a reaction

against self-antigens (autoimmunity) or from an overreaction against environmental

antigens (hypersensitivity). Such an "overraction" was first described by Paul Portier and

Charles Richet in 1902 (Cohen and Zelaya-Quesada, 2002; Haas, 2001). These two French

scientists discovered that a second injection of fluids isolated from jellyfish induced a

violent reaction in dogs that showed no symptoms to an earlier injection. They called this

phenomenon "anaphylaxis" to mean the opposite of prophylaxis. Anaphylactic reactions

are cunently refened to as immediate or type I hypersensitivity. Hypersensitivity reactions

have been classified into four types by Gell and Coombs (Coombs RRA, 1963): type I, IgE-

mediated hypersensitivity; type II, IgG-mediated cytotoxic hypersensitivity; type III,

immune complex-mediated hypersensitivity; and type IV, cell-mediated hypersensitivity.

Type I hypersensitivity is induced by antigens that elicit secretion of specific IgE. High

affinity IgE receptors, FcsRI, highly expressed on mast cells and basophils, capture

secreted IgE (Wedemeyer et al., 2000). Such sensitized cells can be activated within

minutes following re-exposure to the same antigen (Figure 3). Signaling pathways induced

by antigen-dependent crosslinking of IgE bound to FcsRI trigger degranulation of the cell

and result in the release of preformed inflammatory mediators including histamine, neutral

proteases, and proteoglycans (Boyce, 2004). Additionally, activated mast cells and

basophils secrete newly synthesized lipid mediators such as prostaglandin D2, leukotriene

C4, and platelet-activating factor as well as numerous proinflammatory cytokines,

chemokines and growth factors, including TNF-a, IL-4, IL-13, and eotaxin (Prussin and

Metcalfe, 2003).

Clinical manifestations related to mediators released during IgE-induced degranulation

include enhanced local vascular permeability, increased cutaneous blood flow, erythema,

and other effects such as itching (Kay, 2001). Pathology induced by type I hypersensitivity

can range from moderate allergic reactions including hay fever and eczema, to life-

threatening conditions, such as systemic anaphylaxis and asthma.

Page 19: The Interplay of IgE and its High Affinity Receptor FceRI

16 Introduction

Figure 3. Activation and degranulation of mast cells. Soluble IgE binds to FceRI expressed on

mast cells. Crosslinking of receptor-bound IgE by specific antigen triggers the release of preformed

mediators (e.g. histamine) from the granules and induces synthesis of additional inflammatory

factors (e.g. lipids and cytokines).

Regulation ofmast cell survival andFcsRI expression

Recent reports have suggested that binding of monomeric IgE to FcsRI, i.e. in the absence

of antigen, can have immuno-regulatory functions. It has been demonstrated that

monomeric IgE can promote mouse mast cell proliferation induced by IL-3 or SCF in vitro

(Asai et al., 2001; Kalesnikoff et al., 2001; Kitaura et al., 2003). The mechanism of

monomeric IgE-mediated mast cell survival has been shown to involve anti-apoptotic

effects, however, its biological relevance remains to be determined. In addition, it has been

reported that IgE binding to FcsRI in the absence of specific antigen induces upregulation

of FcsRI surface expression on mast cells and basophils (Lantz et al., 1997; Yamaguchi et

al., 1997). The enhanced expression of FcsRI by IgE has been shown to be achieved by

receptor stabilization at the membrane as well as recruitment of a preformed pool of

receptors in the presence of continual basal protein synthesis (Borkowski et al., 2001). IgE

binding to FcsRI might also make the receptor resistant to degradation via conformational

changes induced in the a chain (Kubo et al., 2001). The biological relevance of FcsRI

upregulation induced by newly produced IgE may be an enhanced sensitivity to pathogens

due to reactivity to an increased number of antigens. This also implies that prevention of

Page 20: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 17

IgE-mediated FcsRI upregulation may be a useful target to diminish the impact of allergic

disorders.

Protection againstparasites

The first observation that parasites, particularly helminthes, induce strong primary and

secondary IgE responses was described by Ogilvie in 1964 (Ogilvie, 1964). Numerous

reports have since accumulated suggesting that the biological role of IgE-associated

immune responses is to provide host defense against parasites (Hagan, 1993; Janett and

Miller, 1982; Levy, 2004). However, different degrees of importance of IgE in parasite

clearance have been shown and the requirement of IgE for protective host immunity to

parasites has remained controversial. Investigation of Schistosoma mansoni (S. mansoni)

infection in humans revealed host defense mechanisms involving IgE-dependent killing of

larval schistosomes by platelets and macrophages (Capron et al., 1987). Moreover,

resistance to reinfection with S. mansoni was associated with high anti-schistosomular IgE

levels (Rihet et al., 1991). Convincing evidence that protection against reinfection is

associated with high levels of parasite-specific IgE was also provided by studies of

Schistosoma haematobium infection (Hagan et al., 1991). Additionally, a positive

conelation of parasite-specific IgE levels and protection in rodents was found for

Trichinella spiralis (T spiralis) (Ahmad et al., 1991; Bell, 1998; Dessein et al., 1981).

Interestingly, a powerful IgE response, with over 60% parasite-specific IgE, was found

within the intestinal lumen of T spiralis infected mice, while only 10% of serum IgE was

found to be specific (Negrao-Correa, 2001; Negrao-Conea et al., 1996). A recent study of

IgE-deficient mice infected with T spiralis revealed that IgE was required for both the

elimination of adult worms from the intestine and the killing of larvae present in the

skeletal muscle through mechanisms involving mast cell homeostasis and secretion of mast

cell protease-1 (Gurish et al., 2004).

However, other reports have speculated that IgE production in response to helminth

infection is merely the consequence of a strong Th2 response induced by the parasites but

plays no crucial role in parasite expulsion. In some cases this may indeed be true. For

example, it has been shown that mice lacking functional IgE receptors expel Trichuris

muris with normal kinetics (Betts and Else, 1999).

Page 21: The Interplay of IgE and its High Affinity Receptor FceRI

18 Introduction

Intriguingly, the presence of disproportionately high levels of total (non-parasite specific)

IgE has been observed in response to many parasitic helminthes (Dessaint et al., 1975;

Turner et al., 1979). The generation of such polyvalent IgE has been explained in two ways

(Pritchard, 1993). First, the presence of non-specific IgE might imply the existence of a

protective strategy used by the parasite whereby competition with specific IgE is induced.

Second, high amounts of unrelated IgE could possibly reduce the risk of anaphylaxis,

thereby protecting the host from potentially lethal consequences of hyperreactivity to

parasite antigens.

Taken together, IgE participation in the protective mechanisms involved in parasite

infections may strongly depend on the parasite species. Complex life cycles involving

different tissue localizations of helminthes, such as lung, stomach, or intestine, may induce

various mechanisms leading to protective immunity of the host. For example, it has been

proposed that mucosal penetration by the nematode may be required for induction of an IgE

response that could participate locally in worm elimination. Thus, parasite infections result

in multifactoral and redundant immune responses depending on the parasite species and

infection site. While a Th2-associated immune response is known to be a common feature

in immunity to most parasites, the exact effector elements, their regulation and modes of

action that are important for protection against each individual parasite, remain to be

determined.

Page 22: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 19

1.2 The high affinity IgE receptor, FcsRI

Structure and expression of FceRI

FcsRI is a multimeric cell surface receptor that exists in two isoforms. It can be expressed

either as a heterotetramer composed of one a, one ß, and two y chains (aßy2), or as a

heterotrimer lacking the ß chain (ay2) (Kinet, 1999) (Figure 4). The aßy2 form of FcsRI is

abundantly expressed on IgE-associated effector cells, mast cells and basophils, reaching

3xl05 molecules/cell (Liu et al., 2001). In humans, the trimeric form of FcsRI has been

demonstrated to be expressed on Langerhans' cells (Bieber et al., 1992; Wang et al., 1992),

monocytes (Maurer et al., 1994), eosinophils (Gounni et al., 1994), peripheral blood

dendritic cells (Maurer et al., 1996), and platelets (Joseph et al., 1997). Interestingly

however, mice lack expression of the ay2 form of FcsRI and therefore do not express the

receptor on cells other than mast cells and basophils. The a chain of the receptor (FcsRIa)

is a type I integral membrane protein with two Ig-like domains (Dl and D2) in the

extracellular (N-terminal) region, a single transmembrane domain, and a short cytoplasmic

tail (Garman et al., 1998). FcsRIa is heavily glycosylated due to seven N-linked

glycosylation sites involved in interactions between the a chain and the folding machinery

of the endoplasmic reticulum (Letourneur et al., 1995b). Crystal structure analysis of IgE-

Fc bound to FcsRIa revealed that the Cs3 domains of IgE-Fc bind two distinct sites located

in the D2 domain of FcsRIa (Garman et al., 2000). The ß subunit (FcsRIß) has four

transmembrane domains separating amino and carboxy terminal cytoplasmic tails

containing an immunoreceptor tyrosine-based activation motif (ITAM) near the carboxy

terminus (Kinet et al., 1988). Two major functions of FcsRIß are known. First, FcsRIß has

been shown to amplify signals produced by the y chain of the receptor, even though ß does

not appear to possess any signaling capacity through its own ITAM (Jouvin et al., 1994;

Lin et al., 1996). Second, it has been demonstrated that FcsRIß facilitates surface

expression of the receptor by promoting the processing and export of the a chain to the cell

surface and by enhancing the stability of the receptor complexes (Donnadieu et al., 2000).

The y chains (FcsRIy) form a disulfide-linked homodimer with short extracellular portions

and long cytoplasmic tails containing an ITAM on each subunit (Küster et al., 1990).

Several other receptors, including FcyRI, FcyRIIIA, FcaR, and the CD3 complex of the T

Page 23: The Interplay of IgE and its High Affinity Receptor FceRI

20 Introduction

cell receptor (TCR), share the ubiquitously expressed y subunit, which is crucial for

induction of signaling cascades through phosphorylation of the ITAMs (Daeron, 1997).

HUMAN+MOUSE; HUMAN:

mast cells Langerhans' cells

basophils monocytes

eosinophilsdendritic cells

platelets

Figure 4. Structure and cellular distribution of the high affinity IgE receptor, FcsRI. FceRI is

expressed either as a aßy2-tetramer (A) or a a^-trimer (B). The aßy2 form of FceRI is expressed on

mast cells and basophils of both humans and mice, while the 0,72 structure is found only in humans

on Langerhans' cells, monocytes, eosinophils, peripheral blood dendritic cells, and platelets. The

extracellular region of the a chain contains two lg-like domains (D1 and D2) responsible for binding

of IgE. The ß subunit has four transmembrane domains and an ITAM in the cytoplasmic tail. The y

chains form a disulfide-linked (S-S) homodimer with long cytoplasmic tails containing an ITAM on

each subunit.

IgE-FcsRI interactions

The unique binding kinetic of IgE to FcsRI is the most characteristic feature of this high

affinity receptor. FcsRI binds monomeric IgE with an association constant Ka of 1010 M"1

(Kulczycki and Metzger, 1974) which is considerably higher than binding of IgG to

FcyRIII, the closest homologue of FcsRI, characterized by a Ka of 105 M"1 (Maenaka et al.,

2001). This exceptionally high affinity of IgE binding to FcsRI reflects a very slow

dissociation rate, k0ff ~10"5 s"1 for FcsRI, compared with k0ff ~1 s"1 for FcyRIII (Ishizaka et

al., 1986). The most important implication for the biological function of IgE resulting from

such a slow dissociation rate is the long half-life of receptor-bound IgE, reaching 1-2 weeks

Page 24: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 21

(Tada et al., 1975). This results in persistent sensitization of cells expressing FcsRI, such as

mast cells and basophils, and is responsible for their capacity to react very fast to antigen

challenge, a feature characteristic of allergic responses. Recent investigation of crystal

structures of IgE and its high affinity receptor provided explanations for their unusual

binding kinetics (Wan et al., 2002). Binding of the Cs3 domain of IgE-Fc to the

extracellular domain of FcsRIa results in a profound conformational change of the adjacent

Cs2 domain. This conformational change is believed to greatly stabilize the IgE-FcsRI

complex and therefore lead to the slow dissociation rate (Novak and Bieber, 2002).

Biological functions of FcsRI

The best-known role of FcsRI is the induction of mast cell and basophil degranulation

when multivalent antigens crosslink the receptor by interacting with specific receptor-

bound IgE. The resulting processes, involving rapid release of granule-stored mediators as

well as synthesis and secretion of cytokines and chemokines, have been widely described

and shown to initiate allergic disorders and mediate immune responses against parasites

(Kawakami and Galli, 2002). The requirement for FcsRI in induction of IgE-dependent

allergic reactions has been shown in mice that lack either the a or ß chain gene of the

receptor as these mice do not exhibit cutaneous or systemic anaphylaxis (Dombrowicz et

al., 1993; Dombrowicz et al., 1998). In addition, FcsRI-deficient mice have an impaired

immunity to S. mansoni (Jankovic et al., 1997). In the S. mansoni model IgE-FcsRI

interactions are not directly involved in development of the Th2 response or resistance to

primary infection, but have been proposed to play a role in down-regulation of host

pathology by controlling hepatic fibrosis and egg granuloma formation. Moreover,

investigation of FcsRI on human eosinophils has revealed that during S. mansoni infection

FcsRI participates in eosinophil-mediated cytotoxicity against the parasite by promoting

eosinophil degranulation (Gounni et al., 1994).

Demonstration of FcsRI expression on human antigen presenting cells (APCs) has infened

a novel function of FcsRI in IgE-mediated antigen presentation pathways. Important

differences between FcsRI present on the classical IgE-associated effector cells (mast cells

and basophils) and professional APCs have to be pointed out. APCs express the trimeric

form of the receptor (ay2), which shows substantially lower expression density and

mediates weaker signals (Novak et al., 2003a). This suggests a distinct function for the high

Page 25: The Interplay of IgE and its High Affinity Receptor FceRI

22 Introduction

affinity IgE receptor on APCs. The importance of FcsRI in IgE-dependent antigen

presentation to T cells has been demonstrated on monocytes and circulating dendritic cells

(Maurer et al., 1995; Maurer et al., 1998). Studies of FcsRI-positive monocytes revealed

that high affinity IgE receptor-mediated endocytosis is more efficient than pinocytotic

allergen uptake. Thus, allergen presentation to T cells is highly enhanced when allergen-

IgE complexes are targeted to FcsRI-bearing APCs compared to T cell activation in

absence of IgE. This mechanism may therefore critically lower the atopic individual's

threshold to mount allergen-specific T cell responses. Interestingly, APCs of patients with

atopic diseases have been shown to express increased levels of FcsRI correlating with high

serum IgE concentrations (Kraft et al., 1998). Thus, FcsRI-mediated antigen presentation

has been proposed to mediate delayed-type hypersensitivity reactions (in contrast to

immediate-type hypersensitivity mediated by mast cells and basophils) involved in

diseases, such as atopic dermatitis (AD) and atopic eczema/dermatitis syndrome (AEDS),

in which aeroallergens penetrating the skin banier can be directly taken up by FcsRI-

bearing Langerhans' cells (LCs) leading to T cell activation and inflammation (Figure 5).

Moreover, a surprising functional difference in LCs from normal individuals and

individuals with AD has been observed: calcium mobilization upon crosslinking of FcsRI

can only be detected in LCs freshly isolated from patients with AD, but not in those from

skin of healthy individuals, despite the presence of significant amounts of the receptor

(Jürgens et al., 1995). The differential regulation of FcsRI expressed on LCs from atopic

patients has been reported to involve upregulated expression and activity of protein tyrosine

kinases (PTK) leading to activation of phospholipase C-y (PLCy) induced after IgE and

antigen ligation to FcsRI (Kraft et al., 2002b). Furthermore, signals through FcsRI in LCs

of atopic patients have been shown to induce expression of NF-kB resulting in activation of

proinflammatory cytokines, such as TNF-a and monocyte chemoattractant protein-1

(MCP-1) (Kraft et al., 2002a). These findings support the view that FcsRI-expressing APCs

may contribute to the inflammatory reactions found in atopic disease.

Taken together, recent investigations into the role of FcsRI on APCs have shed new light

on mechanisms controlling allergic and inflammatory hypersensitivity reactions. Additional

studies need to be undertaken to elucidate the detailed function of the high affinity IgE

Page 26: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 23

receptor expressed on different cell types in order to determine whether intervention with

FcsRI-mediated signaling cascades might be used for therapeutic strategies.

Stein

Allergen -.

NFLAMMATION

Lymph node

Figure 5. Contribution of FcsRI* LCs in development of AD. Allergen taken up through the skin

(a) binds to specific IgE present in high amounts in atopic patients. FceRI expressed on LCs

facilitates the uptake of allergens via IgE bound to the receptor leading to a more efficient

presentation of antigens and activation of LCs (b). In the regional lymph node LCs present the

antigen to naïve T cells resulting in activation of the T cells (c). Effector T cells migrate to the skin

where they trigger allergic inflammatory reactions, e.g. mediated by type-2 cytokines, which may

lead to dermatitis (d).

Page 27: The Interplay of IgE and its High Affinity Receptor FceRI

24 Introduction

1.3 Allergy

Definition of allergy

The term "allergy" was introduced by von Priquet in 1906 and it originally refened to all

forms of altered reactivity to antigenic stimulation, which may result in a protective

response (immunity) or an adverse clinical reaction (hypersensitivity) (Priquet, 1963).

However, the cunent usage of the term allergy describes potentially harmful immune

responses to environmental antigens, known as allergens. The pathology of allergic

diseases reflects activation of allergen-specific T cells, especially Th2 cells, and generation

of allergen-specific antibodies, mostly IgE. The term "atopy" (from Greek atopos, meaning

out of place) is used to describe IgE-mediated diseases. Patients with atopy produce IgE

antibodies against common environmental allergens and suffer from atopic diseases, such

as rhinitis, asthma, and eczema. Importantly, some allergic diseases, such as contact

dermatitis and hypersensitivity pneumonitis, develop through IgE-independent mechanisms

and can be therefore considered as nonatopic allergic conditions (Kay, 2001). Allergic

disease can usually be divided into three types of responses induced by allergen challenge:

(i) acute allergic responses, which can be elicited within minutes after exposure to allergen

reflecting the actions of mediators released from mast cells; (ii) late-phase reactions, which

develop several hours after allergen challenge and result in the recruitment of circulating

leukocytes; and (iii) chronic allergic inflammation, which occurs in response to repeated

allergen challenge and is characterized by long-lasting changes in the tissue and enhanced

propensity for Th2 cell-driven immune responses.

Allergens

An important question in the field of allergic disorders was asked by Aas in 1978: "What

makes an allergen an allergen?" (Aas, 1978). Although the first investigation of allergens

took place as early as the 1870s when Charles Blackley identified grass pollens as the cause

of his own disease, it is still not clear whether allergens are a specific subset of antigens or

whether any antigen can potentially become an allergen (Mohapatra and Lockey, 2001).

Molecular cloning and characterization of a large number of allergens has allowed the

identification of certain general characteristics of allergens. Allergens typically are proteins,

of which many show enzymatic activity (Cromwell, 1997). This led to the hypothesis that

Th2-type responses to enzymes evolved because of selection pressure that favored the

Page 28: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 25

development of immune responses to parasites that often require enzymes for successful

invasion of host tissues (Stewart et al., 1993). Other allergens are glycoproteins and it has

been proposed that oligosaccharide side chains may favor the binding to lectin receptors on

APCs and promote allergen uptake (Cromwell, 1997). The clinical classification of

allergens is based on their origin or patterns of distribution within the environment. Thus,

aeroallergens, further subdivided into indoor and outdoor aeroallergens, constitute airborne

particles, such as pollen grains, dust mite feces, and animal dander. Food allergens refer to

allergens ingested with food and the most prominent derive from peanuts, fish, cow's milk,

and cereal grains. Insect venoms represent an important group of allergens, because of their

high risk of causing anaphylaxis (Valentine, 1992). Although about 70 major allergens,

meaning allergens that induce immediate skin test responses in >90% of allergic

individuals, have been identified, it has not yet been revealed why these substances induce

Th2 cell-driven, IgE-associated responses. Thus, given such a wide range of allergens,

further research is required to understand the complexity of allergic disorders.

Factors involved in the development of allergy

Atopic diseases such as asthma, hives, eczema, atopic dermatitis, allergic rhinitis, and food

allergy have increased remarkably over the last two decades, and are now a major source of

disability worldwide, affecting mainly industrialized countries (1998; Holgate, 1999;

Sunyer et al., 1999). Due to the large number of people affected and the high annual costs

of treating allergic disorders, a large body of literature has accumulated discussing factors

that promote development of allergy.

Genetics

Investigation of asthma and eczema within families has revealed that allergy has a genetic

basis. Twin studies have shown that the heritability of asthma may be as high as 75%

(Duffy et al., 1990) and the presence of eczema-specific genes has been suggested from the

observation that parental eczema confers a higher risk of eczema in offspring than parental

asthma or rhinitis (Dold et al., 1992). Techniques used to identify genes that are relevant to

allergy and asthma include the candidate-gene approach, which depends on the

identification of polymorphisms in a known gene, and positional cloning, which links the

inheritance of a specific chromosomal region with the inheritance of a disease (Cookson,

1999). Studies using the candidate-gene method have revealed an association between an

Page 29: The Interplay of IgE and its High Affinity Receptor FceRI

26 Introduction

allele of the human leukocyte antigen (HLA)-DR locus and reactivity to the ragweed

allergen Ra 5 (Marsh et al., 1982), and the linkage of atopy to a polymorphism of FcsRIß

(Hill and Cookson, 1996) and to the IL-4 family of cytokine genes on chromosome 5

(Marsh et al., 1994). Positional cloning has shown that chromosomes 2q, 5q, 6q, 12q, and

13q contain loci linked both to asthma and atopy (Cookson, 1999). However, the clinical

relevance of these findings remains to be determined.

Feto-maternal events

It is widely recognized that most atopic disorders originate in childhood and therefore

research on the early development of the immune system is important to understand the

chronology of events leading to allergic reactions. Experimental data has suggested that

infants can develop an immune response to common environmental antigens whilst in utero

(Warner et al., 2000). Specific allergen-induced responses have been shown to occur as

early as 22 weeks of pregnancy and maternal exposure to birch pollen from this point

onwards results in increased infant allergen-specific responses at birth, suggesting that

antigen passage can occur across the placenta (Jones et al., 1996). Furthermore, peripheral

blood mononucleocytes (PBMCs) from babies with a family history of allergy have been

demonstrated to lack production of the Thl cytokine IFN-y (Holt et al., 1992; Tang et al.,

1994). Interestingly, several reports have proposed that during pregnancy the immune

response at the materno-fetal level is biased towards a Th2 response (Prescott et al., 1998).

This implies that fetal IFN-y production may be required to counterbalance the Th2

environment of the placenta in order to prevent an allergic Th2 phenotype in infants.

However, if the mother is atopic and exposed to allergens that trigger her own allergy

during pregnancy, Th2-type cytokines could potentially cross the placenta and hamper the

fetal IFN-y response. Early exposure of the infant to the same allergens may additionally

influence the immune response, resulting in atopic disease, as shown by the observation

that the level of house dust mite exposure during the first year of life correlates with the

subsequent risk of childhood asthma (Sporik et al., 1990).

Lifestyle

Results of international epidemiological studies have provided ample evidence that the

increase in the prevalence of allergic disease correlates with western lifestyle. Substantial

regional differences in importance of atopy have been shown over the past 20 years (1998)

Page 30: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 27

including studies of West and East Germany (von Mutius et al., 1998) as well as West and

East Europe (Bjorksten et al., 1998). Furthermore, rural lifestyle has been shown to have

protective effects on the development of allergies (Braun-Fahrlander et al., 1999). Large

families or animal pets have also been listed amongst the factors protecting against atopy

leading to the hypothesis that an excessively clean environment may result in inappropriate

immune responses (Rook and Stanford, 1998). It has been proposed that microbial

stimulation in the gastrointestinal tract could possibly explain the association of atopic

disease with clean western lifestyle as microbial antigens stimulate Thl cell activation. An

example of a link between bacteria colonizing the gastrointestinal tract and atopic

sensitizations has been described in a study of Swedish and Estonian children (Bjorksten et

al., 1999; Sepp et al., 1997). While lactobacilli and eubacteria were predominant in non-

atopic individuals, coliforms and Staphylococcus auerus dominated in allergic children.

The findings that atopic sensitization might be influenced by the bacterial species

colonizing the gut may explain why living in a rural community, which increases the

likelihood of exposure to bacteria found in barns, might protect from allergic disease.

The hygiene hypothesis versus the counter-regulatory model

The argument that bacterial and viral infections during early life direct the maturing

immune system toward Thl responses, which counterbalance allergy-associated responses

of Th2 cells, has led to the so-called hygiene hypothesis, first postulated in 1989 by David

Strachan (Strachan, 1989). The hygiene hypothesis argues that the increased prevalence of

allergic disease results from a reduced exposure to microbes that are more common on

farms and in less developed countries. This view has been supported by findings suggesting

that childhood infections show a negative association with atopy. In particular, exposure to

food and orofecal pathogens, such as hepatitis A, Toxoplasma gondii, and Helibacter

pylori, has been shown to reduce the risk of allergic disease by >60% (Matricardi et al.,

2000). However, studies reporting that the prevalence of Thl-dependent autoimmune

diseases is also increasing and that Th2-associated parasite infections do not increase the

risk of allergy, contradict the hygiene hypothesis and imply that the Thl/Th2 balance model

should be re-evaluated (Allen and Maizels, 1997; Gor et al., 2003). Thus, recent reviews

have favored a novel, so called "counter-regulatory" model, which offers a unifying

explanation for the increasing incidence of both allergy and autoimmunity in the absence of

persistent immune challenge characteristic for a westernized lifestyle (Wills-Karp et al.,

Page 31: The Interplay of IgE and its High Affinity Receptor FceRI

28 Introduction

2001; Yazdanbakhsh et al., 2002). Anti-inflammatory cytokines, such as IL-10 and

transforming growth factor (TGF)-ß, have been suggested to be the key players in the

counter-regulatory model. As most pathogenic infections have been shown to upregulate

IL-10 production, consequently suppressing allergic and autoimmune disease, decreased

infectious pressure found in developed countries might result in decreased levels of anti¬

inflammatory cytokines required for prevention of deleterious immune responses (Moore et

al., 2001; van den Biggelaar et al., 2000). This is supported by a previous finding of

decreased IL-10 production in asthmatic patients (Borish et al., 1996).

In summary, the interrelation between pathogenic infections and development of atopic

disorders seems to involve complex regulatory mechanisms that remain to be

experimentally proven. Factors required for counter-regulation, such as cell types and

anatomical locations involved, should be addressed in animal models to increase our

understanding of the mechanisms regulating the development of allergic disease.

Animal models used in the study of allergy

Various animal models, including mice (Levine and Vaz, 1970), rats (Byars and Fenaresi,

1976), guinea pigs (Tanaka et al., 1988), monkeys (Turner et al., 1996), and dogs (Ermel et

al., 1997), have been established in an attempt to provide insights into the immunology and

pathophysiology of human atopic disorders. The murine system has been widely used in

immunologic research because of the availability of a multitude of genetically engineered

mice as well as detailed knowledge of the murine genome and immune system.

Additionally, various techniques and reagents have been developed to assess

immunological responses in mice. Models to investigate allergic-type diseases require close

resemblance to the pathology of the disease in humans and should allow measurement of

physiologic parameters. Numerous sensitization protocols with food- and aero-allergens

have been used to induce an atopic phenotype in mice (Lloyd et al., 2001). Generation of

recombinant allergens has been shown to be advantageous to obtain reliable and

reproducible experimental data (Herz et al., 2004). Sensitization to ovalbumin (OVA) and

challenge with aerosolized OVA represents the most widly used protocol used in mice to

study human bronchial asthma (Neuhaus-Steinmetz et al., 2000). Nevertheless, methods

based on immunization with high amounts of protein antigen do not reflect the natural

pathway of atopic sensitization leading to allergic reactions in humans. Thus, natural

Page 32: The Interplay of IgE and its High Affinity Receptor FceRI

Introduction 29

murine infections inducing a Th2-type immune response including high levels of IgE, such

as helminth infections, have also been used to investigate mechanisms involved in

development of allergy (Finkelman et al., 1997). Although helminth infections have

provided important insight into the regulation of Th2 responses, they are often cleared by

the mouse and therefore do not induce a persistent IgE response that would mimic chronic

allergic diseases in humans. In consequence, novel improved murine models that would

resemble more closely the features of human allergy are required to allow investigation of

all aspects of the disease.

Page 33: The Interplay of IgE and its High Affinity Receptor FceRI

30 Introduction

1.4 Central Question

The main objective of this thesis was to investigate mechanisms involved in the regulation

of IgE production and to analyze the role of IgE interactions with its high affinity receptor,

FcsRI. To reach this aim, a new model of a murine infection resulting in allergy-like

symptoms was established and characterized (Results Part I). In particular, natural murine

ectoparasites, the fur mites Myocoptes musculinus and Myobia musculi, were used and their

value as a model for atopic disorders evaluated. Furthermore, a transgenic mouse model

mimicking the expression pattern of the human FcsRI was generated in order to provide a

tool to study the role of FcsRI expression on the same cell types as found in humans

(Results Part II). Lastly, an attempt to generate a monoclonal antibody against the a chain

of the murine FcsRI was made to allow specific analysis of the receptor expression (Results

Part III).

Page 34: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 31

2 Results Part I

Induction of IgE and allergic-type responses in fur

mite-infested mice

2.1 Abstract

High serum IgE levels are characteristic of allergic diseases and immune responses to most

parasites. A murine allergy model based on infestation with the fur mites Myocoptes

musculinus and Myobia musculi was investigated. Analysis of mite infestation in various

knockout mice revealed that IgE production in response to these ectoparasites was

dependent on T cells, IL-4, and CD40L interaction. Secretion of IL-4 by CD4+ T cells

obtained from peripheral lymph nodes draining mite-infested skin sites was increased with

progressing mite infestation and correlated with the serum IgE induction. A time course

analysis of the mRNA expression of s germline, switched IgE, and AID transcripts

suggested that switching to IgE in response to fur mites occuned initially in skin-draining

lymph nodes. In addition, mite infestation induced mast cell degranulation in the skin as

well as mast cell infiltrations into skin-draining lymph nodes. Analysis of the immune

response generated in mite-infested mice should provide information useful for better

understanding of mechanisms involved in IgE induction and regulation. Due to the

physiological way of allergen-exposure resembling the atopic sensitization in humans, mite

infestation might offer a highly valuable model for the investigation of allergic disorders.

Page 35: The Interplay of IgE and its High Affinity Receptor FceRI

32 Results Part I

2.2 Introduction

IgE is the main antibody isotype involved in allergic disease and immune responses against

parasites. The direct effects of IgE during an immediate-type hypersensitivity reaction are

well characterized (Corry and Kheradmand, 1999; Kay, 2001; Kinet, 1999). However,

many of the specific factors involved in the initiation and induction of IgE are yet to be

determined. Many components influencing development of atopy have been proposed, such

as lifestyle, diet, and genetic background (Cookson, 1999). Nevertheless, it is still unclear

why some individuals develop allergy while others remain allergy-free. Investigation of

factors regulating the induction of IgE is therefore of crucial importance to elucidate the

etiology of allergic diseases.

Nematode infection models have been widely used to study IgE-related immune responses

(Finkelman et al., 1997; Shea-Donohue and Urban, 2004). However, in contrast to allergic

diseases, which are chronic, frequently used parasites, such as Trichinella spiralis and

Nippostrongylus brasiliensis, induce Th2-type responses leading to protection. Other

frequently employed methods to study allergic disorders have been based on immunization

with high amounts of purified protein antigens in adjuvants, which induce some of the

symptoms of allergy or asthma (Herz et al., 2004). While this can provide information on

the effects of IgE after induction, it is important to investigate additional in vivo models,

which could yield physiologically relevant information on the factors regulating the

initiation of allergic responses.

Murine fur mite infestation was previously suggested to closely mimic the development of

allergic type responses (Jungmann et al., 1996a; Laltoo et al., 1979; Weisbroth et al., 1976),

however, little research has been done on the immune response to ascariasis. Most common

mouse fur mite species are Myocoptes musculinus and Myobia musculi (Gambles, 1952).

These mites are natural parasites of mice and live their entire life cycle on the same host

(Friedman and Weisbroth, 1977). Interestingly, despite the fact that these mites are

classified as ectoparasites and do not penetrate the deeper layers of the skin, infested mice

develop greatly increased total serum IgE levels (Jungmann et al., 1996b; Laltoo et al.,

1979). Following prolonged periods of infestation, mice develop skin lesions known as

mite-associated ulcerative dermatitis (MAUD) of which severity is influenced by the

Page 36: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 33

genetic background of the infested mice (Dawson et al., 1986; Friedman and Weisbroth,

1975).

In the present study we characterized the initial requirements of IgE induction in mite-

infested mice. We show that fur mite infestation represents a good animal model for

investigation of allergic-type immune responses involving activation of Th2 cells and

secretion of IL-4. T cell activation and B cell isotype switching to IgE appear to occur

within skin-draining LN. Furthermore, mast cell degranulation was greatly increased in the

skin and increased mast cell infiltration in skin-draining LN was observed. These data

suggest that mite antigens enter through the skin of infested animals leading to induction of

a Th2-type response and B cell switch to IgE in skin-draining LN. Murine mite infestation

is therefore a promising physiological animal model in which to study induction of allergic

responses.

Page 37: The Interplay of IgE and its High Affinity Receptor FceRI

34 Results Part I

2.3 Materials and Methods

Mice and mite infestation

C57BL/6, CD40L"A (Renshaw et al., 1994), IL4"A (Kopf et al., 1993), IL5"A (Kopf et al.,

1996), and TCRßo"" (Mombaerts et al., 1993) mice were bred and maintained at the

Institute of Laboratory Animal Science of the University of Zurich. BALB/c mice were

purchased from Harlan (Netherlands).

For mite infestation female mice at 6-8 weeks of age were put into a cage with a previously

infested mouse carrying a mixed population of Myocoptes musculinus and Myobia musculi.

Hereafter infestation occurred by direct contact. Transgenic mice were housed in the same

cage with respective control mice in order to avoid cage-cage variation.

Serum IgE and IgGi measurement

Blood was collected weekly and total serum IgE and IgGi concentrations were determined

by enzyme-linked immunosorbent assays (ELISA). ELISA plates were coated with 5 ug/ml

rat anti-mouse IgE (clone 6HD5) or with 1 ug/ml goat anti-mouse IgGi (Southern

Biotechnologies) in 0.1 M NaHCÛ3 (pH 9.6) and blocked with 5% (w/v) bovine serum

albumin (BSA). Serially diluted serum or standards, IgE (clone TIB-141) and IgGi

(Zymed), were added and incubated for 2 hours. Bound IgE was detected with biotinylated

anti-mouse IgE (clone RIE-4) at 2.5 ug/ml followed by horseradish peroxidase-conjugated

streptdavidin (Jackson ImmunoResearch Laboratories) at 1 ug/ml. Bound IgGi was

detected by horseradish peroxidase-conjugated anti-mouse IgG (Sigma). ABTS peroxidase

solution (2,2'-azino-bis-[3-ethylbenzthiazidine-6-sulfonic acid] 0.1 mg/ml, 0.05% H202,

and 0.1 M NaH2P04 pH 4.0) was used for the peroxidase-catalyzed color reaction. Color

reaction was read at 405 nm in a Bio-Rad microplate reader and standard curves and

antibody concentrations analyzed with Microplate Manger III software (Bio-Rad).

Flow cytometry

To obtain single cell suspensions, lymph nodes and spleens were smashed though nylon or

metal grids whereas lungs were digested for 1 hour at 37°C in DNase I (50 ug/ml, Fluka

Chemie) and Collagenase (500 ug/ml, Invitrogen). Cells were restimulated for 6 hours with

PMA (50 ng/ml) and ionomycin (500 ng/ml) in the presence of Brefeldin A (5 ug/ml).

Page 38: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 35

Collected cells were surface-stained with anti-CD4-FITC (5 ug/ml, BD Biosciences), fixed

with 2% paraformaldehyde and permeabilized in 0.1% saponin. Intracellular staining was

performed with anti-IL-4-APC (2 ug/ml, BD Biosciences), anti-IL-5-PE (2 ug/ml, BD

Biosciences), or anti-IFN-y-APC (1 ug/ml, BD Biosciences).

Histochemistry

Freshly removed organs were immersed in Carnoy's fixative (60% Ethanol, 20%

Chloroform, 20% Acetic acid). Sections of 5 urn thickness were cut in paraffin, and stained

with toluidine blue using standard procedures.

RT-PCR and sequencing

B cells were purified to > 95% from spleen and lymph nodes by magnetic-assisted cell

sorting (MACS) (Miltenyi Biotec) using anti-B220 microbeads. Total RNA was isolated

using TRIzol reagent (Invitrogen Life Technologies) according to the manufacturer's

instructions. First-strand cDNA synthesis of 1 ug total RNA was performed with random

hexamers and SuperScript II reverse transcriptase (Invitrogen Life Technologies). 1 ul of

the resulting cDNA was amplified by PCR. Primer sequences and PCR conditions were as

follows: mature IgE switched transcripts (962 bp): 5'-CCTGCCCTCGGTTCTGA-3' and

5'-CTAGGCGACTGAAGATGAAG-3' (94°5'; [94°1'; 58°1'; 72°l']x35; 72°10'; 10°), s

germline transcripts (300 bp): 5'-GCAGAAGATGGCTTCGAATAAGAACAGT-3' and

5'-TCGTTGAATGATGGAGGATGTGTCACGT-3' (94°5'; [94°1'; 60°1'; 72°l']x35;

72°10'; 10°), u germline transcripts (325 bp): 5'-CTCTGGCCCTGCTTATTGTTG-3' and

5'-ATGGTGCTGGGCAGGAAGTC-3' (94°5'; [94°1'; 60°1'; 72°l']x35; 72°10'; 10°),

AID (300 bp): 5'-GGCTGAGGTTAGGGTTCCATCTCAG-3' and 5'-

GAGGGAGTCAAGAAAGTCACGCTAGA-3' (94°5'; [94°1'; 60°1'; 72°l']x35; 72°10';

10°).

For sequence analysis cDNA was synthesized from B cells purified from spleens of day 80

mite-infested mice. V(D)JH-Cs transcripts were amplified by PCR using a degenerative

primer mix (Krebber et al., 1997) specific for the Vh region and a reverse primer that

anneals within exon 1 of the Cs gene (IgEc, 5'-TCTGAATACCAGGTCACAGTC-3').

PCR products were purified using the agarose gel extraction kit (Qiagen) and subcloned

directly into the pGEM-T vector (Promega) for automated sequencing.

Page 39: The Interplay of IgE and its High Affinity Receptor FceRI

36 Results Part I

2.4 Results

Mite infestation induces IgE

To investigate the antibody response induced by ascariasis, total serum IgE levels were

analyzed in C57BL/6 and BALB/c mice infested with two common species of mouse fur

mites, Myobia musculi and Myocoptes musculinus. Blood was collected at weekly intervals

following infestation to determine the kinetics of IgE induction. Upregulation of total

serum IgE was first observed 15 days after mite infestation (Figure 6A) followed by a

gradual increase for about ten weeks. Around 90 days post infestation IgE levels reached a

1000-fold increase from about 3 ng/ml to 2000 ng/ml in C57BL/6 mice and 60 ng/ml to

lO'OOO ng/ml in BALB/c mice. These high serum IgE levels then remained stable up to two

years after first contact with the ectoparasite (data not shown). In addition, total serum IgGi

levels were also determined (Figure 6B). The kinetics for IgGi induction showed a similar

course as observed for IgE, however the increase (10-fold in C57BL/6 and 20-fold in

BALB/c) was less prominent. Thus, fur mite infestation parallels an allergic situation where

high serum IgE levels are chronically induced in response to the constant presence of

naturally occurring allergens.

B104 k__ A- -A

A= 104

enc

UJ

5> 103_^r*rL-*rhr*

h-T-

2 k~

o

O

2 102

io2 ^x'a^t r""

**

10 /

1 £ 10 "

A C57BL/6

A BALB/c

0.1 1

0 15 30 45 60 75 90 105 0 15 30 45 60 75 90 105

Days post mite infestation Days post mite infestation

Figure 6. Mite infestation induces IgE and IgGi. C57BL/6 mice (closed symbols) or BALB/c mice

(open symbols) were infested with fur mites at day 0. Blood was collected at weekly intervals and

total serum IgE (A) and lgG1 (B) was determined by ELISA. The data points represent the mean +

standard deviation (s.d.) of 11 C57BL/6 and 8 BALB/c mice. Results are representative of five

separate experiments. The dotted line in A indicates the lower limit sensitivity (0.7 ng/ml) of the IgE

ELISA.

Page 40: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 37

Induction of IgE in response to mite infestation requires T cells, CD40L, and IL-4

To investigate which factors are required for the immune response induced against mites,

several different knockout mice were infested. At various time points thereafter, blood was

collected and total serum IgE and IgGi concentrations were analyzed. A requirement for T

cells for mite-induced IgE was confirmed by investigating TCRßo""

mice (Mombaerts et

al., 1993), as IgE levels remained below detection limit throughout the time of mite

infestation (Figure 7, upper panel). T cells induce isotype switch to IgE by providing

cytokines and through direct cognate interaction with B cells. To investigate if both forms

of T cell help were required, mice deficient in CD40L, IL-4 or IL-5 were infested with

mites and serum IgE levels monitored. As expected, no increase in serum IgE levels was

observed in either IL-4 (Köpfet al., 1993) or CD40L (Renshaw et al., 1994) deficient mice,

however, the absence of IL-5 (Köpfet al., 1996) did not alter IgE production in response to

mites (Figure 7, upper panel). IgGi measurements led to comparable observations (Figure

7, lower panel).

These data suggest that mite infestation induces B cells to switch to IgE or IgGi under the

influence of signals through CD40 and the Th2-type cytokine IL-4. Thus, the immune

response to fur mites requires T cells and IL-4, as observed for IgE induction during

allergic reactions. Therefore, mite infestation may serve as a suitable model for

investigation of allergic responses causing high serum IgE levels.

Page 41: The Interplay of IgE and its High Affinity Receptor FceRI

38 Results Part I

104

TCRßS-/- CD40L-/- IL-4-/- IL-5-/-

--

--

Jc

m

103^--- A-*- * *"* ,*^^

102/

--

/ -,if

/ *

;/2 10 J

¥*'

/

,o 1 '-

AH

*—^c> o -o- o ' \^ ( o<^^^-^^^-^^S*

0.1

105

' 1

j ,

1 >

¥"ra3.

104 ----*—*- - '„# - -p-

"W

"r 103 - -

o

2 102 -

x:^>- --o- -O^'^"~v—v \

\-

~-<^- -o

"5 ?r-*---<>

':y °

,o 10 i"

t

"

KOh- WT

1 1

r

'

0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100 0 20 40 60 80 100

Days post mite infestation

Figure 7. Absence of IgE or IgGi induction in response to mite infestation in mice deficient in

T cells, CD40L or IL-4. Knock-out (open symbols) and WT control mice (closed symbols) were

infested with mites by direct contact and housed together in the same cage. Total serum IgE (upper

panel) and lgG1 (lower panel) of TCRßS"'", CD40L"'", IL-4_/", and IL-5_/" mice were determined by

ELISA at different time points after infestation with mites. The data points represent the mean + s.d.

of 4-5 mice per group. Results are representative of two separate experiments. The dotted line in

the upper panel indicates the lower limit sensitivity (0.7 ng/ml) of the IgE ELISA.

Page 42: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 39

Mite infestation induces IL-4 secretion in skin-draining LN

To further characterize the immune response mounted against fur mites, we investigated the

site of T cell activation, which could provide insight into the site of antigen entry. For this

purpose, we determined the cytokine profile of T cells in spleen, lung, and LN draining the

skin (brachial and cervical LN), the gut (mesenteric LN), and the lung (mediastinal LN).

Cells were isolated at several time points after mite infestation and re-stimulated in vitro

with PMA and lonomycin. Percentages of IL-4-, IL-5-, and IFNy-producing cells were

determined by intracellular staining. Representative dot plots of IL-4+ CD4+ T cells from

three different time points post mite infestation are shown in Figure 8A. IL-4 secretion in

response to mite infestation was observed in CD4+ T cells from skin-draining LN,

mediastinal LN, and lung, but not spleen or mesenteric LN (Figure 8B). Importantly, while

significant increase in IL-4 production in mediastinal LN and lung occurred only late after

mite infestation (>150 days), the increase in IL-4 levels in skin-draining LN was observed

already 30 days post infestation, conesponding to the time of IgE upregulation. These data

suggest that the IL-4 responsible for B cell switch to IgE in mite-infested mice was

produced in skin-draining LN. In contrast, CD4+ T cell secretion of IL-5 was slightly

increased above background levels only at very late time points (>150 days, data not

shown). This correlates well with the very low levels of peripheral blood eosinophilia

observed in infested mice. In our animal facility the percentage of eosinophils in peripheral

blood of naïve C57BL/6 mice averages 0.6% (±0.1% SEM, n=38) and was increased to

2.5%) (±0.2%) SEM, n=38) on day 60 post infestation. After this time eosinophil levels

remained stable or even declined. T cell secretion of IFNy remained stable at background

levels except late time points after mite infestation (data not shown).

Taken together, these results show that T cell activation in response to contact with fur

mites was induced in skin-draining LN, suggesting that mite antigens enter initially via the

skin and not through other possible routes, such as the lung or the gut. Moreover, skin-

draining LN were increased in size following mite infestation while the size of other LN

remained stable (data not shown). This provides additional evidence that after uptake of

mite antigen T cells are activated in skin-draining LN.

Page 43: The Interplay of IgE and its High Affinity Receptor FceRI

40 Results Part I

day 0 day 60

0.17% 1.99%

day 320

4.52%

B

CD4

brachial+cervical LNs mediastinal LNs mesenteric LNs

+

o

ü

+

0 15 30 45 60 165 320 0 15 30 45 60 165 320 0 15 30 45 60 165 320

spleen lung

CZ3 PMA/lonomycmUnstimulated

1kzz:ez n

i i3 X rL

}\

jJ J

0 15 30 45 60 165 320 0 15 30 45 60 165 320

Days post mite infestation

Figure 8. CD4+ T cells in skin-draining LN produce IL-4 in response to mite infestation. A)

Representative dot plots of brachial and cervical LN isolated on day 0, 60, and 320 post mite

infestation after PMA/lonomycin restimulation. The percentage of IL-4+ CD4+ cells is indicated. B)

Cells isolated from LN, spleen, and lung on indicated time points after mite infestation and

stimulated with PMA/lonomycin (light bars) or with medium alone as controls (dark bars). Gates

were set on live CD4+ cells and IL-4 expression was analyzed. Results represent the mean + s.d. of

5 mice per group. Results are representative of two separate experiments.

Switching to IgE is induced in skin-draining lymph nodes

In order to determine at which sites B cell switching to IgE was occuning, B cells were

purified from the spleen and LN draining the skin, lung, and gut of naive and mite-infested

mice RT-PCR was then performed to detect s germline transcripts (GLT), uGLT, mature

switched IgE, and activation-induced cytidine deaminase (ADD) The presence of sGLT

provides evidence for the initial steps leading to IgE class-switch recombination (CSR)

The "sterile" sGLT, which lacks the VDJ region, is an essential prerequisite for CSR AID

Page 44: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 41

has been shown to play a crucial role in CSR (Muramatsu et al., 2000) and its expression

indicates initiation of CSR. Mature IgE transcripts, resulting from DNA excision and

ligation of the VDJ sequences to the constant s locus, indicate the presence of IgE switched

B cells. uGLT expression was also analyzed and used as an internal control as uGLT result

from heavy chain VDJ rearrangement during B cell development and characterize

functional B cells. Our results show an upregulation of sGLT and mature IgE transcripts

induced in B cells isolated from skin-draining LN, while in B cells from spleen and

mesenteric LN the expression of all transcripts remained comparable over the time of

infestation (Figure 9). AID expression was also upregulated in skin-draining LN but to a

lesser extent. Importantly, the levels of uGLT remained constant, indicating that B cell

isolation and cDNA synthesis were comparable in all samples (Figure 9).

Thus, these data suggest that initial B cell switching to IgE in response to mite infestation

occurs in skin-draining LN, corresponding to the site of initial IL-4 production.

eGLT IgE AID uGLT

day 15 ^^J^^^^^J ^^^^^^^^5 pBlBfijj^^^s ^^^^^^^^3

day 23 ^SS^^^^S ^^^EiHiiS ^H^^^^^S

ci 8.y 35 ^^B^^^M^^j ^^^^^^^^^^3 ''"I ^^^^^^^^^^3

_j-~~ _L~~ + — + —

Hl^p||^mi|||||||^^B ||^^^^BiiSiii"iiiBi JiiiiiiiiiiiiiiiiiiiiiiiiiiillimHHHHHHHHHm^^M tfjJiiiittMMIIMEflflflSfll

Pill k^ riHInco k^ EÜ^B^nD hn HIHwhn

Figure 9. Messenger RNA expression of eGLT and mature switched IgE transcripts is

upregulated in skin-draining LN upon mite infestation. B cells were isolated from spleen (Spl),

brachial and cervical LN (sLN), and mesenteric LN (mLN) of naïve or fur mite-infested C57BL/6

mice at 15, 23, and 35 days post infestation. Positive controls (+) derive from RNA isolated from B

cells stimulated in vitro with IL-4 and anti-CD40 for 40 hours. Negative controls (-) are H20. Results

are representative of two independent experiments.

Page 45: The Interplay of IgE and its High Affinity Receptor FceRI

42 Results Part I

IgE induced by mite infestation is polyclonal

Parasite infections are known to induce an increase in total IgE although the levels of

parasite-specific IgE are usually low (Hagan, 1993; Pritchard, 1993; Turner et al., 1979).

This implies that parasites may induce a polyclonal non-specific response. In order to

investigate the diversity of the IgE repertoire induced by mite infestation, we determined

the usage of the V, D, and J segments of IgE heavy chains present in B cells of mite-

infested mice. For this purpose, B cells were isolated from spleens of infested mice and

V(D)J Cs transcripts were amplified by RT-PCR and sequenced. Sequences containing an

open reading frame and the correct Cs sequence were then aligned to the closest germ-line

sequence using the DNAplot database (www.dnaplot.de) to determine the Vh, Dh, and Jh

gene usage and the number of mutations within the framework (FR) and the

complementarity-determining regions (CDR). Analysis of twelve sequences obtained from

three C57BL/6 mice is shown in Table 1 and 2. The majority (11/12) of analyzed clones

were found to align to different germline sequences suggesting that the antibody response

to fur mites is not directed against a single antigen. Furthermore, the sequences of Vs

transcripts of mite-infested mice were not highly mutated suggesting that they may not

have undergone somatic hypermutation. This reveals the possibility that the IgE produced

in response to mite infestation might not result from a germinal center reaction. However,

many more sequences need to be analyzed to allow for representative conclusions.

Table 1. V(D)J Ce rearrangements in mite-infested C57BL/6 mice.

Clone VH family3 DH family JH family -

No. of mutations'3

FR CDR FR

R/Sc

CDR

B6#1-VDJ-1.6 v„i DFL16.2 J„3 21 3 0.8 2 .0

B6#l-VDJ-1.12a v„i DFL16.1 J„l 2 0 1.0 NA

B6#1-VDJ-1.18 v„i - J„3 15 5 1.1 0.6

B6#1-VDJ-1.23 v„i DFL16.1 J„4 2 1 NA NA

B6#l-VDJ-2.3 v„i DFL16.1 J„2 19 6 2 .8 5.0

B6#l-VDJ-2.47 v„i - J„3 12 3 1.4 NA

B6#2-VDJ-1.46 V„5 DSP2.9-C J„2 2 3 1.0 NA

B6#3-VDJ-1.52 V„3 DSP2.X J„3 15 10 1.1 4 .0

B6#3-VDJ-1.53 v„i DFL16.1 J.4 16 4 1.0 1.0

B6#3-VDJ-1.65 v„i DFL16.1 J.2 11 7 2 .7 2 .5

B6#3-VDJ-1.70 v„i DQ52-C J„2 11 6 1.8 NA

aClosest germline VH families of clones isolated from three mite-infested C57BL/6 mice (B6#1-3) were

assigned using the DNAplot database (www dnaplot de) bNumbers of mutations in the framework (FR) and the

complementarity-determining region (CDR) were determined for each sequence cRatio of replacement to

silent mutations (R/S) was calculated unless not applicable (NA) dSequence was isolated in two clones after

PCR reaction

Page 46: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 43

Table 2. V(D)J Ce sequences in mite-infested C57BL/6 mice.

Clone VH (P)Na DH (P)Na JH

B6#1-VDJ-1.6 TGTGCAAGA GAGACCTCG ATTACTACG ATAC CCTGGTTTG

B6#l -VDJ-1.12 TGTGCAAGA GAGA ATTACTACGGTAGTAGCTAC CC CTACTGGTA

B6#1-VDJ-1.18 TGTGCAAG GGGG - - GCTTACTGG

B6#1-VDJ-1.2 3 TGTGCAAG GAATGGACCCTT CTACGGTAGTAÇCTAC GAGGT TGCTATGGA

B6#l-VDJ-2.3 TGTACAAGA GGGA ATTTCTACGGTAGTAGCAAC CCA TACTTTGAC

B6#l-VDJ-2.47 TGTGCAAGA G - - GCTTACTGG

B6#2-VDJ-1.46 TGTGCAAGACTA CCCT ATAGTAACCTT - TACTTTGAC

B6 #3-VDJ-1.52 TGTGCAAAA AA TAACTAC GGGA GGTTTACTT

B6#3-VDJ-1.53 TGTGCAA TGCAATTCCCCC TTATTACTTC TT TGGACTACT

B6 #3-VDJ-1.6 5 TGTGCAAGA TCCC TÇTATTTCTTCGGTAGTAGTT - TTTGACTAC

B6#3-VDJ-1.7 0 TGTGCAAGA - TCCCÇGÇG GGCTAACTGGG ACTACTTTG

Nucleotides inserted between the Vh, Dh, and Jh sequences Differences to the closest germline sequence

are underlined

Mite infestation induces mast cell degranulation in skin and mast cell recruitment to

skin-draining LN

To investigate the effects of the high IgE concentrations induced upon mite infestation, we

performed histochemistry analysis of mast cells by staining sections of several organs with

toluidine blue. Total mast cell numbers as well as numbers of degranulated mast cells were

determined. While total mast cell numbers in skin sections did not change throughout the

time of mite infestation (Figure 10A), the numbers of degranulated mast cells increased

greatly upon exposure to mites (Figure 10B). The dramatic increase in degranulation is

clearly visible in figures 10C-F, while no such increase was observed in mice deficient in T

cells (Figure 10G) or CD40L (Figure 10H) even late after mite infestation. Induction of

mast cell degranulation suggests the presence of mite-specific IgE, which would be

crosslinked by mite antigens. It also demonstrates that scratching alone resulting from mite

presence on the fur cannot be responsible for mast cell degranulation as scratching was also

observed in TCRßo"" and CD40L"" mice. Thus, our results strongly suggest that the

immune response resulting from mite infestation is directed against specific antigens.

Moreover, we observed an accumulation of mast cells in skin-draining LN (Figure 10I-L)

but not in gut-draining LN (Figure 10M-N). No mast cell infiltration was found in any

other organs such as spleen, lung, jejunum, ileum or colon (data not shown). Mast cells

infiltrating skin-draining LN upon mite infestation may interact with lymphocytes and

influence mite-induced immune responses. However, the relevance of such interactions

remains to be determined.

Page 47: The Interplay of IgE and its High Affinity Receptor FceRI

44 Results Part I

B

0 15 30 45 60 165 320

Days post mite infestation

_ D

^i'

.J • ^

E"

ff;'*:•«. F..%»- •

w'/(* „ ,#

Hm f.

f

*. 'I; '-•#•. 'V- '*''.'"» % m- V ,% »

' • • fer'"

-

« tt ' 'V*

«s

ü

S 60

I| 40

c

E

g» 20

•a

5?.

0 15 30 45 60 165 320

Days post mite infestation

m

Figure 10. Mast cell degranulation occurs in skin of mite-infested mice. Total numbers + s.d. of

mast cells per 10 high power fields (HPF) of a 20x magnification (A) and the percentage + s.d. of

degranulated mast cells (B) were determined in skin sections of WT C57BL/6 mice at different time

points after mite infestation. Tissue sections stained with Toluidine blue are depicted as follows:

skin of naïve (C) or mite-infested WT C57BL/6 mice on day 45 (D), 165 (E), and 320 (F) post mite

infestation; skin of TCRßS-deficient (G) and CD40L-deficient (H) mice on day 200 post mite

infestation; cervical LN of naïve (I) or mite-infested WT C57BL/6 mice on day 45 (J), 165 (K), and

320 (L) post mite infestation; mesenteric LN of naïve (M) or mite-infested WT C57BL/6 mice on day

320 (N) post mite infestation. The arrows indicate examples of degranulated mast cells. Results are

representative of three mice per group and two independent experiments.

Page 48: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 45

Additionally, we investigated whether mucus production was induced in the lung of mite-

infested mice by staining lung sections with periodic acid Schiff (PAS). Accumulation of

mucus was never observed in the lungs of mice at various time points post mite infestation

(data not shown), indicating absence of lung inflammation in this model.

Induction of mast cell degranulation in the skin as well as infiltration of mast cells in skin-

draining LN, in agreement with the observations of IL-4 production and B cell switch to

IgE, provide strong evidence that after mite infestation antigen uptake takes place in the

skin and an immune response is induced in skin-draining LN.

Page 49: The Interplay of IgE and its High Affinity Receptor FceRI

46 Results Part I

2.5 Discussion

In the present study we showed that murine ectoparasite infestation can serve as a valuable

model to investigate allergic disorders. We characterized the immune response against

Myocoptes musculinus and Myobia musculi, two fur mite species naturally occurring in

mice. Our results demonstrate that mite infestation induces a strong Th2-type response

resulting in persistent high serum IgE levels. The fact that mites cause a chronic IgE-

associated response is of particular interest because of its resemblance with chronically

induced atopic reactions in humans that cannot be easily reproduced by common allergy

models based on peptide immunizations.

In this report we provide evidence that induction of the immune response against fur mites,

involving IL-4 secretion by CD4+ T cell and B cell switching to IgE, takes place in LN

draining mite-infested skin. Our results thus suggest that mite antigen is taken up via the

skin. Although other antigen entry sites, such as ingestion through the gut or inhalation

through the lung, may also be possible, the absence of marked signs of inflammation in

these organs indicates that priming of the allergic responses occurs in other tissues.

Upregulation of the expression of sGLT and switched IgE transcripts during the course of

mite infestation was explicitly shown in B cells isolated from skin-draining LN. Some

increase of sGLT and switched IgE transcription was also observed in the mesenteric LN,

however, this increase was only detected on day 15 after mite infestation, while in skin-

draining LN the upregulation of sGLT and IgE expression was maintained until at least day

35. The induction of high serum IgE levels observed between 15 and 60 days post mite

infestation correlates therefore with the class switch recombination to IgE in skin-draining

LN. This provides additional evidence for antigen entry to occur via the skin.

As no mite antigen has been identified thus far, we could not directly prove that any of the

IgE produced in response to mite infestation was antigen-specific. However, highly

increased mast cell degranulation in skin of infested mice strongly suggests that at least part

of the total IgE induced was mite antigen specific. This view is supported by a previous

report in which passive and active cutaneous anaphylaxis as well as mast cell degranulation

in the skin have been demonstrated in response to mite extract (Laltoo et al., 1979).

Nevertheless, a large component of the mite-mediated IgE might be antigen non-specific as

sequence analysis of the mature IgE transcripts suggested a polyclonal response to fur mite

Page 50: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 47

infestation. Interestingly, the analyzed IgE was not highly mutated from germline

sequences, suggesting that the mite-induced IgE (whether specific or non-specific) had not

undergone somatic hypermution. Analysis of additional sequences should provide detailed

insight into the nature of the IgE repertoire induced by mites.

In recent years, fur mites have been investigated in the context of atopic dermatitis in

Nishiki Cinnamon (NC) mice. The Japanese NC mice were initially believed to

spontaneously develop dermatitis-like syndromes accompanied by high serum IgE levels

and were therefore suggested as a good model to study human atopic dermatitis (AD)

(Matsuda et al., 1997). Later however, the fur mites Myocoptes musculinus and Myobia

musculi were reported to be the cause of skin lesions and increased IgE production in NC

mice (Iijima et al., 2000; Morita et al., 1999). Mite antigen-specific IgE was demonstrated

in the sera of mite-infested NC mice by histamine release from bone marrow-derived mast

cells stimulated with mite extract (Morita et al., 1999). Additionally, histochemical

examination of mite-induced skin lesions has revealed significantly increased numbers of

mast cells, eosinophils, and T cells as well as the presence of IL-4 (Matsuda et al., 1997).

This suggests that IL-4 released from skin CD4+ T cells and mast cells may be involved in

the pathogenesis of the AD-like lesions occurring in mite-infested NC mice. However, the

mechanism regulating the development of mite-dependent dermatitis has not yet been

clarified. Genetic background has been previously accounted for different susceptibilities to

mite-associated ulcerative dermatitis (MAUD) (Dawson et al., 1986; Friedman and

Weisbroth, 1975). The NC strain appears to be particularly susceptible showing rapid

development of severe skin lesions (at -12 weeks of age) (Matsuda et al., 1997). In the

present study, we investigated mite infestation mainly on C57BL/6 mice. MAUD

symptoms were observed in about 5% of these mice when mite infestation was maintained

for at least one year, which is comparable to previously published data (Dawson et al.,

1986). The choice of an adequate mouse strain seems therefore to be of major importance

depending on the questions investigated with the mite infestation model. NC mice, prone to

readily develop skin lesions, may be most appropriate to study atopic dermatitis. In the

present report however, C57BL/6 mice were chosen as a suitable strain to investigate the

mechanisms required for the induction and regulation of mite-dependent IgE.

The physiological environment of fur mite infestation makes this model highly suitable for

the investigation of initial events inducing IgE production and IgE-mediated

Page 51: The Interplay of IgE and its High Affinity Receptor FceRI

48 Results Part I

pathophysiology leading to allergic disease. The way of antigen exposure resulting from

mite infestation resembles natural allergen sensitization responsible for atopy in humans.

Noteworthy, the importance of house dust mites in the development of human atopic

dermatitis has been suggested by several studies (Cameron, 1997; Tan et al., 1996). House

dust mites are known to trigger airway responses which led to the hypothesis that the

respiratory route may be relevant for the induction and exacerbation of dermatitis after

house dust mite inhalation in patients with high IgE levels and a history of asthma. In the

fur mite model antigens appear to enter the host through the skin and may directly trigger

dermatitis-like responses.

Histochemical analysis of organ sections isolated from mite-infested mice revealed both

mite-dependent mast cell degranulation in the skin and accumulation of large numbers of

mast cells in skin-draining LN. Mast cell degranulation in the skin is likely due to mite

antigen-induced crosslinking of IgE bound to the high affinity IgE receptor (FcsRI). The

effects of inflammatory mediators released by mast cell degranulation are known to induce

symptoms of allergy and may eventually contribute to the dermatitis-like lesions occurring

in mite-infested mice. However, the role of mast cell accumulation in skin-draining LN has

remained controversial. Mite-induced mast cell infiltrations have been previously reported

by Jungmann et al. who speculated that mast cells might recirculate into the lymphatics and

contribute to antigen transport and presentation in draining LN (Jungmann et al., 1996b).

Several studies have shown that mast cells can enhance the development of T cell-

associated responses. For example, mast cells have been shown to be able to present

antigen in vitro through MHC class I- and class Il-dependent pathways (Fox et al., 1994;

Frandji et al., 1993; Malaviya et al., 1996). Moreover, mast cells have been demonstrated to

express a number of molecules that may allow them to interact with lymphocytes. These

molecules include the costimulatory molecules CD40L (Gauchat et al., 1993) and

CD80/CD86 (Frandji et al., 1996) and the accessory molecules intercellular adhesion

molecule (ICAM)-l (Valent et al., 1991), ICAM-3 (Babina et al., 1999), and lymphocyte

function-associated antigen (LFA)-l (Toru et al., 1997). Thus, mast cells may contribute to

B cell switching to IgE by inducing IL-4- and CD40L-mediated pathways. Interestingly,

mast cell migration to LN has also been demonstrated during dinitrofluorobenzene

(DNFB)-induced contact hypersensitivity in mice (Wang et al., 1998). Wang et al. have

suggested that mast cells may contribute to the induction of primary immune responses by

Page 52: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part I 49

migration from the site of antigen encounter to draining lymph nodes, where they may

promote T cell recruitment by secretion of the chemokine macrophage inflammatory

protein (MlP)-lß. In fur mite-infested mice mast cell infiltration was observed exclusively

in skin-draining LN. The absence of mast cell accumulation in any other organs suggests

the involvement of mite antigens taken up through the skin leading to activation of immune

responses in the skin-draining LN. Further investigation is required to evaluate the

significance of mast cell accumulation in the skin-draining LN and its possible involvement

in mite-induced responses.

Great increase in the prevalence in allergic diseases has been observed over the past two

decades and has been recognized to positively correlate with westernized lifestyle (1998).

Ample epidemiologic data has suggested that allergy may to some extent result from

diminished infectious burden in childhood (Cookson, 1999). The mechanism behind this

hypothesis has recently been proposed to involve anti-inflammatory cytokines, such as IL¬

IO or TGF-ß, induced by exposure to pathogens and responsible for suppression of allergic

and autoimmune diseases (Wills-Karp et al., 2001). The fur mite infestation model

presented in this report could serve to evaluate the regulatory theory of protection against

atopic reactions. Mite infestation could be investigated following exposure to viral or

bacterial pathogens to study their effect in naturally occurring IgE-mediated immune

responses. Thus, the mite allergy model could provide insights into factors regulating the

development of atopic disease.

Page 53: The Interplay of IgE and its High Affinity Receptor FceRI

50 Results Part I

Page 54: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 51

3 Results Part II

Generation and characterization of hmFcsRIaß Tg

mice

3.1 Abstract

Major differences in the expression pattern of the high affinity IgE receptor, FcsRI, exist

between humans and rodents. While the tetrameric form of the receptor (aßy2) is expressed

on mast cells and basophils of both mice and humans, the trimeric complex (0,72) has only

been found to be expressed in humans and is present on eosinophils, dendritic cells,

Langerhans' cells, monocytes, macrophages, and platelets. To elucidate the role of FcsRI

expression on these multiple potential effector and antigen presenting cells, we generated

two transgenic (Tg) mouse models that mimic the human situation. The hmFcsRIa Tg

mouse expresses the murine a chain of the receptor under control of the human FcsRIa

promoter, while the hmFcsRIß Tg mouse expresses the murine ß chain under control of the

human FcsRIa promoter. hmFcsRIaß double Tg mice were generated by crossing the two

Tg lines together. mRNA expression of the transgenes was successfully demonstrated by

RT-PCR and Northern blot, which revealed the intended distribution of all three chains of

the high affinity IgE receptor on same cells as found in humans. Nevertheless, FcsRI

protein expression, investigated by flow cytometry, Western blot, and

immunohistochemistry, could not be found in the targeted cells of the hmFcsRIaß Tg mice.

Thus, the generated mouse model requires further investigation to determine the reason of

the absence of FcsRI protein expression and to evaluate whether this drawback can be

corrected.

Page 55: The Interplay of IgE and its High Affinity Receptor FceRI

52 Results Part II

3.2 Introduction

The high affinity receptor for IgE, FcsRI, is an essential component of IgE-mediated

diseases, such as allergy and asthma. After specific receptor crosslinking via IgE/antigen

interactions, FcsRI signal transduction leads to cell degranulation and release of pro¬

inflammatory mediators. In both humans and mice FcsRI is expressed on mast cells and

basophils as a tetramer composed of one a, one ß, and two y chains (aßy2) (Perez-Montfort

et al., 1983). The extracellular domain of the a chain, containing two Ig-like domains, is

responsible for binding to IgE (Blank et al., 1991). Although both the ß and y chains

contain immunoreceptor tyrosine-based activation motifs (ITAMs), ß is believed to act

merely as an amplifier of signals generated by y (Jouvin et al., 1994; Lin et al., 1996).

Additionally, the ß chain has been demonstrated to have a second amplifier function by

stabilizing the receptor complex on the cell surface and facilitating maturation of the a

chain (Donnadieu et al., 2000). In rodents, the ß chain has been reported to be

indispensable for efficient surface expression of the receptor (Blank et al., 1989; Ra et al.,

1989). In contrast, the human FcsRI can also be found in a trimeric structure, composed

only of one a and two y chains (ay2), expressed on Langerhans' cells (Bieber et al., 1992;

Wang et al., 1992), monocytes (Maurer et al., 1994), eosinophils (Gounni et al., 1994),

peripheral blood dendritic cells (Maurer et al., 1996), and platelets (Joseph et al., 1997).

The discovery of FcsRI expression on cells other than mast cells and basophils raised many

questions regarding the role of the high affinity IgE receptor, in particular concerning its

function in antigen presentation (Novak et al., 2003a; von Bubnoff et al., 2001). However,

the lack of FcsRI expression on antigen presenting cells in mice makes it difficult to use

murine models for investigation of the biological relevance of FcsRI-mediated antigen

presentation. Thus, a "FcsRI-humanized" mouse model was generated with the aim of

assessing the functions of FcsRI on antigen presenting cells in a situation more resembling

humans (Dombrowicz et al., 1996; Dombrowicz et al., 1998). In these human FcsRIa

transgenic mice (hFcsRIa Tg) the endogenous gene encoding the a chain of the FcsRI was

replaced with its human homologue. The human FcsRIa promoter used in hFcsRIa Tg

mice controlled expression of the transgene such that it retained the cell specificity and

structure as found in humans. However, the presence of the human FcsRIa protein in these

Page 56: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 53

mice complicates assessment of the physiological role of the trimeric FcsRI because

binding of mouse IgE to the human a chain is characterized by lower affinity than that of

human IgE (Pruzansky and Patterson, 1986). Thus, our aim was to generate a different

FcsRI Tg mouse model, where the murine FcsRIa was expressed under control of the

human FcsRIa promoter, such that in the resulting hmFcsRIa Tg mice the murine high

affinity IgE receptor should be expressed with the same cell distribution as observed for the

human FcsRI. Such a transgenic animal model should allow investigation of the role of

FcsRI expression in host defense or development of allergic diseases using in vivo murine

assays.

Page 57: The Interplay of IgE and its High Affinity Receptor FceRI

54 Results Part II

3.3 Materials and Methods

Mice

C57BL/6 mice were bred and maintained at the Institute of Laboratory Animal Science of

the University of Zurich. FcsRIa"" mice (Dombrowicz et al., 1993) were kindly provided

by Jean-Pierre Kinet (Boston, USA). BALB/c mice were purchased from Harlan

(Netherlands).

Generation of the transgenic mice

hmFcsRIa Tg mice:

The pGL2 expression vector containing the 2.9 kb sequence of the human FcsRIa

promoter, subcloned into Sma I and Xho I restriction digestion sites, was kindly provided

by Jean-Pierre Kinet (Boston, USA). The pGEM-7 expression vector containing the

palH6.8 sequence coding for all 5 exons of the murine FcsRIa genomic DNA, subcloned

into Hind III restriction digestion sites, was kindly provided by William E. Paul (Bethesda,

USA). A Xho I restriction site was inserted immediately upstream of the ATG codon of the

palH6.8 sequence to allow cloning of the murine FcsRIa genomic DNA into the pGL2

vector downstream of the 2.9 kb human FcsRIa promoter. The correct cloning of the

resulting pGL2 vector (14.8 kb) containing the 2.9 kb human FcsRIa promoter followed by

the 6.3 kb murine FcsRIa sequence was verified by sequencing. Thereafter, the vector was

digested with Sma I and Nar I resulting in excision of a 9.2 kb sequence coding for the

human FcsRIa promoter and the murine FcsRIa. This sequence was microinjected into

male pronuclei of fertilized C57BL/6 oocytes at the Institute of Laboratory Animal Science

of the University of Zurich.

hmFcsRIß Tg mice:

The complete cDNA sequence of the murine FcsRIß sequence was kindly provided by

Chisei Ra (Tokyo, Japan) and was subcloned into the pGL2-phFcsRIa-mFcsRIa vector

replacing the mFcsRIa sequence. Microinjection was performed as described above.

Page 58: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 55

Bone marrow-derived mast cell (BMMC) culture

Femur and tibia bones were collected from C57BL/6 mice and bone marrow cells were

flushed out with BSS. Cells were washed and resuspended at an initial concentration of 106

cells/ml in FMDM containing 10% FCS, 50 uM 2-Mercaptoethanol, 20 ng/ml recombinant

mouse stem cell factor (SCF, R&D Systems), and 5% X63-IL-3 cell-conditioned medium

corresponding to about 5 ng/ml IL-3. Cells were maintained in tissue culture petri dishes at

20 ml/dish in a humidified incubator at 37°C with 5% CO2. Non-adherent cells were

initially transferred to new dishes every other day, later once a week and subsequently

expanded as necessary. Fresh SCF and IL-3 were provided weekly. 6 week cultures

contained >80% mast cells as confirmed by FACS analysis of CD117 expression.

Cell isolation

Macrophages

Peritoneal macrophages were induced by injection of 1 ml of 5% thioglycollate i.p.. The

peritoneal cavity was washed with 10 ml ice-cold BSS 72 hours post thioglycollate

injection. Macrophages were purified by MACS (Miltenyi Biotec) using anti-CD lib

microbeads.

Dendritic cells

To obtain single cell suspensions, spleens or lymph nodes were smashed on metal or nylon

grids, respectively. Dendritic cells were then purified by MACS (Miltenyi Biotec) using

anti-CD 1 lc microbeads.

Langerhans'

cells

Epidermal cells were isolated as previously described (Schüler and Steinman, 1985).

Briefly, ears were collected, split and digested with trypsin at 37°C (dorsal halves in 0.6%

trypsin for 20 min and ventral halves in 1% trypsin for 45 min). Epidermal sheets were

peeled from the underlying dermis and epidermal cells were collected into HBSS

containing 10% FCS. Langerhans' cells were purified from the epidermal cell suspension

by MACS (Miltenyi Biotec) using anti-MHC II microbeads.

Page 59: The Interplay of IgE and its High Affinity Receptor FceRI

56 Results Part II

Eosinophils

Bronchoalveolar lavage (BAL) was performed on Nippostrongylus brasiliensis-mfected

mice on day 14 post infection by inserting a blunted 18G plastic needle into the trachea and

flushing the lung three times with 1 ml ice-cold PBS. The recovered cells were analyzed

directly without any further purifying step because of low cell yields.

RT-PCR

Total RNA was isolated from about 5xl06 cells using TRIzol reagent (Invitrogen Life

Technologies) according to the manufacturer's instructions. First-strand cDNA synthesis

was performed with random hexamers and SuperScript II reverse transcriptase (Invitrogen

Life Technologies). Transcripts encoding mFcsRI were amplified using the following

primers and conditions: a chain (467 bp band) 5'-CGATGGTCACTGGAAGGTCTGC-3'

and 5'-ATAACTGTAGTTGAAAGCATGG-3' (94°5'; [94°1'; 54°30"; 72°l']x35; 72°10';

10°), ß chain (306 bp band) 5'-AACATTGTCAGTAGCATCGC-3' and 5'-

CCCTTTGTCTTCCAACTCAC-3' (94°5'; [94°1'; 58°30"; 72°l']x35; 72°10'; 10°), and y

chain (402 bp band) 5'-CAGCCGTGATCTTGTTCTTGC-3' and 5'-

TTAACGGAGATGGGGACCTGC-3' (94°5'; [94°1'; 58°30"; 72°l']x35; 72°10'; 10°).

Northern blot

Total RNA was isolated from 107 cells using TRIzol reagent (Invitrogen Life Technologies)

according to the manufacturer's instructions. Equal amounts of RNA (5 ug/lane) were

separated on a 1.5% agarose-formaldehyde gel. The RNA was transferred onto a Hybond-

N+ nucleic acid transfer membrane (Amersham Biosciences) using the Turbo Blotter

system (Schleicher&Schuell). The specific mRNA was detected by hybridization of the

membrane with a 32P-labeled RNA probe specific for the murine FcsRIa gene. The DNA

template for the riboprobe was generated by cloning a 630 bp FcsRIa RT-PCR product

(primers: FcsRIaNhe 5'-CTAGCTAGCATGGTCACTGGAAGGTCTG-3' and

FcsRIaXba 5'-GCTCTAGAAGTTGTAGCCAATAATACTTGC-3', conditions: 94°5';

[94°1'; 54°30"; 72°l']x35; 72°10'; 10°) into the pGEM-T vector (Promega) and

subsequently linearizing the vector by Nde I restriction digestion. The probe was

synthesized by incubating 1 ug of the DNA template for 3 h at 37°C in presence of 50 uCi

[a32P]UTP (800 Ci/mmol, Amersham Bioscience), 1 U T7 polymerase, and 10 uM

Page 60: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 57

Ribonuclease Triphosphate set (rATP, rCTP, and rGTP) in a total volume of 25 ul of T7

buffer (Roche). The RNA-containing membrane was prehybridized for 1 h at 68°C in

presence of the ULTRAhyb hybridization buffer (Ambion). 5-15xl06 cpm of the probe was

then added and hybridized overnight at 68°C. The membrane was washed twice in 2xSSC

and twice in 0.2xSSC, both containing 0.2% SDS, for 15 min at 60°C and exposed

overnight to a BIOMAX MR film (Kodak).

Western blot

Cells were lysed at 5xl07 cells/ml in lysis buffer containing 1% digitonin, 150 mM NaCl,

50 mM Hepes, and complete protease inhibitor cocktail (one tablet per 50 ml lysis buffer,

Roche). 500 ul lysate was immunoprecipitated with biotinylated anti-FcsRIa antibody (10

ug/ml, eBioscience) for 2 h on a helicopter rotor at 4°C. Streptavidin-conjugated sepharose

(Amersham Biosciences) was added and incubated for an additional 2 h on a helicopter

rotor at 4°C. The sepharose beads were then washed 3x with PBS and samples denaturated

by incubating for 5 min at 95°C in SDS reducing loading buffer. After centrifugation (3

min, 13000 RPM) proteins were separated by running on a 12.5% SDS-polyacryamide Tris

gel (Bio-Rad Laboratories) and then electrotransferred to Protran nitrocellulose membranes

(Schleicher&Schuell) using a semi-dry transfer unit (SemiPhor Hoefer). Membranes were

blocked in 4% milk/PBS overnight at 4°C. Detection of FcsRIß was performed with mouse

anti-mouse FcsRIß antibody (Rivera et al., 1988) (1 ug/ml, clone JRK, kindly provided by

Juan Rivera, Bethesda, USA) followed by goat anti-mouse IgG antibody (1 ug/ml, EY

Laboratories). Detection of FcsRIy was performed with rabbit anti-mouse FcsRIy antibody

(1 ug/ml, Upstate Biotech) followed by goat anti-rabbit IgG (1 ug/ml, Sigma) antibody. All

antibodies were diluted in 1% milk/PBS and incubated with the membranes for 2 h. For

both ß and y chains HRPO-conjugated donkey anti-goat IgG (1 ug/ml, Abeam) was used as

tertiary antibody and incubated with the membranes for 1 h. Revelation was performed

using the SuperSignal West Pico Chemiluminescent Substrate (Pierce) according to the

manufacturer's instructions.

Flow cytometry

Purified cells were stained using standard procedures with the following antibodies: anti-

mouse CD117-APC (0.7 ug/ml, BD Pharmingen), anti-mouse CDllb-PE (0.4 ug/ml, BD

Page 61: The Interplay of IgE and its High Affinity Receptor FceRI

58 Results Part II

Pharmingen), anti-mouse CDllc-PE (0.7 ug/ml, BD Pharmingen), anti-mouse I-Ab-PE (0.7

ug/ml, BD Pharmingen), anti-mouse GR-l-PE (2 ug/ml, BD Pharmingen), anti-mouse

FcsRIa-FITC (15 ug/ml, eBiosciences), and FITC-labeled Golden Syrian Hamster isotype

control (15 ug/ml, eBiosciences). For intracellular staining cells were fixed with 2%

paraformaldehyde and permeabilized in 0.1% saponin. Cells were acquired in a FACS

Calibur (BD) and analyzed using FlowJo software.

Immunofluorescence

Cytospins of 105 cells were performed in a Shandon Southern centrifuge. Cells were fixed

in acetone, air-dried, and rehydrated in PBS immediately prior to staining. Anti-mouse

CD117-APC (2 ug/ml, BD Pharmingen) or anti-mouse FcsRIa-biotin (10 ug/ml,

eBiosciences) antibodies were applied onto the fixed cells and incubated for 45 min at room

temperature (RT). When staining for FcsRIa, streptavidin-Cy3 (1.3 ug/ml, Jackson

Immunoresearch) was applied for 30 min at RT. Nuclei were stained with DAPI (1 ug/ml,

Sigma) for 5 min at RT. After each staining step slides were washed 3x in PBS. Slide

covers were mounted using DakoCytomation fluorescent mounting medium. Fluorescence

was monitored on a Zeiss Axiophot microscope with a JVC KYF70 camera, using the

Analysis software (Soft Imaging System, Münster, Germany).

Nippostrongylus brasiliensis infection

Parasite maintenance and injection

N brasiliensis was maintained by serial passage through 6 week-old Lewis rats. To

establish rat infections, 5000 infective L3 stage larvae were resuspended in 0.5 ml H20 and

injected s.c. Rat fecal pellets were collected daily between day 7 and 11 post infection and

cultured with granular charcoal at 27°C for 14-28 days. Exsheathed L3 stage larvae were

harvested and resuspended in H20. Mice were injected s.c. with 550 L3 larvae in 200 ul

H20.

Determination ofparasite egg numbers

Infected mice were transferred to gridded cages lined with moist paper and fecal pellets

were collected daily between day 5 and 11 post iV. brasiliensis infection. Pellets were

resuspended in equal volumes of H20 and saturated NaCl solution, vortexed and a sample

Page 62: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 59

quickly removed and transferred to a McMaster Worm Egg Counting Chamber (Weber

Scientific International Ltd.) for quantification. The number eggs per gram feces was

calculated knowing that the counting chamber holds 150 ul volume and that 25 mouse fecal

pellets correspond to 1 gram.

Determination ofperipheral blood eosinophilia

Blood smears were prepared from a drop of freshly collected blood, air-dried, and stained

with Diff-Quik (Dade Behring) for differential cell counts of lymphocytes, macrophages,

neutrophils, and eosinophils. A total of 200 cells were counted and the percentage of

eosinophils determined.

Determination oftotal serum IgE levels

Total serum IgE concentrations were determined by enzyme-linked immunosorbent assay

(ELISA). ELISA plates (Greiner Microion) were coated with 5 ug/ml rat anti-mouse IgE

(clone 6HD5) in 0.1 M NaHC03 (pH 9.6) and blocked with 5% (w/v) bovine serum

albumin (BSA). Serially diluted serum or standards (IgE clone TIB-141) were added and

incubated for 2 hours. Bound IgE was detected with biotinylated anti-mouse IgE (clone

RIE-4) at 2.5 ug/ml followed by horseradish peroxidase-conjugated streptdavidin (Jackson

ImmunoResearch Laboratories) at 1 ug/ml. ABTS peroxidase solution (2,2'-azino-bis-[3-

ethylbenzthiazidine-6-sulfonic acid] 0.1 mg/ml, 0.05% H202, and 0.1 M NaH2P04 pH 4.0)

was used for the peroxidase-catalyzed color reaction. Color reaction was read at 405 nm in

a Bio-Rad microplate reader and standard curves and IgE concentrations analyzed with

Microplate Manger III software (Bio-Rad).

Page 63: The Interplay of IgE and its High Affinity Receptor FceRI

60 Results Part II

3.4 Results

Generation of hmFcsRIaß Tg mice

The strategy used to generate mice with FcsRI expression patterns comparable to those

found in humans was based on the results obtained by Dombrowicz et al. (Dombrowicz et

al., 1996). Thus, to obtain the hmFcsRIa Tg mouse, the 2.9 kb promoter region of the

human FcsRIa encoding the information necessary for cell-specific expression was

subcloned into the pGL2 expression vector. The murine FcsRIa genomic DNA fragment of

6.3 kb was then subcloned downstream of the promoter into the Xho I and Hind III

restriction digestion sites (Figure IIA). For microinjection into C57BL/6 oocytes the

promoter and gene were excised from the vector by Sma I and Nar I restriction digestion.

Integration and germline transmission of the transgene were monitored by PCR and

Southern blot analysis (data not shown).

Expression of the human FcsRIa chain in the previously described hFcsRIa Tg mice

(Dombrowicz et al., 1996; Dombrowicz et al., 1998) was reported to be independent of the

presence of the ß chain and trimeric ay2 complexes were identified on peritoneal

macrophages isolated from hFcsRIa Tg mice. However, in the present approach we used

the murine FcsRIa chain, which has been demonstrated to require the presence of the ß

chain for correct surface expression of FcsRI (Blank et al., 1989; Ra et al., 1989). Thus, in

order to ensure proper expression, in addition to the hmFcsRIa Tg mouse we generated an

hmFcsRIß Tg mouse, which should express the ß chain of the murine FcsRI with the same

cellular distribution characteristics as found in humans. The transgene construct for the

hmFcsRIß Tg mouse was generated by replacing the mFcsRIa gene in the pGL2-

phFcsRIa-mFcsRIa with the mFcsRIß sequence (Figure 1 IB). Lastly, the two generated

Tg strains, hmFcsRIa and hmFcsRIß, were crossed to obtain double transgenic

hmFcsRIaß mice. The phenotype of the double transgenic mice was analyzed by RT-PCR,

Northern blot, Western blot, FACS analysis, and immunohistofluorescence as described

below. Additionally, the immune response of hmFcsRIaß mice to Nippostrongylus

brasiliensis infection was assessed.

Page 64: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 61

A

PolfA Sifnit

Sma I (4)B

3OTR

intron f

Nar I (9229)

Hind 111 (9167)

hFct-RIa

promoter

Xho I (2902)

mFceRIa

/

PolfA Signal

3'yTR

Sma I (4)

PGL2-phFceRI»mFccRip

909? Tap

FceRIa

promoter

Xho I (2902)

tnFcvlRlR1111 %^c i %i ^.j

Nar I (9229)

Figure 11. Transgene constructs for hmFceRlot and hmFceRIß Tg mice. The 2.9 kb human

FceRIa promoter was subcloned into the Sma I and Xho I restriction sites of the pGL2 expression

vector. For the hmFcsRIa mouse, the 6.3 kb murine FceRIa genomic DNA was subcloned

downstream of the promoter into the Xho I and Hind III restriction sites (A). For the hmFceRIß

mouse, the 0.7 kb murine FceRIß cDNA was subcloned downstream of the promoter into the Xho I

and Nar I restriction sites (B). Both vectors were digested with Sma I and Nar I to excise the

transgene, which was then used for microinjections into male pronuclei of fertilized C57BL/6

oocytes to generate hmFceRIa and hmFceRIß Tg mice.

Page 65: The Interplay of IgE and its High Affinity Receptor FceRI

62 Results Part II

RT-PCR analysis of FcsRI expression in hmFcsRIaß Tg mice

In order to investigate mRNA expression of FcsRI in different cell types of hmFcsRIa and

hmFcsRIaß Tg mice, RT-PCR primers were generated for each of the chains of the

receptor. The primers were tested on the MC/9 mast cell line as well as on bone marrow-

derived mast cells (BMMC) of WT C57BL/6, hmFcsRIa Tg, and FcsRIa-deficient mice

(Figure 12A). All mast cell samples expressed all three chains of FcsRI (a, ß, and y) except

BMMC from FcsRIa""

mice (Dombrowicz et al., 1993) in which FcsRIa was not detected.

As expected, peritoneal macrophages of C57BL/6 mice, used as negative control, expressed

only the y chain of the FcsRI, which is known to be ubiquitously expressed.

Analysis of hmFcsRIa Tg mice revealed mRNA expression of the a chain of the high

affinity IgE receptor in macrophages, dendritic cells, eosinophils, and Langerhans' cells,

while no such expression could be detected in WT C57BL/6 mice (Figure 12B). The

quality of the cDNA was controlled by the presence of y chain transcripts in all samples.

mRNA expression of both FcsRIa and FcsRIß was confirmed in macrophages and

dendritic cells of the double transgenic hmFcsRIaß mice (Figure 12C). We have not yet

confirmed expression of FcsRIa or FcsRIß in Langerhans' cells and eosinophils due to the

large numbers of cells required for RNA isolation coupled with limited availability of the

double transgenic mice. Surprisingly, we also detected FcsRIa mRNA expression in

splenic B cells isolated from hmFcsRIa Tg mice after infection with N brasiliensis and in

splenic B cells of naive double transgenic mice but at very low levels (data not shown).

Further investigation is required to confirm expression of the transgenic high affinity IgE

receptor in B cells. As expected, investigation of T cells isolated from both hmFcsRIa and

hmFcsRIaß Tg mice did not reveal any FcsRIa mRNA expression (data not shown).

Taken together, these data indicate that mRNA expression of the high affinity receptor was

successfully achieved on the targeted cells of the hmFcsRIaß Tg mice.

Page 66: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 63

BMMC

500—1

500—1

L MC/9 B6 aTg a-'- ft» »P

FceRIa

467 bp

FceRIß"*~

306 bp

500-1m

FceRIy407 bp

B

bp l

500-

M<î>

B6 aTg

SpIDC

B6 aTg

LNDC

B6 aTg

BAL eos

B6 aTg

LC

B6 aTg

BMMC

B6 aTg

Hfi

500-

FceRIa

"467bp

FceRIy407 bp

bp

500-

500 —

500-

B6 aß

SpIDC

B6 aß

LNDC

B6 aß

BMMC

B6 aß

Hfi

FceRIa

487 bp

FceRIß306 bp

FceRIy407 bp

Figure 12. mRNA expression of the individual chains of FceRI in hmFceRIa and hmFcsRIaß

Tg mice. RT-PCR for detection of the a, ß, and y chains of FceRI was performed on mast cells

(MC/9 line and BMMC), thioglycollate-induced peritoneal macrophages (MO), naïve splenic

dendritic cells (spl DC), DC from lymph nodes (LN DC), eosinophils (eos) from the BAL of N.

Jbras/7/'ens/'s-infected mice, and naïve Langerhans' cells (LC). Cells were isolated from WT C57BL/6

(B6), FceRIa"'" (a"/_), hmFceRIa Tg (aTg), or hmFceRlaß Tg (aß) mice. Controls (A) as well as

analysis of hmFceRIa (B) and hmFceRlaß (C) Tg mice are depicted. H20 was used as negative

control. The size of each PCR product is indicated. The 100 bp Ladder (L) was used for the size

marker and the 500 bp band is indicated. Results are representative of two to four separate

experiments.

Page 67: The Interplay of IgE and its High Affinity Receptor FceRI

64 Results Part II

Northern blot analysis of FcsRI expression in hmFcsRIaß Tg mice

To provide a quantitative method to investigate mRNA expression of the high affinity IgE

receptor, Northern blot analysis for FcsRIa expression was developed. Initially, Northern

blot analysis was used to quantify FcsRIa expression levels in different hmFcsRIa Tg lines

derived from founders 2, 3, and 14 (Figure 13A). The Tg line with the highest FcsRIa

expression was identified (founder 3) and this line was then crossed with hmFcsRIß Tg

mice to generate double transgenic mice.

Northern blot analysis was also performed in order to confirm the RT-PCR results showing

that the murine a chain gene was expressed in hmFcsRIa and hmFcsRIaß Tg mice in the

same cells as found in humans. mRNA of the a chain of the receptor was detected in

splenic dendritic cells from both hmFcsRIa and hmFcsRIaß Tg mice, as indicated by the

presence of a 1 kb band corresponding to the 1005 bp of the murine FcsRIa mRNA (Ra et

al., 1989) (Figure 13B). 18 S rRNA bands were also observed due to unspecific binding of

the probe and could be eliminated by more stringent washing (data not shown). However,

as they served as a convenient internal control for the amount of loaded RNA they were not

routinely washed away. Contrary to what was observed by RT-PCR no expression of

FcsRIa was observed in macrophages of either single or double Tg mice. The lack of

detection by Northern blot analysis is likely to be due to sensitivity of this assay. It is

possible that the levels of FcsRIa expression in macrophages may be lower than in

dendritic cells and therefore detectable only by RT-PCR and not by Northern blot. As

Northern blot analysis requires large cell numbers to obtain sufficient RNA, investigation

of other cell types, such as Langerhans' cells and eosinophils, could not be performed due

to limited availability of the transgenic mice.

Page 68: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 65

SpIDC BMMC

2a 2b 14a 14b B6 B6

Kb

1.77«

1.28-

18 S rRNA

FceRIa

1005 bp

B

B6

M4>

aTg aßTg

SpIDC

B6 aTg aßTg

Kb

1.77«

1.28-

18 S rRNA

FceRIa

1005 bp

Figure 13. Northern blot analysis of FceRIa expression. FceRIa expression levels in splenic

dendritic cells (spl DC) purified from hmFceRIa Tg lines derived from founders 2, 3, and 14 were

investigated using a FceRla-specific radioactive probe (A). Thioglycollate-induced peritoneal

macrophages (MO) and naïve spl DC purified from WT C57BL/6 (B6), hmFceRIa Tg (aTg), or

hmFceRlaß Tg (aßTg) mice were equally analyzed (B). BMMC from WT C57BL/6 mice were used

as positive control. The 18 S rRNA bands reflect the amount of loaded RNA. The 0.16-1.77 Kb RNA

Ladder was used as marker and the positions of the 1.77 Kb and 1.28 Kb bands are indicated.

Results are representative of two (A) or four (B) separate experiments.

Page 69: The Interplay of IgE and its High Affinity Receptor FceRI

66 Results Part II

Western blot analysis of FcsRI expression in hmFcsRIaß Tg mice

The results presented thus far confirmed mRNA expression of FcsRI in hmFcsRIaß Tg

mice. We next investigated whether the mRNA was also transcribed into protein in the

different cell types of the transgenic mice. For this purpose a method for Western blot

analysis of the murine high affinity IgE receptor was developed and tested on mast cells. To

assess cytosolic proteins, cells were lysed and lysates were immunoprecipitated with a

monoclonal anti-mouse FcsRIa antibody to subsequently identify proteins that were bound

to and therefore co-immunoprecipitated with FcsRIa. Precipitated proteins were then

analyzed for the presence of FcsRIß and FcsRIy by probing with specific antibodies against

the ß and y chains of FcsRI. This method allowed demonstration of the presence of the ß

and y chains and their association with the a chain. As expected, both the ß (Figure 14A)

and y (Figure 14B) chains were found to be associated with the a chain in the mast cell line

MC/9 as well as in BMMC of WT mice. Importantly, anti-a chain antibody-mediated

immunoprecipitation of BMMC lysates derived from FcsRIa-deficient mice did not reveal

the presence of neither ß nor y chains, indicating the specificity of the co-precipitation of

the ß and y chains. Thus far we did not succeed in detecting the presence of the a chain in

cell lysates by probing membranes containing fractionated proteins with a specific anti-

FcsRIa antibody. This may be due to conformational changes induced in FcsRIa while

running the samples on a reducing SDS-gel or transferring them to nitrocellulose

membranes. Such conformational changes may mask or destroy binding sites of the anti-

FcsRIa antibody and therefore inhibit its binding to the a chain.

Analysis of peritoneal macrophages and splenic dendritic cells derived from either

hmFcsRIa or hmFcsRIaß Tg mice did not reveal co-precipitation of the ß nor the y chain

with the a chain of FcsRI (Figure 14C). Weak detection of a protein corresponding to the

size of the y chain in the macrophage samples was present, as shown by the weak bands in

the lower panel of Figure 14C. However, this protein was also detected in macrophages

isolated from FcsRIa-deficient mice, suggesting that the detected band did not result from

co-immunoprecipitation with the a chain. It is possible that this band reflects non-specific

binding of the anti-y antibody to a protein present in macrophages. Alternatively, this band

may indicate the ability of the y chain from macrophages to non-specifically "stick" to the

sepharose beads used in the immunoprecipitation. Thus, although we could detect mRNA

Page 70: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 67

expression of the subunits composing the high affinity IgE receptor in macrophages and

dendritic cells of the generated transgenic mice, we were not able to show their association

at the protein level. Importantly, this method was based on immunoprecipitation with

FcsRIa leaving it open whether the absence of FcsRI protein detection in the Tg mice was

due to lack of a chain protein expression in the peritoneal macrophages and dendritic cells

of the Tg mice or to the lack of receptor complex formation in these cells. Alternative

approaches may be envisaged to answer this question and possibly demonstrate the

presence of FcsRI complexes in the cells of hmFcsRIa and hmFcsRIaß Tg mice.

Therefore, we are currently developing methods for Western blot analysis where cell

lysates are immunoprecipitated with either the ß or the y chain of FcsRI and fractionated

proteins are subsequently probed with specific antibodies against the a, ß, and y chains of

the receptor.

Taken together, further investigation is required to determine whether translation of the

FcsRI chains from mRNA into protein or their association into receptor complexes was

inhibited in the targeted cells of hmFcsRIa and hmFcsRIaß Tg mice. Furthermore, it is

possible that the expression level of FcsRI in the transgenic mice is below the detection

limit of the Western blot, implying that additional assays are required to confirm the data.

Page 71: The Interplay of IgE and its High Affinity Receptor FceRI

68 Results Part II

B

BMMC

kDa

50 —

37 —

25 —

20 —

15 —

10 —

MC/9

FceRIß"32kD

BMMC

kDa B6 a-'-

50 ¥

37 -- *

#1ï

25 ,

20—f},

-

15 /;""

* J" ''

i

10— A"

MC/9

Vm. 1

FceRIy'9 kDa

MO spl DC

kDa MC/§ B6 a aß or'- B6 a aß a-/_

37

»-• FceRIß", 32 kDa

20"

-

15

il,10—

-%§ iii?-

1 '•,

*, _

1'

37-

25-

20-

15-

10-

'r

- -

§*-itsitf

..* !FceRIy9 kDa

Figure 14. Immunoprecipitation and Western blot of FceRI. Digitonin-solubilized protein extracts

were prepared from the mast cell line MC/9, BMMC, thioglycollate-induced peritoneal macrophages

(MO) or naïve splenic dendritic cells (spl DC) isolated from WT C57BL/6 (B6), hmFceRIa Tg (a),

hmFceRlaß Tg (aß) or FceRla-deficient (a"/_) mice. Immunoprecipitation was carried out with anti-

mFceRIa Ab and co-precipitation of ß and y chain proteins was analyzed by Western blot using

antibodies specific for the mFceRIß and mFceRly. The molecular mass is indicated in Kilodaltons.

Results are representative of four (mast cells) or one (MO and DC) separate experiments.

Page 72: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 69

FACS analysis of FcsRI expression in hmFcsRIaß Tg mice

To further investigate whether hmFcsRIaß Tg mice express FcsRI, flow cytometry

analysis was performed to detect both intra- and extracellular protein expression of FcsRIa.

In contrast to the Western blot analysis, which detects the association of the receptor

subunits, this approach provides direct information on the presence of translated FcsRIa.

Several different cell types were isolated from WT C57BL/6, hmFcsRIa Tg, and

hmFcsRIaß Tg mice and FcsRIa expression was assessed on each cell population. In

contrast to BMMC, which were found to express high amounts of the receptor (Figure 15,

top panel), FcsRIa protein expression was not detected in macrophages, Langerhans' cells

or dendritic cells isolated from naive hmFcsRIa or hmFcsRIaß Tg mice (Figure 15, middle

panel).

A positive correlation between FcsRI cell surface expression and serum IgE concentrations

has been shown previously (Yamaguchi et al., 1997). The mechanisms controlling

upregulation of FcsRI by IgE involve receptor stabilization at the membrane and continued

basal synthesis of receptors (Borkowski et al., 2001). It has also been reported that in

humans FcsRI expression on APCs is greatly upregulated in atopic individuals, which are

characterized by high levels of IgE (Maurer et al., 1994; Novak et al., 2003b). Therefore, it

was possible that the inability to detect expression of FcsRI in APCs isolated from

hmFcsRIa and hmFcsRIaß Tg mice may reflect the lack of significant levels of serum IgE.

Thus, in order to achieve maximal expression of FcsRI we attempted to induce high levels

of serum IgE in the transgenic mice prior to cell isolation. Mice were therefore infected

with the nematode Nippostrongylus brasiliensis, which is known to induce up to 1000-fold

increase in total serum IgE (Finkelman et al., 1988). Cells were then isolated 15 days

following iV. brasiliensis infection when IgE levels had reached a peak of 1769 ng/ml

(±166.7 SEM, n=50). Besides macrophages, dendritic cells, and Langerhans' cells, FcsRIa

expression was additionally investigated in eosinophils isolated from BAL of iV.

brasiliensis-'mfected mice. However, despite the presence of high levels of serum IgE,

FcsRIa expression could not be detected in any of the analyzed cells (Figure 15, lower

panel).

Page 73: The Interplay of IgE and its High Affinity Receptor FceRI

70 Results Part II

BMMC(CD117+)

i 'I

i p.

C57BL/6

FcsRIa-'-

,! I

Isotype control

: ,- \

Spl DC(CD11c+) LN DC (CD11 c+) LC (MHCII+)MO(CD11b+)

fi

f\ |1 C57BL/6

>

'reC

! i

I jil i

l" \

11,i i

? I » \

aTg

aßTg

J i^ 1 \\i \

eos(GR-1+)

c

.0)

'552-Q

i

il 11

\\i Ï

1M

fi

I!li

j

« tI s

S 11 ./ 1^

; \ j\

FcsRIa

Figure 15. Intracellular FACS analysis of FcsRIa expression. BMMC, thioglycollate-induced

peritoneal macrophages (MO), splenic dendritic cells (spl DC), DC from lymph nodes (LN DC),

Langerhans' cells (LC), and eosinophils (eos) from the BAL isolated from either naïve or N.

Jbras;'/;'ens;'s-infected WT C57BL/6 (black), hmFceRIa Tg (aTg, green), or hmFceRlaß Tg (aßTg, red)

mice were analyzed for FceRIa expression by intracellular flow cytometry. Cells were gated for the

specific cell type markers of each cell population as indicated in brackets. Control stains were

performed using BMMC from FceRIa"'" (blue) and with an isotype control antibody (grey). Results

are representative of three (naïve) or two (A/. Jbras;'/;'ens;'s-infected) separate experiments.

These data therefore suggest that although there is expression of the a chain of FcsRI at the

mRNA level, there does not appear to be expression of FcsRIa protein in the analyzed cells

from either hmFcsRIa or hmFcsRIaß Tg mice. Intracellular FACS analysis should allow

detection of both intracellular FcsRIa protein expression and FcsRIa expressed on the cell

surface as part of the receptor complex. Thus, these results could be explained by a

potential inhibition of the translation of FcsRIa in macrophages, dendritic cells,

Langerhans' cells, and eosinophils of the generated transgenic mice. The reason for such

inhibition remains to be determined. Alternatively, it is possible that the a chain is

translated into protein but then retained in the endoplasmic reticulum (ER) and rapidly

Page 74: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 71

degraded, which would hinder its detection by FACS. The retention in the ER could be due

to incomplete interactions of the ß with the a chain, which in mice is required to release

FcsRIa from the ER (Ra et al., 1989). Possible mutations in the transgene could lead to

structural changes of the receptor chains inhibiting their interactions. Thus, sequence

analysis of the transgenic FcsRI may provide further insight into the factors responsible for

the absence of FcsRI protein expression in hmFcsRIaß Tg mice.

Immunofluorescence analysis of FcsRI expression in hmFcsRIaß Tg mice

Although the data from flowcytometry suggested that FcsRIa protein was not expressed in

the targeted cells of hmFcsRIa and hmFcsRIaß Tg mice, we decided to confirm these

results by an additional method. For this purpose cytospins of several cell types isolated

from WT C57BL/6, hmFcsRIa or hmFcsRIaß Tg mice were investigated for FcsRIa

protein expression by immunofluorescence. To begin with the method was tested on

BMMC from WT C57BL/6 and FcsRIa""

mice, which were used as positive and negative

controls, respectively. The results showed clear expression of the mast cell marker CD117

on both cell types, while FcsRIa expression was detected only on WT cells (Figure 16A).

However, no FcsRIa expression could be detected in macrophages, dendritic cells, or

Langerhans' cells isolated from either WT or transgenic mice as shown by fluorescence

comparable to background levels (Figure 16B). As not all cell surface maker antibodies

were available for immunofluorescence analysis, the purity and viability of each cell

sample was confirmed by FACS prior to preparation of the cytospins. Analysis of FcsRIa

expression in cells isolated from iV. brasiliensis-mfected mice equally revealed only

background levels of fluorescence (data not shown), implying that even in presence of high

serum IgE levels no detectable FcsRIa expression was induced.

Taken together, these results are consistent with the FACS data leading to the conclusion

that despite of significant FcsRIa mRNA expression in the targeted cells of hmFcsRIa and

hmFcsRIaß Tg mice no a chain protein could be detected.

Page 75: The Interplay of IgE and its High Affinity Receptor FceRI

72 Results Part II

A CD117 FceRIa DAPI Merge

B I» spl DC LNDC LC

aßTg

Figure 16. Immunofluorescence of cytospins. Cytospins of BMMC isolated from WT C57BL/6

(B6) and FceRIa"'" (a"'") mice were analyzed for CD117 and FceRIa expression using specific

fluorescent antibodies; nuclei were stained with DAPI (A). FceRIa expression was investigated in

thioglycollate-induced peritoneal macrophages (MO), splenic dendritic cells (spl DC), dendritic cells

from lymph lodes (LN DC), and Langerhans' cells (LC) isolated from WT C57BL/6 (B6), hmFceRIa

Tg (aTg) or hmFceRlaß Tg (aßTg) (B). Results are representative of two separate experiments.

Page 76: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 73

Immune response of hmFcsRIaß Tg mice to Nippostrongylus brasiliensis infection

hmFcsRIa and hmFcsRIaß Tg mice were infected with the nematode parasite iV.

brasiliensis to induce high serum levels of IgE and to drive maximal FcsRI expression.

Additionally, the immune response of the transgenic mice to iV. brasiliensis was assessed.

This approach was based on the hypothesis that the presence of FcsRI on the surface of

APCs or eosinophils could provide mechanisms leading to a more efficient immune

response against the parasite. For example, FcsRI-mediated antigen uptake by APCs could

lead to enhanced antigen presentation and therefore increased T cell activation.

Furthermore, activation of eosinophils via FcsRI could improve killing of the parasitic

larvae during their passage through the lung. Thus, the level of protection against iV.

brasiliensis was assessed in hmFcsRIa and hmFcsRIaß Tg mice and compared to that in

WT C57BL/6 mice.

Within 10 days post iV. brasiliensis infection total serum IgE levels increased greatly and

remained high in both WT and transgenic mice (Figure 17A). There was no significant

difference in the kinetics nor magnitude of IgE induction between WT and transgenic mice.

Peripheral blood eosinophil levels were also investigated as N brasiliensis infection is

known to induce blood and tissue eosinophilia, which has been suggested to play a role in

host protection and expulsion of the parasite (Shin et al., 1997). The percentage of

eosinophils in the blood started to increase at day 7 post infection and reached a peak of

about 10% around day 15 (Figure 17B). Comparable results were observed for WT as well

as hmFcsRIa and hmFcsRIaß Tg mice. In WT mice the immune response induced by iV.

brasiliensis leads to expulsion of the worms within two weeks after infection. During that

period infecting larvae develop into the adult stage within the gut lumen and produce eggs

that are expelled via the fecal pellets. The numbers of eggs in the feces are inversely

proportional to the strength of the protective immune response. Fecal worm egg counts

were therefore monitored in iV. brasiliensis-'mfected WT as well as hmFcsRIa and

hmFcsRIaß Tg mice. Comparable worm egg numbers and comparable kinetics of worm

expulsion were found between WT and both Tg mouse strains (Figure 17C). The eggs

found in hmFcsRIaß Tg mice had a slightly delayed onset and were cleared one day

earlier, however, data of several experiments showed no significant difference between the

analyzed mouse strains indicating the they had a comparable degree of protection to iV.

Page 77: The Interplay of IgE and its High Affinity Receptor FceRI

74 Results Part II

brasiliensis. Although our results were variable, especially the range of eosinophilia and

egg counts was very wide, repeated iV. brasiliensis injections demonstrated that the overall

response did not differ between WT, hmFcsRIa, and hmFcsRIaß Tg mice.

A

B

C

5 6 7 8 9 10 11

Days post Nb infection

Figure 17. Nippostrongylus brasiliensis (Nb) infection of hmFceRlaß Tg mice. C57BL/6

(closed circles), hmFceRIa Tg (open triangles), and hmFceRlaß Tg (open squares) mice were

infected with 550 Nb L3 larvae on day 0. Blood was collected on several time points after infection

(as indicated) and total serum IgE levels (A) and blood eosinophilia (B) were determined. The data

points represent the mean + s.d. of 3-5 mice per group. The dotted line in A indicates the lower limit

sensitivity (0.7 ng/ml) of the IgE ELISA. Numbers of Nb eggs per gram feces were monitored for

each mouse strain (C). Results are representative of five separate experiments.

• C57BL/6

A hmFceRIa

D hmFceRlaß

30'000

ra 20'000

5»o>mœ

10'000

Page 78: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 75

It is possible that FcsRI expressed on APCs may only play a role in secondary parasite

infections, where IgE produced during the primary response has already bound to the

receptor. To investigate this hypothesis we performed a booster infection of iV. brasiliensis

ten weeks after the primary infection. Analysis of IgE serum levels during the secondary

immune response did however not reveal any significant differences between WT,

hmFcsRIa, and hmFcsRIaß Tg mice (data not shown).

Page 79: The Interplay of IgE and its High Affinity Receptor FceRI

76 Results Part II

3.5 Discussion

FcsRI transgenic mice were generated to provide a novel tool for the investigation of

various aspects of the biology of the high affinity IgE receptor. Using the human FcsRIa

promoter we generated transgenic mice with the aim of expressing the murine FcsRI on the

same cell types as demonstrated for the human FcsRI. We first generated hmFcsRIa Tg

mice that express the murine a chain of the receptor under control of the human FcsRIa

promoter. Although previously reported transgenic mice generated with the same technique

but using the human a chain of the receptor have been shown to express the ay2-trimeric

receptor complex (Dombrowicz et al., 1998), in the present approach important species

differences between the murine and human FcsRIa chain had to be considered, such as the

different requirements for receptor surface expression. Transfection studies had previously

demonstrated that the murine a chain contains endoplasmic reticulum (ER) retention

signals located in the cytoplasmic tail (Letourneur et al., 1995a). The other chains of the

FcsRI complex appear to prevent retention and degradation of the a chain in the ER by

masking the ER retention signals and allowing export of the receptor to the Golgi apparatus

and subsequently to the plasma membrane. While the y chain has been shown to be

sufficient to mask the ER retention signals of the human a chain, the murine a chain

requires both the ß and y chains to allow receptor release from the ER (Ra et al., 1989).

Therefore, in order to achieve surface expression of FcsRI in hmFcsRIa Tg mice, we were

obliged to induce expression of the ß chain in the cell types that express the transgenic a

chain. For this purpose, hmFcsRIß Tg mice were generated. Nevertheless, even after

crossing the hmFcsRIa and hmFcsRIß strains together to obtain double transgenic mice,

detectable levels of receptor expression could not be found in antigen presenting cells. This

result was unexpected as FcsRI mRNA expression was observed in these cells. Several

reasons for the lack of detectable protein expression of the high affinity IgE receptor in the

generated transgenic mice can be considered. First, a considerable difference in the

expression density of FcsRI between the classical IgE effector cells (mast cells and

basophils) and other FcsRI-expressing cells has been reported (Donnadieu et al., 2000). It

was therefore possible that very low expression levels of the receptor in the targeted cells of

our transgenic models could have prevented us from detecting the receptor by commonly

Page 80: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part II 77

used methods. However, this expression density difference has been accredited to the lack

of the ß chain on cells other than mast cells and basophils, which was overcome in our

approach by additional generation of hmFcsRIß Tg mice. Second, as Western blot analysis

did not show any association of the receptor chains, it can be speculated that regardless of

the addition of the ß chain to the system, the receptor complex might still be retained and

degraded in the ER. Additional analysis, such as confocal microscopy, would be required to

determine the exact location of the FcsRI chains in the cytosol. Moreover, to generate the

hmFcsRIß Tg strain rather than using the genomic sequence of FcsRIß for the transgene

construct, ß chain cDNA was employed. Thus, it cannot be excluded that the absence of

normally occurring mRNA maturation events, such as splicing, could modify the cell

processes and prevent the transgene from being translated. Lack of the ß chain would

consequently lead to degradation of the a chain in the ER. Lastly, it has to be envisaged

that mRNA of both the a and ß FcsRI chains detected by RT-PCR or Northern blot may

have not been translated into protein. This would explain the complete lack of FcsRIa

detection by both surface and intracellular FACS analysis. At the moment we do not have

an explanation for the absence of FcsRI translation. Many factors are involved in the

machinery of gene expression and translation and can account for absence of an expected

phenotype in transgenic mouse models. Mutations of the sequence could inhibit transgene

expression. Therefore, we are currently analyzing the mRNA sequences of the a and ß

chains in macrophages of hmFcsRIaß Tg mice.

iV. brasiliensis infection did not reveal any differences in protection against the parasite

between WT and hmFcsRIaß Tg mice. This may obviously be due to the lack of FcsRI

protein expression in the Tg mice. However, it is also important to evaluate whether iV.

brasiliensis infection was the appropriate model to investigate hmFcsRIaß Tg mice.

Clearance of iV. brasiliensis has been suggested to be IgE-independent (Watanabe et al.,

1988) and may therefore not involve FcsRI-mediated mechanisms. This would easily

explain the absence of differences in protection against iV. brasiliensis between WT and

hmFcsRIaß Tg mice. Thus, additional models should be investigated to analyze the

immune response in hmFcsRIaß Tg mice, such as parasite infections where IgE is required

for protection (e.g. T. spiralis (Gurish et al., 2004)).

Page 81: The Interplay of IgE and its High Affinity Receptor FceRI

78 Results Part II

Taken together, we generated two mouse models, the hmFcsRIa and the hmFcsRIß Tg

mice, which were crossed together to obtain double transgenic hmFcsRIaß mice. Although

presence of the transgenes was detected in the genomic DNA of all these models and their

mRNA expression was shown in the targeted cell types, no detectable protein expression

was observed. Additional investigation is required to determine the exact reason for lack of

protein expression in order to modify the transgenic mice and to enable them to be suitable

models for investigation of the role of FcsRI expression on APCs and eosinophils.

Page 82: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 79

4 Results Part III

Generation of a muFcsRIa-huIgGiFc fusion protein

4.1 Abstract

Studies investigating the interaction of IgE and its high affinity receptor, FcsRI, have

revealed novel functions and expression patterns of FcsRI. Mouse models have been

widely used to study the downstream effects of IgE binding to FcsRI and their impact on

immune responses. However, the lack of a specific anti-mouse FcsRIa antibody hampers

investigations of the role of FcsRI in mice. Here we describe the successful generation of a

fusion protein composed of the extracellular domain of murine FcsRIa and the human IgGi

Fc domain. This fusion protein was used for immunization of FcsRIa"" mice in an attempt

to induce production of anti-mouse FcsRIa antibodies.

Page 83: The Interplay of IgE and its High Affinity Receptor FceRI

80 Results Part III

4.2 Introduction

Cell surface receptors for immunoglobulin (Ig) Fe regions, Fe receptors (FcRs), are

involved in a broad range of tasks within the immune system and are important regulators

of immune responses by inducing either activation or inhibition of cellular effector

functions. FcRs are widely expressed on various hematopoietic cells allowing interactions

of the Fc domain of antibodies with many specialized cells leading to a diverse range of

immune responses (Daeron, 1997). Thus, FcRs are able to provide a link between humoral

and cell-mediated immunity. The functions of FcRs can be divided into three groups

(Takai, 2002). First, the most important function of FcRs is the regulation of cellular

immune responses. Triggering of FcRs leads to biological events such as degranulation of

mast cells (Metzger et al., 1986), phagocytosis by macrophages (Anderson et al., 1990),

proliferation of B lymphocytes (Phillips and Parker, 1983), or induction of cytokine and

chemokine expression (Turner and Kinet, 1999). Although most FcRs induce activating

signals through immunoreceptor tyrosine-based activation motifs (ITAMs), there is one

inhibitory FcR, FcyRIIB, which contains in its cytoplasmic domain an inhibitory motif

(ITIM) allowing down-regulation of many of the above cited effector responses (Ravetch

and Bolland, 2001). Second, FcRs play an important role in internalization of immune

complexes leading to enhanced antigen presentation (Regnault et al., 1999). The third

major function of FcRs involves transcellular transfer of Igs, such as transplacental transfer

of maternal IgG (Simister and Mostov, 1989) or transfer of IgA to mucosal surfaces

(Phalipon and Corthesy, 2003).

Two Fc receptors for IgE have been identified, the high affinity IgE receptor, FcsRI, and

the low affinity IgE receptor, FcsRII (CD23). FcsRI is the major surface molecule involved

in induction of allergic diseases (Kinet, 1999). Initially it was believed that the sole role of

FcsRI was to induce immediate-type hypersensitivity reactions through allergen/IgE-

mediated crosslinkage on the surface of mast cells and basophils. However, in the last

decade evidence has accumulated that in humans FcsRI is expressed not only on mast cells

and basophils but also on Langerhans' cells (Bieber et al., 1992; Wang et al., 1992),

monocytes (Maurer et al., 1994), eosinophils (Gounni et al., 1994), peripheral blood

dendritic cells (Maurer et al., 1996), and platelets (Joseph et al., 1997). More recent

investigations have shown that even in the absence of specific antigen, binding of

Page 84: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 81

monomeric IgE to FcsRI can induce signals leading to FcsRI upregulation (Yamaguchi et

al., 1997) and mast cell survival (Asai et al., 2001; Kalesnikoff et al., 2001). These novel

findings have shed new light on the functions of FcsRI and increased the interest in this

receptor.

In order to investigate surface expression of FcsRI in humans, several monoclonal

antibodies (mAbs) against the human a chain of the receptor have been generated (Rigby et

al., 2000; Riske et al., 1991; Wang et al., 1992). However, the lack of a mAb against the

murine FcsRIa makes it difficult to study specific FcsRIa expression in mouse models.

Binding of labeled IgE has traditionally been used to study expression of the muFcsRI, but

since IgE binds also to the low affinity receptor, CD23 (Delespesse et al., 1992), and an

additional receptor, Mac-2 (Cherayil et al., 1989; Frigeri and Liu, 1992), specific muFcsRI

binding cannot be observed. Therefore, the objective of the present study was to generate a

mAb against the murine a chain of the high affinity IgE receptor (muFcsRIa). As a first

step, we generated a fusion protein composed of the extracellular domain of muFcsRIa and

the human IgGi Fc domain (hulgGiFc). HulgGiFc allowed isolation of the fusion protein

via a protein A column. In this report we describe the successful production and isolation of

the muFcsRIa-huIgGiFc fusion protein as well as the results obtained from immunizations

of FcsRIa""

mice with the muFcsRIa-huIgGiFc fusion protein.

Page 85: The Interplay of IgE and its High Affinity Receptor FceRI

82 Results Part III

4.3 Materials and Methods

Bone marrow-derived mast cell (BMMC) culture

Femur and tibia bones were collected from C57BL/6 mice and bone marrow cells were

flushed out with BSS. Cells were washed and resuspended at an initial concentration of 106

cells/ml in FMDM containing 10% FCS, 50 uM 2-Mercaptoethanol, 20 ng/ml recombinant

mouse stem cell factor (SCF, R&D Systems), and 5% X63-IL-3 cell-conditioned medium

corresponding to about 5 ng/ml IL-3. Cells were maintained in tissue culture petri dishes at

20 ml/dish in a humidified incubator at 37°C with 5% C02. Non-adherent cells were

initially transferred to new dishes every other day, later once a week and subsequently

expanded as necessary. Fresh SCF and IL-3 were provided weekly. 6 week cultures

contained >80% mast cells confirmed by FACS analysis of CD117 expression.

Generation of muFcsRIa-huIgGiFc fusion protein

Total RNA was isolated from bone marrow-derived mast cells using TRIzol reagent

(Invitrogen Life Technologies). First-strand cDNA synthesis from 1 ug total RNA was

performed using random hexamers and SuperScript II reverse transcriptase (Invitrogen Life

Technologies). A 631 bp fragment encoding the extracellular domain of the muFcsRIa

gene was amplified using primers FcsRIaNhe 5'-

CTAGCTAGCATGGTCACTGGAAGGTCTG-3' and FcsRIaXba 5'-

GCTCTAGAAGTTGTAGCCAATAATACTTGC-3' (94°5'; [94°1'; 54°30"; 72°l']x35;

72° 10'; 10°). These primers introduce Nhe I and Xba I restriction sites respectively, both of

which were utilized for the cloning strategy. The PCR-product was purified from a 1%

agarose gel using the QIAquick gel extraction kit 250 (Qiagen). The isolated fragment was

subcloned into pGEM-T (Promega) and transformed into XL-1 blue competent bacteria.

Plasmid DNA was isolated from single colonies grown on agar-LB/ampicillin plates using

the QIAprep Spin Miniprep kit (Qiagen). Restriction digestion with Nhe I and Xba I

(Roche) was performed to select for clones containing the FcsRIa DNA insert. Positive

clones were sequenced and checked for mutations. Sequences were compared with the

mRNA sequence of the murine FcsRIa (NCBI: NM010184) (Ra et al., 1989). The FcsRIa

DNA with the correct sequence was then excised from plasmids by Nhe I and Xba I

restriction digestion and ligated into the pCMVKanaRFcB7H expression vector (kindly

Page 86: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 83

provided by J. Weber, Zürich) containing the sequence coding for the Fc portion of the

human IgGi. Prior to ligation, the pCMVKanaRFcB7H vector was also digested with Nhe I

and Xba I in order to replace the B7H fragment with the FcsRIa gene. The resulting

pCMV-KanaR-muFcsRIa-huIgGiFc vector was then transformed into XL-1 blue competent

bacteria. Plasmid DNA was isolated from colonies grown on agar-LB/kanamycin plates and

restriction-digested with Nhe I and Xba I as described above. Positive colonies were

confirmed by sequencing. Three samples of pCMV-KanaR-muFcsRIa-huIgGiFc plasmid

DNA were selected for transfection. Unless stated otherwise, all procedures were

performed according to the manufacturer's instructions.

Transfection

HEK293T human embryonic kidney cells were cultured in DMEM containing 10% FCS

and 50 uM 2-Mercaptoethanol. For transfection, 3-4xl06 cells were incubated in 25 uM

Chloroquine (7-Chloro-4-[4-diethylamino-l-Methyl-butylamino] Quinoline) (Sigma) for 1

hour at 37°C. DNA (1 ug GFP/hygR and 10 ug pCMV-KanaR-muFcsRIa-huIgGiFc

expression vectors) was prepared in 1 ml 250 mM CaCb, added dropwise to 1 ml 2xBBS

(BES-buffered solution) and incubated 2-5 min. Cells were supplemented with 10 ml fresh

medium and mixed gently with the DNA solution. After 8 hours of incubation at 37°C

medium was exchanged to remove the CaCl2 DNA solution. For stable transfection, cells

were cultured in presence of 200 ug/ml Hygromycin B.

ELISA

ELISA plates were coated with 1.6 ug/ml goat anti-human IgG (Jackson ImmunoResearch

Laboratories) in 0.1 M NaHC03 (pH 9.6) overnight at 4°C and then blocked with 5% (w/v)

bovine serum albumin (BSA). For detection of the muFcsRIa-huIgGiFc fusion protein in

supernatant of transfected HEK293T cells, serially diluted supernatant was added and

incubated for 2 hours. Detection of bound muFcsRIa-huIgGiFc was performed by adding

horseradish peroxidase (HRPO)-conjugated goat anti-human IgG (Jackson

ImmunoResearch Laboratories) at 0.8 ug/ml for 1 hour. For detection of anti-muFcsRIa-

huIgGiFc antibodies in serum of immunized mice or in supernatant of hybridoma, the

muFcsRIa-huIgGiFc fusion protein or a control hulgGiFc fusion protein at 10 ug/ml was

added to plates coated with goat anti-human IgG and incubated for 1 hour. Thereafter,

Page 87: The Interplay of IgE and its High Affinity Receptor FceRI

84 Results Part III

serially diluted sera or hybridoma supernatants were incubated for 2 hours, followed by

incubation of HRPO-conjugated goat anti-mouse IgG (Sigma) at 1 ug/ml. ABTS

peroxidase solution (2,2'-azino-bis-[3-ethylbenzthiazidine-6-sulfonic acid] 0.1 mg/ml,

0.05%) H202, and 0.1 M NaH2P04 pH 4.0) was used for the peroxidase-catalyzed color

reaction. Color reaction was read at 405 nm in a Bio-Rad microplate reader.

Western blot

Transfected HEK293T cells were grown in serum-free InVitrus medium (R&D Laboratory)

for 3 days and supernatant from a confluent T25 flask was collected by centrifugation (15

min, 4000 RPM). 25 ul protein A sepharose CL-4B (Pharmacia Biotech) was added per 1

ml supernatant and incubated on a rotor at 4°C overnight. Sepharose beads were then

washed twice with PBS and samples denaturated by incubating for 5 min at 95°C in SDS

reducing loading buffer. After centrifugation (3 min, 13000 RPM) proteins were separated

on 10%) SDS-polyacryamide Tris gels (Bio-Rad Laboratories) and then electrotransferred to

Protran nitrocellulose membranes (Schleicher&Schuell) using a semi-dry transfer unit

(SemiPhor Hoefer). Membranes were blocked in 4% milk/PBS overnight at 4°C and probed

with HRPO-conjugated goat anti-human IgG antibody (Caltag Laboratories) at 0.7 ug/ml

for 2 hours. Revelation was performed using SuperSignal West Pico Chemiluminescent

Substrate (Pierce) according to the manufacturer's instructions.

Flow cytometry

To analyze the presence of anti-muFcsRIa antibodies in sera of immunized mice or

supernatants of generated hybridoma, BMMC were incubated with 1:2 diluted sera or

supernatants for 20 min on ice. Thereafter cells were surface-stained with anti-mouse IgG-

FITC (2.5 ug/ml, Caltag Laboratories) and anti-mouse CD117-PE (0.7 ug/ml, BD

Biosciences). To test for expression of FcsRIa on BMMC, cells were incubated with

mouse IgE (clone RIE4) at 50 ug/ml and stained thereafter with anti-mouse IgE-FITC (20

ug/ml, Southern Biotechnology). Cells were aquired in a FACS Calibur (BD) and analyzed

using FlowJo software.

Page 88: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 85

Mice

C57BL/6 mice were bred and maintained at the Institute of Laboratory Animal Science of

the University of Zurich. FcsRIa"" mice (Dombrowicz et al., 1993) were kindly provided

by Jean-Pierre Kinet, Boston, USA.

Immunization

muFcsRIcc-huIgGiFcfusion protein injections:

FcsRIa"" mice were tolerized against human IgGi by an i.v. injection with 500 ug of

human IgGiK (Sigma). Two weeks later an initial immunization with 20 ug muFcsRIa-

huIgGiFc fusion protein in complete Freund's adjuvant (CFA) s.c. was administrated

followed by five booster immunizations at 2-week intervals: four with 20 ug muFcsRIa-

huIgGiFc fusion protein in incomplete Freund's adjuvant (IFA) s.c. and one with 50 ug

muFcsRIa-huIgGiFc fusion protein in IFA i.p.. A final immunization with 50 ug

muFcsRIa-huIgGiFc fusion protein in PBS i.p. was administrated 1 day after the last boost.

Mast cells injections:

FcsRIa"" mice were immunized five times with 106-107 BMMC in PBS i.p. at 2-week

intervals. A final immunization with 106 BMMC in PBS i.v. was administrated 1 day after

the last boost.

For both immunization protocols, splenocyte fusion to myeloma cells was performed 3

days after the final immunization.

Hybridoma production

Spleens of immunized animals were isolated and single cell suspensions prepared. 2xl07

splenocytes and 2xl07 cells of the mouse myeloma cell line X63AG8.653 were mixed.

Fusion was induced by adding 1% PEG 4000 (Polyethylenglycol, Merck) in BSS to the cell

pellet at 37°C. After fusion, cells were resuspended in pre-warmed HAT-IMDM medium

(Hypoxanthine-Aminopterine-Thymidine, Gibco) and distributed into 96-well plates

containing peritoneal macrophages isolated from C57BL/6 mice and plated one day before

fusion. Hybridoma colonies were detectable 7-10 days after fusion and screening of

supernatant was performed once medium had turned orange.

Page 89: The Interplay of IgE and its High Affinity Receptor FceRI

86 Results Part III

4.4 Results

Cloning of the fusion gene muFcsRIa-huIgGiFc

The initial objective of the present study was to generate a fusion protein composed of the

extracellular domain of the murine a chain of the high affinity IgE receptor (muFcsRIa)

and the Fc portion of the human IgGi (hulgGiFc). The hulgGiFc was added to the

muFcsRIa protein to enable isolation of the expressed protein via binding to a protein A

column. To achieve expression of the muFcsRIa-huIgGiFc fusion protein, the sequences

coding for both the muFcsRIa extracellular domain and the hulgGiFc fragment were

cloned together into an expression vector (pCMV-KanaR). To obtain the muFcsRIa

sequence, bone marrow-derived mast cells (BMMC) from C57BL/6 mice were prepared

and total RNA was isolated from the cultured cells, which had been tested for purity by

CD117 expression analysis and of which FcsRIa expression was confirmed by IgE binding

(Figure 18A). The extracellular domain of the murine a chain of the high affinity IgE

receptor was then amplified using specific primers, which also introduced restriction sites

for the enzymes Nhe I and Xba I. As shown in Figure 18B, the primers aligned in exon 1

and exon 5 of the muFcsRIa gene yielding a PCR-product of 631 bp. The PCR-product

was then subcloned into the pGEM-T vector and transformed into XL-1 blue competent

bacteria. Single clones containing the insert were identified by restriction digestion using

Xba I and Nhe I and then sequenced. Three samples (4c, 5b, 8a) with the correct sequence

of muFcsRIa were then cloned into the pCMV-KanaR-huIgGiFc expression vector

containing the hulgGiFc (Figure 18C).

Page 90: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 87

-24.1 75.3 99.2 0.054

Q

O

m m

0.42 0.11 0.67

IgE + anti-lgE

0.027

anti-lgE control

B

Nhe I

EXTRACELLULAR TM CP

5

IB

100 bp BMMC cDNA H20Ladder

500-

Xbal

muFceRIa protein domains

^^m muFceRIa gene exons

muFceRIa'

631 bp

CMVNhe I

KanaR

muFctKI«

831 bp

Xbai

pCMV-muFceRIcx-huIgG-]Fc5274 bp ^huig^Fc

p "" 723 bp

SV40 ori

Figure 18. Amplification of the muFceRIa sequence and cloning of the pCMV-muFceRloc-

hulgG.|Fc expression vector. BMMC were analyzed for purity (CD117 expression) and expression

of FceRI (binding of IgE) (A). The percentage of the cells present in each quadrant is indicated. A

primer set aligning within exon 1 and exon 5 of the muFceRIa sequence and containing restriction

sites for Nhe I and Xba I was used in RT-PCR on total RNA isolated from BMMC (B). The upper line

indicates the protein domains of FceRIa (TM=transmembrane, CP=cytoplasmic). The resulting 631

bp PCR-product coding for the extracellular domain of muFceRIa was cloned into the pCMV-KanaR-

hulgG^c vector (C). Kanamycin resistance was used for amplification of the vector.

Page 91: The Interplay of IgE and its High Affinity Receptor FceRI

88 Results Part III

Transfection and screening for cells expressing the muFcsRIa-huIgGiFc fusion

protein

The human embryonic kidney cell line HEK293T was transfected with the pCMV-

muFcsRIa-huIgGiFc vector to achieve expression of the muFcsRIa-huIgGiFc fusion

protein. To allow selection of transfected cells, a vector expressing the green fluorescence

protein (GFP) and Hygromycin resistance (HygR) was co-transfected into HEK293T cells

together with the pCMV-muFcsRIa-huIgGiFc vector. Transfected cells were identified by

GFP expression, using a fluorescent microscope. All three transfections obtained from the

constructs 4c, 5b, and 8a showed positive cells based on the presence of green fluorescence

(Figure 19A). At different time points after transfection, presence of the muFcsRIa-

huIgGiFc fusion protein in the supernatant of transfected cells was determined by ELISA

(Figure 19B). Cells transfected with the construct 4c produced the highest concentration of

protein, however, cells transfected with constructs 5b and 8a also showed substantial fusion

protein expression. As all three transfected cell lines produced the fusion protein, all three

were subcloned in order to obtain clonal cell lines. All cell lines derived from single cells

were tested by ELISA to determine if fusion protein expression was maintained. Over 300

clones were tested (data not shown) of which 31 with highest protein expression were

chosen to be expanded and tested again by ELISA. Three clones, labeled 8a3B, 5b4B, and

4c 1 A, with the highest protein expression were selected for further investigation. Western

blot analysis revealed that clone 8a3B appeared to produce the highest amounts of the

muFcsRIa-huIgGiFc fusion protein (Figure 19C). This clone was therefore selected for

further expansion and subsequent isolation of the fusion protein.

Page 92: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 89

Figure 19. Transfection of HEK293T cells with three constructs coding for the muFceRloc-

hulgG.|Fc fusion protein. pCMV-muFceRla-hulgG^c vectors were co-transfected into HEK293T

cells together with a vector coding for GFP and HygR. After transfection cells were grown in

presence of Hygromycin and tested for GFP expression. Cells transfected with three selected

constructs of pCMV-muFceRla-hulgG^c (4c, 5b, and 8a) are depicted 16 days after transfection

(A). ELISA analysis for presence of hulgG^c in supernatants of transfected cells 19 days post

transfection (B). A 3-fold serial dilution was performed on initially undiluted supernatants.

Background of the ELISA was 0.16. Western blot analysis for presence of hulgG^c in the

supernatants of three clonal transfected cell lines (8a3B, 5b4B, and 4c1A) (C). A control hulgG^c

fusion protein was used as reference for the results. ELISA and Western blot results are

representative of three separate experiments.

Isolation of the muFcsRIa-huIgGiFc fusion protein

In order to purify the muFcsRIa-huIgGiFc fusion protein, four liters of supernatant of the

clone 8a3B were concentrated down to 100 ml and the fusion protein was isolated by

binding to a protein A column. Bound protein was eluted from the column and collected in

4 ml fractions. As shown in Figure 20A, fractions 5 to 11 contained high amounts of

protein with the protein peak eluting in fraction 6. The presence of the muFcsRIa-

Page 93: The Interplay of IgE and its High Affinity Receptor FceRI

90 Results Part III

hulgGiFc fusion protein in fractions 5 through 11 was then tested by specific ELISA with

all fractions revealing the presence of high amounts of fusion protein (Figure 20B).

Fractions 5 to 11 were therefore pooled and the total amount of protein determined. The

final yield of the isolated muFcsRIa-huIgGiFc fusion protein was 6 grams at a

concentration of 0.1 mg/ml.

A

Ecoeo

£S

Q

o

B

Ecmo

Q

O

r

3 4 5

Dilution [log3]

Fraction 5

Fraction 6

-- Fraction

-•- Fracti

-o- Fracti

-A- Fraction

-v- Fraction

7

on 8

on 9

10

Figure 20. Elution and analysis of the muFceRloc-hulgGiFc fusion protein. Four liters of

supernatant was collected from transfected clone 8a3B and concentrated to 100 ml. Fusion protein

was purified by binding to a protein A column and bound protein eluted in 4 ml fractions. The OD

(280 nm) of the eluent was measured (A). Fractions with a high OD are depicted in yellow.

Presence of hulgG-iFc in each of the fractions 5-11 was measured by ELISA (B). Samples were

pre-diluted 1:10 and then a 3-fold serial dilution was performed. Background of the ELISA was 0.14.

Page 94: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 91

Immunization of FcsRIa""

mice with the muFcsRIa-huIgGiFc fusion protein

After successful generation of the muFcsRIa-huIgGiFc fusion protein, the second aim of

this study was to generate monoclonal anti-muFcsRIa antibodies. To obtain such

antibodies, mice deficient in FcsRIa (Dombrowicz et al., 1993) were chosen for

immunization with the fusion protein as these mice should recognize FcsRIa as a foreign

protein and therefore produce anti-FcsRIa antibodies. Prior to immunization with the

fusion protein, FcsRIa"" mice were tolerized against human IgGi by injection of a high

amount of human IgGi. Tolerance against human IgGi was induced in an attempt to

prevent production of antibodies against the hulgGiFc portion of the fusion protein. To

generate an anti-FcsRIa antibody response, FcsRIa""

mice were then immunized with the

muFcsRIa-huIgGiFc fusion protein six times at two-week intervals. Blood was collected

prior to each immunization in order to investigate the presence of anti-muFcsRIa

antibodies in the serum. The immunization protocol was performed three times with 3-4

mice per group. Representative results obtained from one immunization regime are shown

in Figure 21 (FACS) and Figure 22 (ELISA).

The presence of anti-muFcsRIa antibodies in serum of immunized FcsRIa""

mice was

analyzed by FACS using BMMC. These BMMC were CD 117 positive and expressed high

amounts of FcsRIa as visualized by staining with IgE and anti-lgE (Figure 21 A). Thus, in

order to detect anti-muFcsRIa antibodies, serum was incubated with BMMC and anti-

muFcsRIa binding was subsequently detected using fluorochrome-labeled anti-mouse IgG

antibody, as illustrated in Figure 21B. None of the immunized FcsRIa"" mice had

detectable amounts of anti-muFcsRIa antibodies in their serum, although serum from

mouse #3 may have contained small amounts of antibody as a small increase in mast cell

binding was observed (Figure 21C).

Page 95: The Interplay of IgE and its High Affinity Receptor FceRI

92 Results Part III

A iL1°4

32 8 65 6

-m0

0 71 0 87

B

10 u i(r u 1G*

IgE + anti-lgE

mouse #1

0498 9 0 27

10 10 10 10J 104

anti-CD117 J» JLanti-mouse IgG

CD117^^*SerUmIPFceRIa

BMMC

mouse #2 mouse #3 mouse #4

98 5 0 52114

94 4 4 39"

981 0 25

#103-

102-

10 -

0 I «p

101

3° boost

0 95 0 05 0 88 0 280

1 41 0 25

10u 10 10' 10 104 10 10' 10 101 IQ"1 10u 10 10y 105 10*

98 2 0 54 98 3

1 12 017

10 10' 10 10^ 10"*

0 83

0 25

10'

94 7 41

^o'>-

m-io2-

10 -

0 88 0 33

98 6 0 047

4° boost

1 35 0 023

10 10 10y 10 104 10 10' 10 10J 104 10u 10 10' 101 10*

1V

97 6 0 92

n3-»

102-

fP»

10

1 18 0 28

97 7 1 39

o2

o1

.n0 81 0 14

it*

95 5 3 29

10*-

Iffio9-

10 -

0 87 0 32

98 6 0 27

5° boost

10 10' 10 101 10"* iou 10 ioy io5 iC 10 10 10 10J 104 10u 10 10' 10 104

serum + anti-mouse IgG

Figure 21. FACS analysis of sera of FcsRIa"'" mice immunized with the muFceRla-hulgGiFc

fusion protein. BMMC (CD117+) expressing high levels of FceRI (binding to IgE) were used for

FACS analysis (A). To detect anti-muFceRIa antibodies in blood of immunized mice, serum was

incubated with BMMC and binding to FceRIa was subsequently detected by fluorochrome-labeled

anti-mouse IgG antibodies (B). Analysis of serum of four FceRIa' mice collected before the third,

fourth and fifth boost with the muFceRla-hulgG^c fusion protein is shown (C). The percentage of

the cells present in each quadrant is indicated.

Serum samples were also analyzed by ELISA for the presence of antibodies binding to the

muFcsRIa-huIgGiFc fusion protein To confirm that antibodies present in the sera of

immunized mice were binding to the muFcsRIa portion of the fusion protein and not to the

hulgGiFc fragment, a control hulgGiFc fusion protein was included in the analysis, as

illustrated in Figure 22A Results of ELISA analysis using the muFcsRIa-huIgGiFc fusion

Page 96: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 93

protein for antibody binding detection showed high levels of protein binding in sera

collected before the third, fourth, and fifth boost with the muFcsRIa-huIgGiFc fusion

protein (Figure 22B, orange symbols). However, sera collected prior to the fifth boost also

revealed high protein binding to a control hulgGiFc fusion protein (Figure 22B, green

squares). Moreover, analysis of serum collected prior to the first immunization with the

muFcsRIa-huIgGiFc fusion protein also revealed binding to the control hulgGiFc fusion

protein (Figure 22B, green circles). These data indicated the presence of anti-huIgGi

antibodies in the sera of immunized FcsRIa""

mice. Such antibodies were most likely

generated after injection of the human IgGi used for tolerization and could be detected in

the ELISA but not in the FACS analysis due to the different experimental setup.

A

}VtiHmouse IgG

BiuFccRlrif-huloGFc or controI-huIgGFc

jfanti-huIgG

Dilution [log3]

Figure 22. ELISA analysis of sera of FceRIa"'" mice immunized with the muFceRla-hulgGiFc

fusion protein. Plates were coated with anti-human IgG and incubated with the muFceRla-

hulgG^c fusion protein or a control hulgG^c fusion protein. Subsequently, plates were incubated

with sera of immunized mice and protein binding was detected with anti-mouse IgG (A). Results of

serum from mouse #3 collected prior to the first (•), third (A), fourth (T), and fifth () boost using

either the muFceRla-hulgG^c fusion protein (orange) or a control IgG^c fusion protein (green) for

detection are shown (B). Serum was pre-diluted 1:50 and then a 3-fold serial dilution was

performed. Background of the ELISA was 0.25.

Surprisingly, immunization of FcsRIa""

mice with the muFcsRIa-huIgGiFc fusion protein

caused development of skin lesions, which were not observed in control mice injected with

adjuvant alone. Due to the skin lesion development as well as the apparent absence of

specific anti-muFcsRIa antibodies, a different strategy of immunization was attempted

where FcsRIa""

mice were immunized with BMMC expressing high levels of FcsRI.

Page 97: The Interplay of IgE and its High Affinity Receptor FceRI

94 Results Part III

Immunization of FcsRIa""

mice with bone marrow-derived mast cells.

Immunization of FcsRIa""

mice with the muFcsRIa-huIgGiFc fusion protein revealed two

difficulties. First, rather than generating specific anti-muFcsRIa antibodies, immunized

mice produced antibodies against the human IgGi. Second, the immunization caused severe

skin lesions to a degree that some mice had to be removed from the experiment. This

prompted us to develop a new strategy for generation of anti-muFcsRIa antibodies. We

decided to immunize FcsRIa""

mice with BMMC, which express high levels of FcsRIa on

the cell surface. Blood was collected before each BMMC injection and serum was screened

for presence of anti-muFcsRIa antibodies using the same FACS and ELISA protocols as

described for immunization with the muFcsRIa-huIgGiFc fusion protein (Figures 2IB and

22A). The immunization regime was performed twice on 3-4 mice per group and

representative results of two mice are depicted in Figure 23. Interestingly, the presence of

mouse IgG antibodies binding to BMMC in serum of FcsRIa""

mice increased upon

immunization with BMMC, as monitored by FACS showing a shift of stained BMMC from

about 1% to 8% (Figure 23A). This suggested that increasing amounts of BMMC-binding

antibodies were induced upon BMMC immunization. ELISA analysis also revealed the

presence of antibodies binding to the muFcsRIa-huIgGiFc fusion protein in the sera of

FcsRIa""

mice immunized with BMMC (Figure 23B, closed symbols). No such binding

was observed in sera from naive FcsRIa""

mice (Figure 23B, open symbols). Importantly,

no antibody binding was detected using a control hulgGiFc fusion protein (data not shown)

indicating that antibodies present in serum of immunized mice bound specifically to the

muFcsRIa portion of the muFcsRIa-huIgGiFc fusion protein.

Thus, both the FACS and ELISA data confirmed induction of antibodies in FcsRIa""

mice

in response to immunization with BMMC. B cells from immunized mice were therefore

used to generate hybridoma cell lines producing anti-muFcsRIa antibodies. Splenocytes

were fused with myeloma cells and supernatants of successfully generated hybridoma

single cell clones were tested for the presence of anti-muFcsRIa antibodies by specific

FACS and ELISA. Over 100 hybridoma clones were generated, however, none of these

hybridoma were found to produce antibodies binding to either BMMC by FACS (Figure

24) or to the muFcsRIa-huIgGiFc fusion protein by ELISA (data not shown). Thus, the

hybridoma production did not lead to generation of a monoclonal anti-muFcsRIa antibody.

Page 98: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 95

A BMMC control 2nd Ab control

93 8 0 38

JÉif

5.8 0.18,

IgE + anti-lgE anti-mouse IgG

mouse #2 mouse #6

87 2 2.33

i*l"

4 22 6 25

76 9 101

r

#

434 86

,93

<? ^

serum + anti-mouse IgG

1 23

1° boost

2° boost

3° boost

-A- mouse #2 naive

—O- mouse #6 naive

—A- mouse #2 5° boost

mouse #6 5° boost

Dilution [log3]

Figure 23. Detection of anti-FceRIa antibodies in FceRIa"'" mice immunized with BMMC. FACS

(A) and ELISA (B) were performed on sera of immunized mice as described in Figure 21B and 22A,

respectively. The top panels show control stains of BMMC with IgE to confirm high levels of FceRI

expression and with anti-mouse IgG used as secondary Ab in the subsequent stains.

Representative results of two mice (#2 and #6) are shown. The percentage of the cells present in

each quadrant is indicated. For ELISA analysis, serum was pre-diluted 1:10 and then a 3-fold serial

dilution was performed. Background of the ELISA was 0.1.

Page 99: The Interplay of IgE and its High Affinity Receptor FceRI

96 Results Part III

o

o

BMMC control

44 1 43 1

2nd Ab control

86 5 0 25

126

/

0 14 ,132, .

J 1t!JG 028

10° 101 10^ 10J 104 10 101 109 10* 10

IgE + anti-lgE anti-mouse IgG

Hybridoma 2B7t

Hybridoma 2D3

0 261°

389 5 0 16

10u 10' 10 10 10H

4Hybridoma 6F9

'89 5 0 14

103 0

10 101 Vf 10" 104

Hybridoma 11H10

186 8 0 098

4

131

104

10" 10' 10z 10 10

Hybridoma 9F9'

Î86 5 0 29

o'- '

10" 10' 10z Vf V

Hybridoma 12A2

86 1 0 46

£.

134

10u 10' 10 10 10H 10" 10' Wf 10 10

serum + anti-mouse IgG

Figure 24. Analysis of hybridoma generated from BMMC-immunized FcsRIa" mice.

Supernatant of generated hybridoma clones was tested by FACS using BMMC as described in

Figure 21B. Six representative clones of over 100 tested hybridoma are depicted. The percentage

of the cells present in each quadrant is indicated.

Page 100: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 97

4.5 Discussion

The high affinity IgE receptor, FcsRI, regulates signals leading to immediate-type

hypersensitivity reactions by binding of IgE on mast cells or basophils. In the presence of

specific antigen, the receptor is crosslinked leading to degranulation of the cell and release

of allergic mediators (Kinet, 1999). Signaling through FcsRI can also be triggered by

monomeric IgE, in the absence of antigen, resulting in FcsRI upregulation and cell survival

(Asai et al., 2001; Kalesnikoff et al., 2001; Yamaguchi et al., 1997). In addition to its

classical funcions on mast cells and basophils, in humans, FcsRI has also been shown to be

expressed on Langerhans' cells (Bieber et al., 1992; Wang et al., 1992), monocytes (Maurer

et al., 1994), eosinophils (Gounni et al., 1994), peripheral blood dendritic cells (Maurer et

al., 1996), and platelets (Hasegawa et al., 1999).

To allow specific investigation of FcsRI expression and function in mouse models, we

undertook to generate a monoclonal anti-mouse FcsRIa antibody. The first aim was to

generate a fusion protein composed of the extracellular domain of the murine FcsRIa and

the human IgGi Fc fragment. For this purpose, sequences coding for muFcsRIa and

hulgGiFc were cloned into an expression vector and transfected into a mammalian cell line.

Expression of the fusion protein was confirmed by ELISA and Western blot analysis

detecting hulgGiFc in the supernatant of transfected cells. The muFcsRIa-huIgGiFc fusion

protein was isolated from the supernatant using binding properties of hulgGiFc to protein

A.

After successful generation and isolation of a considerable amount of purified fusion

protein, the second aim of this project was to use this muFcsRIa-huIgGiFc fusion protein

to immunize FcsRIa""

mice in an attempt to generate anti-muFcsRIa antibodies. FcsRIa""

mice are deficient in the a chain of the high affinity IgE receptor, thus immunization with

the fusion protein should induce production of antibodies against FcsRIa. However, as the

fusion protein was also composed of the hulgGiFc domain, we were obliged to tolerize the

mice with hulgGi in order to prevent generation of antibodies against the hulgGiFc portion

of the fusion protein. Nevertheless, our results suggest that rather than tolerization, FcsRIa-

deficient mice developed anti-huIgGi antibodies. One possible way to overcome such a

problem would be to remove the hulgGiFc portion of the fusion protein from the

muFcsRIa portion prior to immunization. A separation of the two portions would have

Page 101: The Interplay of IgE and its High Affinity Receptor FceRI

98 Results Part III

been possible by introducing a cleavage site, such as for Factor Xa, into the expression

vector between the sequences coding for muFcsRIa and hulgGiFc.

We encountered an additional difficulty with the muFcsRIa-huIgGiFc fusion protein when

attempting to investigate binding to IgE. Although we could detect binding of muFcsRIa-

huIgGiFc to anti-huIgG antibodies by both ELISA and Western blot, despite numerous

protocols used, we were not able to show binding of muFcsRIa-huIgGiFc to murine IgE.

This may indicate that the muFcsRIa portion of the fusion protein was not properly folded

such that the three-dimensional structure did not reveal the IgE-binding site. Alternatively,

the presence of the hulgGiFc portion may have prevented binding of IgE, possibly due to

steric hindrance. In either case, removal of the hulgGiFc portion by Factor Xa, as suggested

above, might have allowed successful binding of IgE to the muFcsRIa portion.

Surprisingly, FcsRIa"" mice immunized with the muFcsRIa-huIgGiFc fusion protein

responded with unexpected skin lesions. At present the mechanism behind induction of

these skin lesions is unclear, even if the response appeared to be specific for the fusion

protein, as adjuvant alone had no effect. Although the formation of these lesions may be

immunologicaly interesting, they nevertheless prevented us from pursuing the

immunization protocol using the muFcsRIa-huIgGiFc fusion protein. As an alternative we

developed a different immunization method based on injections of FcsRIa"" mice with

BMMC expressing high levels of FcsRI. Analysis of serum of FcsRIa""

mice immunized

with BMMC revealed the presence of antibodies binding to BMMC, however, generation

of hybridoma clones from splenocytes of these mice did not result in production of anti-

muFcsRIa antibodies. The reason for unsuccessful generation of hybridoma producing

anti-muFcsRIa antibodies is presently unclear. It is possible that generation of additional

hybridoma would eventually yield specific antibody production. However, during the

completion of this study a commercial anti-muFcsRIa antibody became available and the

project was therefore terminated.

In conclusion, a muFcsRIa-huIgGiFc fusion protein was successfully generated, expressed

in the HEK293T cell line, and substantial amounts of protein were purified. The fusion

protein was shown to bind specifically to anti-huIgG but not to IgE. Immunization of

FcsRIa""

mice with the muFcsRIa-huIgGiFc fusion protein or with BMMC did not result

Page 102: The Interplay of IgE and its High Affinity Receptor FceRI

Results Part III 99

in generation of anti-muFcsRIa antibodies, however, the fusion protein is available in large

amounts and can be used in future if needed.

Page 103: The Interplay of IgE and its High Affinity Receptor FceRI

100 Results Part III

Page 104: The Interplay of IgE and its High Affinity Receptor FceRI

Discussion 101

5 Discussion

5.1 The functions of IgE

IgE, formerly referred to as "reagin", was first recognized as a new immunoglobulin class

in 1967 (Ishizaka and Ishizaka, 1967; Johansson and Bennich, 1967). However, initial

description of reagin was performed much earlier, when Cooke and Vander-Veer published

a first report on the genetic predisposition to allergy in 1916 (Cooke, 1916). The role of

reagin in hypersensitivity was further confirmed by Prausnitz and Küstner who

demonstrated in 1921 that this serum factor was responsible for allergic reactions

(Prausnitz, 1921). Thus, the first identified function of IgE was related to its involvement in

the development of allergy. However, it seems rather astonishing that IgE would evolve to

induce harmful immune responses suggesting that there must also be a beneficial function

of IgE in immunity that has led to the evolution of this antibody. Indeed, in 1964 Bridget

Ogilvie proposed that IgE (at that time still called reaginic antibody) may play an important

role in immunity to parasitic helminth infections (Ogilvie, 1964). Since then, a large body

of literature has accumulated to demonstrate the host-protective function of IgE in parasite

infections, as reviewed by Hagan (Hagan, 1993). Although high IgE levels are associated

with most parasite infections and a protective role has been assigned to IgE against

parasites such as Schistosoma mansoni (Capron et al., 1987) and Trichinella spiralis

(Dessein et al., 1981), the involvement of IgE in host-defense mechanisms remains

controversial for many parasite species. This suggests that IgE participation in parasite

infections may strongly depend on the characteristics of individual parasites, including

different life cycles, host penetration routes, and tissue localizations. Thus, further

investigation is required to determine the mechanisms regulating effector elements involved

in protection against each individual parasite species.

Taken together, IgE is involved in two types of immune responses. On one hand, IgE is

produced in response to parasite infections and may be involved in anti-parasitic host-

defense. On the other hand, IgE mediates adverse immune responses resulting in allergic

disease. The most prevalent parasite infections (ascariasis, trichriasis, and hookworm

infections) are believed to affect nearly two billion people worldwide, mostly in East Asia

and Sub-Saharan Africa (de Silva et al., 2003). These numbers are far from declining and

Page 105: The Interplay of IgE and its High Affinity Receptor FceRI

102 Discussion

afflict considerable human suffering and economic hardship. Understanding the factors

mediating protection against parasites is therefore of high importance. Contrary to

nematode infections, allergy mostly affects western countries, which may explain why

significantly more research has been done on IgE in the context of allergic disease than on

immunity to parasites. Interestingly, both branches of IgE biology, parasite protection and

allergy, appear to rely on the same effector mechanisms. Thus, investigation of either of the

two "faces" of IgE may provide valuable means to diminish the harm of both allergy and

parasite infections.

5.2 The epidemic of allergy

The benefits of a contemporary westernized lifestyle, such as low infant mortality due to

improved sanitation and access to drinkable water, strongly correlate with an increased

prevalence of allergy. The incidence of allergic disorders has risen greatly in the past

decades reaching epidemiological dimensions, particularly in the developed world. Most

allergic reactions are not life threatening, however they cause distress for millions. Quality

of life is significantly decreased by allergic rhinitis, eczema, and asthma, which interfere

with sleep, intellectual functioning, and recreational activities. Food allergies cause

constant concern with eating habits and fear of unnoticed ingestion of the respective

allergen. More dramatic outcomes of allergy-related pathologies are illustrated with

anaphylaxis and severe asthma, which can cause death. Statistical studies of allergic

disorders report alarming results. In countries such as the UK, Australia, or New Zealand,

around 25% of children under the age of 14 are reported to be affected with asthma and/or

eczema (1998). A few years ago the cost of treating asthma in the United States was

estimated to be about 6 billion US dollars per year (Smith et al., 1997) and may now be

significantly higher. Allergic diseases are a feature of westernized societies and have

therefore been suggested to result from reductions in the nature and frequency of childhood

infections. To explain the correlation between reduced infectious stress and increased

prevalence of allergy, different models have been proposed, the two most prominent being

the hygiene hypothesis and the counter-regulatory model (Wills-Karp et al., 2001).

However, both of these models have been disputed and additional evidence is required to

clearly demonstrate the environmental factors and immunological regulators responsible for

the observed increase in allergic disorders.

Page 106: The Interplay of IgE and its High Affinity Receptor FceRI

Discussion 103

5.3 Mechanisms of IgE-mediated immune responses

Allergic conditions are believed to share a unique mechanism of disease induction, which

involves high levels of IgE production. Indeed, total serum IgE concentrations have been

shown to positively correlate with the severity of allergic reactions (Burrows et al., 1989;

Laske and Niggemann, 2004). Binding of IgE to its high affinity receptor, FcsRI, expressed

most importantly on mast cells, followed by antigen-mediated crosslinking of receptor-

bound IgE, induces the release of allergic mediators leading to symptoms of atopic disease

(Kay, 2001). The pathology of atopy includes weal and flare eruptions of the skin (hives

and urticaria), rhinitis, asthma, and anaphylaxis. Consequently, investigating the factors

that control IgE production is essential to understand the pathogenesis of allergy. The

regulation of IgE production is based on the activation of Th2 cells and secretion of Th2-

type cytokines, most importantly IL-4 and IL-13 (Geha et al., 2003). Understanding the

mechanisms regulating the induction of Th2-mediated immune responses is crucial to

provide effective means against allergic disease.

5.4 Remaining questions to be answered

Despite much progress in allergy research, many questions concerning the development of

the disease remain open and hinder the development of effective therapies. A great number

of factors have been shown to influence the outcome of allergic responses, including early

events during fetal life and childhood, genetic background, and lifestyle (Cookson, 1999).

This illustrates the complexity of the disease and explains the difficulty of successful

therapy and prevention.

One of the most basic questions regarding IgE production concerns the polarization of the T

helper response. Naive CD4+ T cells differentiate upon encounter of specific antigens into

two functionally distinct effector subtypes, Thl and Th2 (Abbas et al., 1996). Polarization

of T helper cells depends mainly on the cytokine milieu. Thus, induction of Thl cells

requires the presence of IL-12, which is produced by APCs after pathogen-dependent

activation of Toll-like receptors (Dabbagh and Lewis, 2003). On the other hand,

development of Th2 cells requires the cytokine IL-4, which is also crucial for the

production of IgE (Finkelman et al., 1988). However, the molecular basis for Th2

polarization is poorly understood. Recent findings have suggested that thymic stromal

lymphopoietin (TSLP) produced by epithelial cells at the site of antigen entry in skin and

Page 107: The Interplay of IgE and its High Affinity Receptor FceRI

104 Discussion

mucosa may interact with human dendritic cells. These interactions have been suggested to

induce production of Th2-attracting chemokines, such as CCL17 and CCL22, and Th2

differentiation characterized by secretion of IL-4, IL-5, and IL-13 (Soumelis et al., 2002).

However, in mice TSLP has no effect on dendritic cells (Leonard, 2002) implying that

other mechanisms must be involved in Th2 differentiation in the murine system. Ample

research has been performed to investigate the relevant source of IL-4, however, the

mechanisms involved in its induction remain unclear (Liew, 2002). Thus, as long as the

factors inducing a Th2-type response are not elucidated, the initiation of IgE-mediated

responses will remain unresolved. Several cell subsets have been shown to have the

capacity of producing IL-4. Mature CD4+ T cells have been reported to be a sufficient

source of IL-4 for initial induction of IgE synthesis (Schmitz et al., 1994). Mast cells (Plaut

et al., 1989) and basophils (Seder et al., 1991) are known to produce IL-4 upon

crosslinkage of high affinity Fes receptors. Interestingly, mast cells and eosinophils have

been shown to express IL-4 in bronchial mucosa of asthmatic patients (Ying et al., 1997)

and cultured basophils have been demonstrated to produce biologically active IL-4 and

express functional CD40L contributing to IgE production by B cells (Yanagihara et al.,

1998). Recent studies using IL-4 reporter mice have shown IL-4 synthesis by basophils and

eosinophils in response to infection with a Th2-inducing parasite (Min et al., 2004;

Voehringer et al., 2004). Furthermore, natural killer (NK) T cells have been suggested to

favor the development of Th2 cells by providing a source of IL-4 (Yoshimoto and Paul,

1994). Thus, initial induction of IL-4 might involve several still unresolved mechanisms,

which may depend on factors such as the tissue localization of pathogenic infection or the

nature of the antigen.

While the cytokine environment can clearly modulate the fate of a naive CD4+ T cell, other

factors have also been described to influence Th development, such as nature, dose, and

localization of antigen (Constant and Bottomly, 1997). These characteristics may explain

why some pathogens preferentially induce one of the Th subsets, such that intracellular

microorganisms induce Thl responses while extracellular forms of pathogens cause Th2

differentiation. The number of antigenic epitopes available to the precursor Th cell during

priming appears to play an important role. This suggests that efficiency of uptake and

processing by APCs, which has been shown to be influenced by the initial form of the

antigen (Constant et al., 1995), may indirectly influence the development of Th cells.

Page 108: The Interplay of IgE and its High Affinity Receptor FceRI

Discussion 105

Controversial data have been reported on the effect of antigen dose on Th cell polarization,

as reviewed by Constant and Bottomly (Constant and Bottomly, 1997). While several

studies have reported that high doses of immunogen induce Thl responses and low doses

favor Th2 responses (Bancroft et al., 1994), others have shown opposite results (Wang et

al., 1996). Thus, the experimental settings of each study need to be evaluated.

Another still intriguing part of IgE-related immunity relates to the function of its high

affinity receptor, FcsRI. Although the role of FcsRI in mediating allergic reactions is well

established, other physiological functions of FcsRI are less clear. Research from the past

decade has greatly improved our knowledge of the biology of FcsRI. The main progress is

due to the discovery of major differences between the composition and cellular distribution

of human and murine FcsRI. While in mice FcsRI is expressed only on mast cells and

basophils with an obligatory tetrameric structure described by the aßy2 chain composition,

the human FcsRI can be found both as a tetramer on mast cells and basophils and

additionally as a trimer (ay2) on Langerhans' cells, monocytes, eosinophils, peripheral

blood dendritic cells, and platelets (Kinet, 1999). These findings revealed an unexpected

participation of FcsRI in antigen presentation (Novak et al., 2003a). Furthermore,

investigation of the ß chain of the high affinity IgE receptor demonstrated that ß serves as

an amplifier of FcsRI functions by enhancing the signaling cascades transduced through the

receptor (Lin et al., 1996) and by increasing FcsRI cell surface expression (Donnadieu et

al., 2000). These important functions of FcsRIß imply distinct physiological roles for the

classical tetrameric receptor complex and the trimeric isoform lacking the ß chain.

Additionally, polymorphism within the FcsRIß gene has been associated with atopic

phenotypes (Hill and Cookson, 1996). Interesting findings have also been reported on the a

chain of the receptor. FcsRIa has been proposed to be target for specific anti-FcsRIa

autoantibodies able to release histamine from basophils and mast cells leading to

development of chronic urticatia (Fiebiger et al., 1995). In particular, an imbalance between

FcsRI occupancy and natural anti-FcsRIa antibodies has been suggested to play an

important role in the pathogenesis of autoimmune urticaria (Horn et al., 2001). Another

remarkable feature of the high affinity IgE receptor is that the surface expression of FcsRI

is regulated by its own ligand, IgE. Binding of IgE to FcsRI has been shown to upregulate

surface expression of the receptor in mast cells and basophils (Lantz et al., 1997;

Page 109: The Interplay of IgE and its High Affinity Receptor FceRI

106 Discussion

Yamaguchi et al., 1997) by receptor stabilization at the membrane (Borkowski et al., 2001).

This implies that increased IgE levels may enhance the sensitivity to pathogens or allergens

due to an increased number of receptors occupied by IgE with more antigen specificities.

Finally, a role in protection against parasites has also been assigned to FcsRI. Increased

liver pathology in response to S. mansoni has been demonstrated in FcsRIa-deficient mice

(Jankovic et al., 1997) and eosinophil-mediated cytotoxicity against S. mansoni has been

shown to involve FcsRI (Gounni et al., 1994). Thus, the high affinity IgE receptor has a

very broad range of functions, of which some are not yet completely understood and

require further investigation.

5.5 Questions addressed in this thesis

The aim of this thesis was to address some of the questions regarding the role of IgE in

allergic diseases, the mechanisms involved in the regulation of IgE induction, and the

interactions of IgE and FcsRI.

Results Part I

Animal models of Th2-type conditions should provide suitable tools for investigation of the

early steps of immune responses leading to IgE production and consequently to allergic

reactions. The fur mite infestation model, described in the first part of the results, was

characterized to determine its value as a tool to investigate mechanisms involved in the

induction of IgE and to determine its suitability as a model to study allergy. The immune

response induced by a mixed population of murine fur mites, Myocoptes musculinus and

Myobia musculi, was therefore characterized. The results showed that high levels of serum

IgE induced in mice in response to mite infestation involved the same mechanisms as those

known for allergy-inducing IgE in human disease, namely Th2 cell-dependent

CD40/CD40L interactions and secretion of IL-4. The kinetics and tissue localization of

isotype switching to IgE in B cells was investigated in detail and revealed that mite

antigens appear to enter via the skin from where they are transported to skin-draining

lymph nodes, the site of T cell activation and B cell switch to IgE.

Furthermore, the IgE repertoire induced in response to mite infestation was analyzed.

Preliminary results revealed little mutations in the Vh, Dh, and Jh segments of mite-induced

IgE. This suggests that the analyzed IgE had not undergone somatic hypermutation and

Page 110: The Interplay of IgE and its High Affinity Receptor FceRI

Discussion 107

therefore may not have gone through a germinal center reaction. It has been postulated that

in certain conditions, such as allergic rhinitis, B cells can undergo class switch

recombination to IgE at the site of inflammation, i.e. outside secondary lymphoid organs

(Takhar et al., 2005). Thus, further characterization of mite-induced IgE is required to

determine whether it may result from local B cell activation.

Importantly, antigen-exposure during mite infestation occurs in a fully physiological way.

Fur mites are natural murine parasites and mice are infested via direct contact with a

previously infested mouse. Thus, no artificial injections are required and mice are exposed

to physiological antigen loads. This implies an enormous advantage of the mite infestation

model compared with other models used in allergy research, such as immunization with

high amounts of purified protein antigens in adjuvants or infection with high doses of

nematode parasites. The close resemblance of the mite infestation model to allergen

sensitization in humans, both involving exposure to antigen loads corresponding to those

present in the natural environment, makes this model a valuable tool to study the

mechanisms regulating IgE induction and development of allergic conditions.

Results Part II

The high affinity IgE receptor, FcsRI, is an important player in IgE-mediated immune

responses. The functions of FcsRI, in particular the role of its expression on cells such as

APCs and eosinophils, are not fully understood. Therefore, the second part of the present

thesis was dedicated to provide better knowledge of the biological role of FcsRI by

generating a transgenic mouse model. The aim was to generate mice that would mirror the

human distribution pattern of FcsRI. If successful, such a model should provide important

insight into the role of FcsRI expression on multiple cells in humans on which the receptor

is lacking in the murine system, including Langerhans' cells, monocytes, eosinophils,

dendritic cells, and platelets. Comparison of immune responses in WT mice with those in

animals expressing FcsRI with a human distribution should provide information on the

importance of these cells in IgE-mediated reactions. For example, the presence of FcsRI-

positive Langerhans' cells and eosinophils in a murine model should allow a novel way for

investigation of atopic dermatitis and asthma and provide new insight into the role of IgE in

these diseases. Generation of hmFcsRIaß Tg mice was based on the usage of the human

FcsRIa promoter to drive expression of the a and ß chains of the murine FcsRI with the

Page 111: The Interplay of IgE and its High Affinity Receptor FceRI

108 Discussion

cellular distribution pattern found in humans. Although mRNA expression of the

transgenes was detected in the targeted cell types of the generated transgenic mice, no

FcsRI protein expression could be shown. The lack of detectable protein expression may be

due to retention and degradation of the receptor complex in the ER or to absence of

transcription of the transgenic mRNA. Further investigation of this transgenic model, such

as confocal microscopy and sequencing of the transgenic receptor chains, is therefore

required to enable its usage for investigation of the role of FcsRI expression.

Results Part III

Investigation of FcsRI requires tools allowing precise detection of its expression. Specific

monoclonal antibodies are commonly used to analyze the expression of cell surface

molecules. As no anti-mouse FcsRIa antibodies were available, we made an attempt to

generate a specific antibody against the a chain of the murine high affinity IgE receptor.

For this purpose a fusion protein composed of the murine FcsRIa and the human IgGi Fc

portion was generated and used to immunize FcsRIa-deficient mice. However, the applied

protocol of a chain protein injections into FcsRIa-deficient mice did not result in the

expected synthesis of anti-FcsRIa antibodies. Moreover, a commercial anti-mouse FcsRIa

antibody became available during the completion of this project prompting us to cease

further investigation. Nevertheless, the successfully generated and purified muFcsRIa-

huIgGiFc fusion protein is available in large amounts and can be used at further point if

needed.

5.6 Concluding remarks

Last but not least, applying the knowledge gained from experimental animal models to the

human condition requires much care. Differences between results obtained from rodent

systems and the pathology of human allergic disease have to be evaluated to avoid

misleading conclusions. The finding that IgE may be produced locally, in the nasal mucosa

of patients with allergic rhinitis (Smurthwaite et al., 2001) and the lung mucosa of

asthmatics (Wilson et al., 2002), implies that serum IgE or skin reactivity to common

allergens might underestimate a critical role of IgE in disease. Locally limited IgE, such as

IgE content of mucosal fluids, has to be additionally investigated to fully elucidate the role

of this intriguing antibody in allergy.

Page 112: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 109

6 References

(1998). Worldwide variation in prevalence of symptoms of asthma, allergicrhinoconjunctivitis, and atopic eczema: ISAAC. The International Study of Asthma and

Allergies in Childhood (ISAAC) Steering Committee. Lancet 351, 1225-1232.

Aas, K. (1978). What makes an allergen an allergen. Allergy 33, 3-14.

Abbas, A. K., Murphy, K. M., and Sher, A. (1996). Functional diversity of helper T

lymphocytes. Nature 383, 787-793.

Aebischer, I., and Stadler, B. M. (1996). TH1-TH2 cells in allergic responses: at the limits

of a concept. Adv Immunol 61, 341-403.

Ahmad, A., Wang, C. H., and Bell, R. G. (1991). A role for IgE in intestinal immunity.Expression of rapid expulsion of Trichinella spiralis in rats transfused with IgE and thoracic

duct lymphocytes. J Immunol 146, 3563-3570.

Allen, J. E., and Maizels, R. M. (1997). Thl-Th2: reliable paradigm or dangerous dogma?Immunol Today 18, 387-392.

Anderson, C. L., Shen, L., Eicher, D. M., Wewers, M. D., and Gill, J. K. (1990).

Phagocytosis mediated by three distinct Fc gamma receptor classes on human leukocytes. J

Exp Med 777, 1333-1345.

Asai, K., Kitaura, J., Kawakami, Y., Yamagata, N., Tsai, M., Carbone, D. P., Liu, F. T.,

Galli, S. J., and Kawakami, T. (2001). Regulation of mast cell survival by IgE. Immunity14, 791-800.

Babina, M., Mammeri, K., and Henz, B. M. (1999). ICAM-3 (CD50) is expressed byhuman mast cells: induction of homotypic mast cell aggregation via ICAM-3. Cell Adhes

Commun 7, 195-209.

Bancroft, A. J., Else, K. J., and Grencis, R. K. (1994). Low-level infection with Trichuris

muris significantly affects the polarization of the CD4 response. Eur J Immunol 24, 31 IS¬

SUS.

Barbera, G., Munoz-Lopez, F., Cruz-Hernandez, M., and Torralba, A. (1977). IgEconcentration in asthmatic children. Relation to other immunoglobulins, histamine-latex

reaction, eosinophilia and skin reactivity. Allergol Immunopathol (Madr) 5, 653-658.

Bell, R. G. (1998). The generation and expression of immunity to Trichinella spiralis in

laboratory rodents. AdvParasitol 41, 149-217.

Betts, C. J., and Else, K. J. (1999). Mast cells, eosinophils and antibody-mediated cellular

cytotoxicity are not critical in resistance to Trichuris muris. Parasite Immunol 21, 45-52.

Page 113: The Interplay of IgE and its High Affinity Receptor FceRI

110 REFERENCES

Bieber, T., de la Salle, H., Wollenberg, A., Hakimi, J., Chizzonite, R., Ring, J., Hanau, D.,and de la Salle, C. (1992). Human epidermal Langerhans cells express the high affinity

receptor for immunoglobulin E (Fc epsilon RI). J Exp Med 775, 1285-1290.

Bjorksten, B., Dumitrascu, D., Foucard, T., Khetsuriani, N., Khaitov, R., Leja, M., Lis, G.,

Pekkanen, J., Priftanji, A., and Riikjarv, M. A. (1998). Prevalence of childhood asthma,rhinitis and eczema in Scandinavia and Eastern Europe. Eur Respir J 12, 432-437.

Bjorksten, B., Naaber, P., Sepp, E., and Mikelsaar, M. (1999). The intestinal microflora in

allergic Estonian and Swedish 2-year-old children. Clin Exp Allergy 29, 342-346.

Blank, U., Ra, C, Miller, L., White, K., Metzger, H., and Kinet, J. P. (1989). Completestructure and expression in transfected cells of high affinity IgE receptor. Nature 337, 187-

189.

Blank, U., Ra, C. S., and Kinet, J. P. (1991). Characterization of truncated alpha chain

products from human, rat, and mouse high affinity receptor for immunoglobulin E. J Biol

Chem 266, 2639-2646.

Borish, L., Aarons, A., Rumbyrt, J., Cvietusa, P., Negri, J., and Wenzel, S. (1996).Interleukin-10 regulation in normal subjects and patients with asthma. J Allergy Clin

Immunol 97, 1288-1296.

Borkowski, T. A., Jouvin, M. H., Lin, S. Y., and Kinet, J. P. (2001). Minimal requirementsfor IgE-mediated regulation of surface Fc epsilon RI. J Immunol 167, 1290-1296.

Boyce, J. A. (2004). The biology of the mast cell. Allergy Asthma Proc 25, 27-30.

Braun-Fahrlander, C, Gassner, M., Grize, L., Neu, U., Sennhauser, F. H., Varonier, H. S.,

Vuille, J. C, and Wuthrich, B. (1999). Prevalence of hay fever and allergic sensitization in

farmer's children and their peers living in the same rural community. SCARPOL team.

Swiss Study on Childhood Allergy and Respiratory Symptoms with Respect to Air

Pollution. Clin Exp Allergy 29, 28-34.

Burrows, B., Martinez, F. D., Halonen, M., Barbee, R. A., and Cline, M. G. (1989).Association of asthma with serum IgE levels and skin-test reactivity to allergens. N Engl J

Med 320, 271-277.

Byars, N. E., and Ferraresi, R. W. (1976). Intestinal anaphylaxis in the rat as a model of

food allergy. Clin Exp Immunol 24, 352-356.

Cameron, M. M. (1997). Can house dust mite-triggered atopic dermatitis be alleviated

using acaricides? Br J Dermatol 737, 1-8.

Capron, A., Dessaint, J. P., Capron, M., Ouma, J. H., and Butterworth, A. E. (1987).

Immunity to schistosomes: progress toward vaccine. Science 238, 1065-1072.

Chaudhuri, J., Tian, M., Khuong, C, Chua, K., Pinaud, E., and Alt, F. W. (2003).

Transcription-targeted DNA deamination by the AID antibody diversification enzyme.

Nature 422, 726-730.

Page 114: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 111

Cherayil, B. J., Weiner, S. J., and Pillai, S. (1989). The Mac-2 antigen is a galactose-

specific lectin that binds IgE. J Exp Med 770, 1959-1972.

Coffman, R. L., and Carty, J. (1986). A T cell activity that enhances polyclonal IgE

production and its inhibition by interferon-gamma. J Immunol 136, 949-954.

Cohen, S. G., and Zelaya-Quesada, M. (2002). Portier, Richet, and the discovery of

anaphylaxis: a centennial. J Allergy Clin Immunol 770, 331-336.

Constant, S., SantAngelo, D., Pasqualini, T., Taylor, T., Levin, D., Flavell, R., and

Bottomly, K. (1995). Peptide and protein antigens require distinct antigen-presenting cell

subsets for the priming of CD4+ T cells. J Immunol 154, 4915-4923.

Constant, S. L., and Bottomly, K. (1997). Induction of Thl and Th2 CD4+ T cell

responses: the alternative approaches. Annu Rev Immunol 15, 297-322.

Cooke, V.-V. (1916). Human sensitization. J Immunol 7, 201-305.

Cookson, W. (1999). The alliance of genes and environment in asthma and allergy. Nature

402, B5-11.

Coombs RRA, G. P. (1963). The classification of allergic reactions underlying disease.

Clinical aspects of Immunology, 317-337.

Cony, D. B., and Kheradmand, F. (1999). Induction and regulation of the IgE response.

Nature 402, B18-23.

Cromwell, O. (1997). Biochemistry of allergens. In: Kay AB, ed. Allergy and allergicdiseases., 797-810.

Dabbagh, K., and Lewis, D. B. (2003). Toll-like receptors and T-helper-l/T-helper-2

responses. Curr Opin Infect Dis 16, 199-204.

Daeron, M. (1997). Fc receptor biology. Annu Rev Immunol 15, 203-234.

Dawson, D. V., Whitmore, S. P., and Bresnahan, J. F. (1986). Genetic control of

susceptibility to mite-associated ulcerative dermatitis. Lab Anim Sei 36, 262-267.

de Silva, N., Brooker, S., and Hotez, P. (2003). Soil-Transmitted Helminthic Infections:

Updating the Global Picture. Disease Control Priorities Project.

de Vries, J. E., Punnonen, J., Cocks, B. G., de Waal Malefyt, R., and Aversa, G. (1993).Regulation of the human IgE response by IL4 and IL13. Res Immunol 144, 597-601.

Delespesse, G, Sarfati, M., Wu, C. Y., Fournier, S., and Letellier, M. (1992). The low-

affinity receptor for IgE. Immunol Rev 125,11-91.

Dessaint, J. P., Bout, D., Wattre, P., and Capron, A. (1975). Quantitative determination of

specific IgE antibodies to Echinococcus granulosus and IgE levels in sera from patientswith hydatid disease. Immunology 29, 813-823.

Page 115: The Interplay of IgE and its High Affinity Receptor FceRI

112 REFERENCES

Dessein, A. J., Parker, W. L., James, S. L., and David, J. R. (1981). IgE antibody and

resistance to infection. I. Selective suppression of the IgE antibody response in rats

diminishes the resistance and the eosinophil response to Trichinella spiralis infection. J ExpMed 153, 423-436.

Dold, S., Wjst, M., von Mutius, E., Reitmeir, P., and Stiepel, E. (1992). Genetic risk for

asthma, allergic rhinitis, and atopic dermatitis. Arch Dis Child 67, 1018-1022.

Dombrowicz, D., Brini, A. T., Flamand, V., Hicks, E., Snouwaert, J. N., Kinet, J. P., and

Koller, B. H. (1996). Anaphylaxis mediated through a humanized high affinity IgE

receptor. J Immunol 757, 1645-1651.

Dombrowicz, D., Flamand, V., Brigman, K. K., Koller, B. H., and Kinet, J. P. (1993).Abolition of anaphylaxis by targeted disruption of the high affinity immunoglobulin E

receptor alpha chain gene. Cell 75, 969-976.

Dombrowicz, D., Lin, S., Flamand, V., Brini, A. T., Koller, B. H., and Kinet, J. P. (1998).

Allergy-associated FcRbeta is a molecular amplifier of IgE- and IgG-mediated in vivo

responses. Immunity 8, 517-529.

Donnadieu, E., Jouvin, M. H., and Kinet, J. P. (2000). A second amplifier function for the

allergy-associated Fc(epsilon)RI-beta subunit. Immunity 12, 515-523.

Duffy, D. L., Martin, N. G., Battistutta, D., Hopper, J. L., and Mathews, J. D. (1990).Genetics of asthma and hay fever in Australian twins. Am Rev Respir Dis 142, 1351-1358.

Durmaz, B., Yakinci, C, Koroglu, M., Rafiq, M., and Durmaz, R. (1998). Concentration of

total serum IgE in parasitized children and the effects of the antiparasitic therapy on IgElevels. J Trop Pediatr 44, 121.

Ermel, R. W., Kock, M., Griffey, S. M., Reinhart, G A., and Frick, O. L. (1997). The

atopic dog: a model for food allergy. Lab Anim Sei 47, 40-49.

Fiebiger, E., Maurer, D., Holub, H., Reininger, B., Hartmann, G., Woisetschlager, M.,

Kinet, J. P., and Stingl, G. (1995). Serum IgG autoantibodies directed against the alphachain of Fc epsilon RI: a selective marker and pathogenetic factor for a distinct subset of

chronic urticaria patients? J Clin Invest 96, 2606-2612.

Finkelman, F. D., Katona, I. M., Urban, J. F., Jr., Holmes, J., Ohara, J., Tung, A. S.,

Sample, J. V., and Paul, W. E. (1988). IL-4 is required to generate and sustain in vivo IgE

responses. J Immunol 141, 2335-2341.

Finkelman, F. D., Shea-Donohue, T., Goldhill, J., Sullivan, C. A., Morris, S. C, Madden,K. B., Gause, W. C, and Urban, J. F., Jr. (1997). Cytokine regulation of host defense

against parasitic gastrointestinal nematodes: lessons from studies with rodent models. Annu

Rev Immunol 75, 505-533.

Fox, C. C, Jewell, S. D., and Whitacre, C. C. (1994). Rat peritoneal mast cells present

antigen to a PPD-specific T cell line. Cell Immunol 755, 253-264.

Page 116: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 113

Frandji, P., Oskeritzian, C, Cacaraci, F., Lapeyre, J., Peronet, R., David, B., Guillet, J. G.,and Mecheri, S. (1993). Antigen-dependent stimulation by bone marrow-derived mast cells

ofMHC class II-restricted T cell hybridoma. J Immunol 757, 6318-6328.

Frandji, P., Tkaczyk, C, Oskeritzian, C, David, B., Desaymard, C, and Mecheri, S.

(1996). Exogenous and endogenous antigens are differentially presented by mast cells to

CD4+ T lymphocytes. Eur J Immunol 26, 2517-2528.

Friedman, S., and Weisbroth, S. H. (1975). The parasitic ecology of the rodent mite Myobiamusculi. II. Genetic factors. Lab Anim Sei 25, 440-445.

Friedman, S., and Weisbroth, S. H. (1977). The parasitic ecology of the rodent mite,

Myobia musculi. IV. Life cycle. Lab Anim Sei 27, 34-37.

Frigeri, L. G., and Liu, F. T. (1992). Surface expression of functional IgE binding protein,an endogenous lectin, on mast cells and macrophages. J Immunol 148, 861-867.

Gambles, R. (1952). Myocoptes musculinus (Koch) and Myobia musculi (Schrank), two

species of mite commonly parasitizing the laboratory mouse. Br Vet J 705, 194-203.

Garman, S. C, Kinet, J. P., and Jardetzky, T. S. (1998). Crystal structure of the human

high-affinity IgE receptor. Cell 95, 951-961.

Garman, S. C, Wurzburg, B. A., Tarchevskaya, S. S., Kinet, J. P., and Jardetzky, T. S.

(2000). Structure of the Fc fragment of human IgE bound to its high-affinity receptor Fc

epsilonRI alpha. Nature 406, 259-266.

Gauchat, J. F., Henchoz, S., Mazzei, G., Aubry, J. P., Brunner, T., Blasey, H., Life, P.,

Talabot, D., Flores-Romo, L., Thompson, J., and et al. (1993). Induction of human IgE

synthesis in B cells by mast cells and basophils. Nature 365, 340-343.

Geha, R. S., Jabara, H. H., and Brodeur, S. R. (2003). The regulation of immunoglobulin E

class-switch recombination. Nat Rev Immunol 3, 721-732.

Gor, D. O., Rose, N. R., and Greenspan, N. S. (2003). TH1-TH2: a procrustean paradigm.Nat Immunol 4, 503-505.

Gould, H. J., Sutton, B. J., Beavil, A. J., Beavil, R. L., McCloskey, N., Coker, H. A., Fear,

D., and Smurthwaite, L. (2003). The biology of IGE and the basis of allergic disease. Annu

Rev Immunol 21, 579-628.

Gounni, A. S., Lamkhioued, B., Ochiai, K., Tanaka, Y., Delaporte, E., Capron, A., Kinet, J.

P., and Capron, M. (1994). High-affinity IgE receptor on eosinophils is involved in defence

against parasites. Nature 367, 183-186.

Grunewald, S. M., Teufel, M., Erb, K., Neide, A., Mohrs, M., Brombacher, F., Brocker, E.

B., Sebald, W., and Duschl, A. (2001). Upon prolonged allergen exposure IL-4 and IL-

4Ralpha knockout mice produce specific IgE leading to anaphylaxis. Int Arch AllergyImmunol 725, 322-328.

Page 117: The Interplay of IgE and its High Affinity Receptor FceRI

114 REFERENCES

Gurish, M. F., Bryce, P. J., Tao, H., Kisselgof, A. B., Thornton, E. M., Miller, H. R.,

Friend, D. S., and Oettgen, H. C. (2004). IgE enhances parasite clearance and regulatesmast cell responses in mice infected with Trichinella spiralis. J Immunol 772, 1139-1145.

Haas, L. F. (2001). Charles Robert Richet (1850-1935). J Neurol Neurosurg Psychiatry 70,255.

Hagan, P. (1993). IgE and protective immunity to helminth infections. Parasite Immunol

75, 1-4.

Hagan, P., Blumenthal, U. J., Dunn, D., Simpson, A. J., and Wilkins, H. A. (1991). Human

IgE, IgG4 and resistance to reinfection with Schistosoma haematobium. Nature 349, 243-

245.

Hasegawa, S., Pawankar, R., Suzuki, K., Nakahata, T., Furukawa, S., Okumura, K., and Ra,C. (1999). Functional expression of the high affinity receptor for IgE (FcepsilonRI) in

human platelets and its' intracellular expression in human megakaryocytes. Blood 93, 2543-

2551.

Herz, U., Renz, H., and Wiedermann, U. (2004). Animal models of type I allergy usingrecombinant allergens. Methods 32, 271-280.

Hill, M. R., and Cookson, W. O. (1996). A new variant of the beta subunit of the high-

affinity receptor for immunoglobulin E (Fc epsilon RI-beta E237G): associations with

measures of atopy and bronchial hyper-responsiveness. Hum Mol Genet 5, 959-962.

Holgate, S. T. (1999). The epidemic of allergy and asthma. Nature 402, B2-4.

Holt, P. G, Clough, J. B., Holt, B. J., Baron-Hay, M. J., Rose, A. H., Robinson, B. W., and

Thomas, W. R. (1992). Genetic 'risk' for atopy is associated with delayed postnatalmaturation of T-cell competence. Clin Exp Allergy 22, 1093-1099.

Horn, M. P., Pachlopnik, J. M., Vogel, M., Dahinden, M., Wurm, F., Stadler, B. M., and

Miescher, S. M. (2001). Conditional autoimmunity mediated by human natural anti-

Fc(epsilon)RIalpha autoantibodies? Faseb J 75, 2268-2274.

Iciek, L. A., Delphin, S. A., and Stavnezer, J. (1997). CD40 cross-linking induces Ig

epsilon germline transcripts in B cells via activation of NF-kappaB: synergy with IL-4

induction. J Immunol 755, 4769-4779.

Iijima, O. T., Takeda, H., Komatsu, Y., Matsumiya, T., and Takahashi, H. (2000). Atopicdermatitis in NC/Jic mice associated with Myobia musculi infestation. Comp Med 50, 225-

228.

Ishizaka, K., and Ishizaka, T. (1967). Identification of gamma-E-antibodies as a carrier of

reaginic activity. J Immunol 99, 1187-1198.

Ishizaka, T., Helm, B., Hakimi, J., Niebyl, J., Ishizaka, K., and Gould, H. (1986).

Biological properties of a recombinant human immunoglobulin epsilon-chain fragment.Proc Natl Acad Sei U S A 83, 8323-8327.

Page 118: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 115

Jabara, H. H., Schneider, L. C, Shapira, S. K., Alfieri, C, Moody, C. T., Kieff, E., Geha,R. S., and Vercelli, D. (1990). Induction of germ-line and mature C epsilon transcripts in

human B cells stimulated with rIL-4 and EBV. J Immunol 145, 3468-3473.

Jankovic, D., Kullberg, M. C, Dombrowicz, D., Barbieri, S., Caspar, P., Wynn, T. A., Paul,W. E., Cheever, A. W., Kinet, J. P., and Sher, A. (1997). Fc epsilonRI-deficient mice

infected with Schistosoma mansoni mount normal Th2-type responses while displayingenhanced liver pathology. J Immunol 759, 1868-1875.

Jarrett, E. E., and Miller, H. R. (1982). Production and activities of IgE in helminth

infection. Prog Allergy 31, 178-233.

Johansson, S. G., andBennich, H. (1967). Immunological studies of an atypical (myeloma)immunoglobulin. Immunology 13, 381-394.

Jones, A. C, Miles, E. A., Warner, J. O., Colwell, B. M., Bryant, T. N., and Warner, J. A.

(1996). Fetal peripheral blood mononuclear cell proliferative responses to mitogenic and

allergenic stimuli during gestation. Pediatr Allergy Immunol 7, 109-116.

Joseph, M., Gounni, A. S., Kusnierz, J. P., Vorng, H., Sarfati, M., Kinet, J. P., Tonnel, A.

B., Capron, A., and Capron, M. (1997). Expression and functions of the high-affinity IgE

receptor on human platelets and megakaryocyte precursors. Eur J Immunol 27, 2212-2218.

Jouvin, M. H., Adamczewski, M., Numerof, R., Letourneur, O., Valle, A., and Kinet, J. P.

(1994). Differential control of the tyrosine kinases Lyn and Syk by the two signaling chains

of the high affinity immunoglobulin E receptor. J Biol Chem 269, 5918-5925.

Jungmann, P., Freitas, A., Bandeira, A., Nobrega, A., Coutinho, A., Marcos, M. A., and

Minoprio, P. (1996a). Murine acariasis. II. Immunological dysfunction and evidence for

chronic activation of Th-2 lymphocytes. Scand J Immunol 43, 604-612.

Jungmann, P., Guenet, J. L., Cazenave, P. A., Coutinho, A., and Huerre, M. (1996b).Murine acariasis: I. Pathological and clinical evidence suggesting cutaneous allergy and

wasting syndrome in BALB/c mouse. Res Immunol 147, 27-38.

Jürgens, M., Wollenberg, A., Hanau, D., de la Salle, H., and Bieber, T. (1995). Activation

of human epidermal Langerhans cells by engagement of the high affinity receptor for IgE,Fc epsilon RI. J Immunol 755, 5184-5189.

Kalesnikoff, J., Huber, M., Lam, V., Damen, J. E., Zhang, J., Siraganian, R. P., and Krystal,G. (2001). Monomeric IgE stimulates signaling pathways in mast cells that lead to cytokine

production and cell survival. Immunity 14, 801-811.

Kawakami, T., and Galli, S. J. (2002). Regulation of mast-cell and basophil function and

survival by IgE. Nat Rev Immunol 2, 773-786.

Kay, A. B. (2001). Allergy and allergic diseases. First of two parts. N Engl J Med 344, 30-

37.

Page 119: The Interplay of IgE and its High Affinity Receptor FceRI

116 REFERENCES

Kinet, J. P. (1999). The high-affinity IgE receptor (Fc epsilon RI): from physiology to

pathology. Annu Rev Immunol 77, 931-972.

Kinet, J. P., Blank, U., Ra, C, White, K., Metzger, H., and Kochan, J. (1988). Isolation and

characterization of cDNAs coding for the beta subunit of the high-affinity receptor for

immunoglobulin E. Proc Natl Acad Sei U S A 55, 6483-6487.

Kitaura, J., Song, J., Tsai, M., Asai, K., Maeda-Yamamoto, M., Mocsai, A., Kawakami, Y.,

Liu, F. T., Lowell, C. A., Barisas, B. G., etal. (2003). Evidence that IgE molecules mediate

a spectrum of effects on mast cell survival and activation via aggregation of the

FcepsilonRI. Proc Natl Acad Sei U S A 700, 12911-12916.

Kopf, M., Brombacher, F., Hodgkin, P. D., Ramsay, A. J., Milbourne, E. A., Dai, W. J.,

Ovington, K. S., Behm, C. A., Kohler, G., Young, I. G, and Matthaei, K. I. (1996). IL-5-

deficient mice have a developmental defect in CD5+ B-l cells and lack eosinophilia but

have normal antibody and cytotoxic T cell responses. Immunity 4, 15-24.

Kopf, M., Le Gros, G., Bachmann, M., Lamers, M. C, Bluethmann, H., and Köhler, G.

(1993). Disruption of the murine IL-4 gene blocks Th2 cytokine responses. Nature 362,245-248.

Kraft, S., Novak, N., Katoh, N., Bieber, T., and Rupee, R. A. (2002a). Aggregation of the

high-affinity IgE receptor Fc(epsilon)RI on human monocytes and dendritic cells induces

NF-kappaB activation. J Invest Dermatol 775, 830-837.

Kraft, S., Wessendorf, J. H., Haberstok, J., Novak, N., Wollenberg, A., and Bieber, T.

(2002b). Enhanced expression and activity of protein-tyrosine kinases establishes a

functional signaling pathway only in FcepsilonRIhigh Langerhans cells from atopicindividuals. J Invest Dermatol 779, 804-811.

Kraft, S., Wessendorf, J. H., Hanau, D., and Bieber, T. (1998). Regulation of the highaffinity receptor for IgE on human epidermal Langerhans cells. J Immunol 161, 1000-1006.

Krebber, A., Bornhauser, S., Burmester, J., Honegger, A., Willuda, J., Bosshard, H. R., and

Pluckthun, A. (1997). Reliable cloning of functional antibody variable domains from

hybridomas and spleen cell repertoires employing a reengineered phage display system. J

Immunol Methods 207, 35-55.

Kubo, S., Matsuoka, K., Taya, C, Kitamura, F., Takai, T., Yonekawa, H., and Karasuyama,H. (2001). Drastic up-regulation of Fcepsilonri on mast cells is induced by IgE binding

through stabilization and accumulation of Fcepsilonri on the cell surface. J Immunol 167,3427-3434.

Kulczycki, A., Jr., and Metzger, H. (1974). The interaction of IgE with rat basophilicleukemia cells. II. Quantitative aspects of the binding reaction. J Exp Med 140, 1676-1695.

Küster, H., Thompson, H., and Kinet, J. P. (1990). Characterization and expression of the

gene for the human Fc receptor gamma subunit. Definition of a new gene family. J Biol

Chem 265, 6448-6452.

Page 120: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 117

Laltoo, H., Van Zoost, T., and Kind, L. S. (1979). IgE antibody response to mite antigens in

mite infested mice. Immunol Commun 5, 1-9.

Lantz, C. S., Yamaguchi, M., Oettgen, H. C, Katona, I. M., Miyajima, L, Kinet, J. P., and

Galli, S. J. (1997). IgE regulates mouse basophil Fc epsilon RI expression in vivo. J

Immunol 755,2517-2521.

Laske, N., and Niggemann, B. (2004). Does the severity of atopic dermatitis correlate with

serum IgE levels? Pediatr Allergy Immunol 75, 86-88.

Leonard, W. J. (2002). TSLP: finally in the limelight. Nat Immunol 3, 605-607.

Letourneur, F., Hennecke, S., Demolliere, C, and Cosson, P. (1995a). Steric masking of a

dilysine endoplasmic reticulum retention motif during assembly of the human high affinity

receptor for immunoglobulin E. J Cell Biol 729, 971-978.

Letourneur, O., Sechi, S., Willette-Brown, J., Robertson, M. W., and Kinet, J. P. (1995b).Glycosylation of human truncated Fc epsilon RI alpha chain is necessary for efficient

folding in the endoplasmic reticulum. J Biol Chem 270, 8249-8256.

Levine, B. B., and Vaz, N. M. (1970). Effect of combinations of inbred strain, antigen, and

antigen dose on immune responsiveness and reagin production in the mouse. A potentialmouse model for immune aspects of human atopic allergy. Int Arch Allergy Appl Immunol

39, 156-171.

Levy, D. A. (2004). Parasites and Allergy: From IgE to Thl/Th2 and Beyond. Clin Rev

Allergy Immunol 26, 1-4.

Liew, F. Y. (2002). T(H)1 and T(H)2 cells: a historical perspective. Nat Rev Immunol 2,55-60.

Lin, S., Cicala, C, Scharenberg, A. M., and Kinet, J. P. (1996). The Fc(epsilon)RIbetasubunit functions as an amplifier of Fc(epsilon)RIgamma-mediated cell activation signals.Cell 55, 985-995.

Litinskiy, M. B., Nardelli, B., Hubert, D. M., He, B., Schaffer, A., Casali, P., and Cerutti,A. (2002). DCs induce CD40-independent immunoglobulin class switching through BLySand APRIL. Nat Immunol 3, 822-829.

Liu, Z. J., Haleem-Smith, H., Chen, H., and Metzger, H. (2001). Unexpected signals in a

system subject to kinetic proofreading. Proc Natl Acad Sei U S A 95, 7289-7294.

Lloyd, C. M., Gonzalo, J. A., Coyle, A. J., and Gutierrez-Ramos, J. C. (2001). Mouse

models of allergic airway disease. Adv Immunol 77, 263-295.

Maenaka, K., van derMerwe, P. A., Stuart, D. I., Jones, E. Y., and Sondermann, P. (2001).The human low affinity Fcgamma receptors IIa, IIb, and III bind IgG with fast kinetics and

distinct thermodynamic properties. J Biol Chem 276, 44898-44904.

Page 121: The Interplay of IgE and its High Affinity Receptor FceRI

118 REFERENCES

Malaviya, R., Twesten, N. J., Ross, E. A., Abraham, S. N., and Pfeifer, J. D. (1996). Mast

cells process bacterial Ags through a phagocytic route for class IMHC presentation to T

cells. J Immunol 156, 1490-1496.

Marsh, D. G, Hsu, S. H., Roebber, M., Ehrlich-Kautzky, E., Freidhoff, L. R., Meyers, D.

A., Pollard, M. K., and Bias, W. B. (1982). HLA-Dw2: a genetic marker for human

immune response to short ragweed pollen allergen Ra5.1. Response resulting primarilyfrom natural antigenic exposure. J Exp Med 755, 1439-1451.

Marsh, D. G, Neely, J. D., Breazeale, D. R., Ghosh, B., Freidhoff, L. R., Ehrlich-Kautzky,E., Schou, C, Krishnaswamy, G., and Beaty, T. H. (1994). Linkage analysis of IL4 and

other chromosome 5q31.1 markers and total serum immunoglobulin E concentrations.

Science 264, 1152-1156.

Matricardi, P. M., Rosmini, F., Riondino, S., Fortini, M., Ferrigno, L., Rapicetta, M., and

Bonini, S. (2000). Exposure to foodborne and orofecal microbes versus airborne viruses in

relation to atopy and allergic asthma: epidemiological study. Bmj 320, 412-417'.

Matsuda, H., Watanabe, N., Geba, G. P., Sperl, J., Tsudzuki, M., Hiroi, J., Matsumoto, M.,

Ushio, H., Saito, S., Askenase, P. W., and Ra, C. (1997). Development of atopic dermatitis¬

like skin lesion with IgE hyperproduction in NC/Nga mice. Int Immunol 9, 461-466.

Maurer, D., Ebner, C, Reininger, B., Fiebiger, E., Kraft, D., Kinet, J. P., and Stingl, G.

(1995). The high affinity IgE receptor (Fc epsilon RI) mediates IgE-dependent allergen

presentation. J Immunol 154, 6285-6290.

Maurer, D., Fiebiger, E., Reininger, B., Ebner, C, Petzelbauer, P., Shi, G. P., Chapman, H.

A., and Stingl, G. (1998). Fc epsilon receptor I on dendritic cells delivers IgE-boundmultivalent antigens into a cathepsin S-dependent pathway ofMHC class II presentation. J

Immunol 161,2131-2139.

Maurer, D., Fiebiger, E., Reininger, B., Wolff-Winiski, B., Jouvin, M. H., Kilgus, O.,

Kinet, J. P., and Stingl, G. (1994). Expression of functional high affinity immunoglobulin E

receptors (Fc epsilon RI) on monocytes of atopic individuals. J Exp Med 779, 745-750.

Maurer, D., Fiebiger, S., Ebner, C, Reininger, B., Fischer, G. F., Wichlas, S., Jouvin, M.

H., Schmitt-Egenolf, M., Kraft, D., Kinet, J. P., and Stingl, G. (1996). Peripheral blood

dendritic cells express Fc epsilon RI as a complex composed of Fc epsilon RI alpha- and Fc

epsilon RI gamma-chains and can use this receptor for IgE-mediated allergen presentation.J Immunol 757, 607-616.

Messner, B., Stutz, A. M., Albrecht, B., Peiritsch, S., and Woisetschlager, M. (1997).

Cooperation of binding sites for STAT6 and NF kappa B/rel in the IL-4-induced up¬

regulation of the human IgE germline promoter. J Immunol 759, 3330-3337.

Metzger, H., Alcaraz, G., Hohman, R., Kinet, J. P., Pribluda, V., and Quarto, R. (1986).The receptor with high affinity for immunoglobulin E. Annu Rev Immunol 4, 419-470.

Page 122: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 119

Min, B., Prout, M., Hu-Li, J., Zhu, J., Jankovic, D., Morgan, E. S., Urban, J. F., Jr., Dvorak,A. M., Finkelman, F. D., LeGros, G., and Paul, W. E. (2004). Basophils produce IL-4 and

accumulate in tissues after infection with a Th2-inducing parasite. J Exp Med 200, 507-517.

Minty, A., Chalon, P., Derocq, J. M., Dumont, X., Guillemot, J. C, Kaghad, M., Labit, C,

Leplatois, P., Liauzun, P., Miloux, B., and et al. (1993). Interleukin-13 is a new human

lymphokine regulating inflammatory and immune responses. Nature 362, 248-250.

Mohapatra, S. S., and Lockey, R. F. (2001). Molecular characterization of allergens. Clin

Rev Allergy Immunol 27, 203-213.

Mombaerts, P., Mizoguchi, E., Grusby, M. J., Glimcher, L. H., Bhan, A. K., and Tonegawa,S. (1993). Spontaneous development of inflammatory bowel disease in T cell receptormutant mice. Cell 75, 274-282.

Moore, K. W., de Waal Malefyt, R., Coffman, R. L., and O'Garra, A. (2001). Interleukin-10

and the interleukin-10 receptor. Annu Rev Immunol 79, 683-765.

Morawetz, R. A., Gabriele, L., Rizzo, L. V., Noben-Trauth, N., Kuhn, R., Rajewsky, K.,

Müller, W., Doherty, T. M., Finkelman, F., Coffman, R. L., and Morse, H. C, 3rd (1996).Interleukin (IL)-4-independent immunoglobulin class switch to immunoglobulin (Ig)E in

the mouse. J Exp Med 184, 1651-1661.

Morita, E., Kaneko, S., Hiragun, T., Shindo, H., Tanaka, T., Furukawa, T., Nobukiyo, A.,and Yamamoto, S. (1999). Fur mites induce dermatitis associated with IgE hyperproductionin an inbred strain of mice, NC/Kuj. J Dermatol Sei 79, 37-43.

Muramatsu, M., Kinoshita, K., Fagarasan, S., Yamada, S., Shinkai, Y., and Honjo, T.

(2000). Class switch recombination and hypermutation require activation-induced cytidinedeaminase (AID), a potential RNA editing enzyme. Cell 702, 553-563.

Muramatsu, M., Sankaranand, V. S., Anant, S., Sugai, M., Kinoshita, K., Davidson, N. O.,and Honjo, T. (1999). Specific expression of activation-induced cytidine deaminase (AID),a novel member of the RNA-editing deaminase family in germinal center B cells. J Biol

Chem 274, 18470-18476.

Negrao-Correa, D. (2001). Importance of immunoglobulin E (IgE) in the protectivemechanism against gastrointestinal nematode infection: looking at the intestinal mucosae.

Rev Inst Med Trop Sao Paulo 43, 291-299.

Negrao-Correa, D., Adams, L. S., and Bell, R. G. (1996). Intestinal transport and

catabolism of IgE: a major blood-independent pathway of IgE dissemination during a

Trichinella spiralis infection of rats. J Immunol 757, 4037-4044.

Neuhaus-Steinmetz, U., Glaab, T., Daser, A., Braun, A., Lommatzsch, M., Herz, U., Kips,J., Alarie, Y., and Renz, H. (2000). Sequential development of airway hyperresponsivenessand acute airway obstruction in a mouse model of allergic inflammation. Int Arch AllergyImmunol 727, 57-67.

Novak, N., and Bieber, T. (2002). To bend or not to bend. Nat Immunol 3, 607-608.

Page 123: The Interplay of IgE and its High Affinity Receptor FceRI

120 REFERENCES

Novak, N., Kraft, S., and Bieber, T. (2003a). Unraveling the mission of FcepsilonRI on

antigen-presenting cells. J Allergy Clin Immunol 777, 38-44.

Novak, N., Tepel, C, Koch, S., Brix, K., Bieber, T., and Kraft, S. (2003b). Evidence for a

differential expression of the FcepsilonRIgamma chain in dendritic cells of atopic and

nonatopic donors. J Clin Invest 777, 1047-1056.

Ogilvie, B. M. (1964). Reagin-Like Antibodies In Animals Immune To Helminth Parasites.

Nature 204, 91-92.

Perez-Montfort, R., Kinet, J. P., and Metzger, H. (1983). A previously unrecognizedsubunit of the receptor for immunoglobulin E. Biochemistry 22, 5722-5728.

Phalipon, A., and Corthesy, B. (2003). Novel functions of the polymeric Ig receptor: well

beyond transport of immunoglobulins. Trends Immunol 24, 55-58.

Phillips, N. E., and Parker, D. C. (1983). Fc-dependent inhibition of mouse B cell

activation by whole anti-mu antibodies. J Immunol 730, 602-606.

Plaut, M., Pierce, J. H., Watson, C. J., Hanley-Hyde, J., Nordan, R. P., and Paul, W. E.

(1989). Mast cell lines produce lymphokines in response to cross-linkage of Fc epsilon RI

or to calcium ionophores. Nature 339, 64-67'.

Prausnitz, C. (1921). Studie über die Ueberempfindlichkeit. Zentralbl Bakteriol 86, 160-

169.

Prescott, S. L., Macaubas, C, Holt, B. J., Smallacombe, T. B., Loh, R., Sly, P. D., and Holt,P. G. (1998). Transplacental priming of the human immune system to environmental

allergens: universal skewing of initial T cell responses toward the Th2 cytokine profile. J

Immunol 160, 4730-4737.

Priquet, v. (1963). Allergic In: Gell PGH, Coombs RRA, eds. Clinical aspects of

immunology.

Pritchard, D. I. (1993). Immunity to helminths: is too much IgE parasite—rather than host-

protective? Parasite Immunol 75, 5-9.

Prussin, C, and Metcalfe, D. D. (2003). 4. IgE, mast cells, basophils, and eosinophils. J

Allergy Clin Immunol 777, S486-494.

Pruzansky, J. J., and Patterson, R. (1986). Binding constants of IgE receptors on human

blood basophils for IgE. Immunology 55, 257-262.

Punnonen, J., Aversa, G., Cocks, B. G., McKenzie, A. N., Menon, S., Zurawski, G., de

Waal Malefyt, R., and de Vries, J. E. (1993). Interleukin 13 induces interleukin 4-

independent IgG4 and IgE synthesis and CD23 expression by human B cells. Proc Natl

Acad Sei U S A 90, 3730-3734.

Page 124: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 121

Ra, C, Jouvin, M. H., and Kinet, J. P. (1989). Complete structure of the mouse mast cell

receptor for IgE (Fc epsilon RI) and surface expression of chimeric receptors (rat-mouse-human) on transfected cells. J Biol Chem 264, 15323-15327.

Ravetch, J. V., and Bolland, S. (2001). IgG Fc receptors. Annu Rev Immunol 79, 275-290.

Regnault, A., Lankar, D., Lacabanne, V., Rodriguez, A., Thery, C, Rescigno, M., Saito, T.,

Verbeek, S., Bonnerot, C, Ricciardi-Castagnoli, P., and Amigorena, S. (1999). Fcgammareceptor-mediated induction of dendritic cell maturation and major histocompatibility

complex class I-restricted antigen presentation after immune complex internalization. J ExpMed 759, 371-380.

Renshaw, B. R., Fanslow, W. C, 3rd, Armitage, R. J., Campbell, K. A., Liggitt, D., Wright,B., Davison, B. L., and Maliszewski, C. R. (1994). Humoral immune responses in CD40

ligand-deficient mice. J Exp Med 750, 1889-1900.

Rigby, L. J., Trist, H., Snider, J., Hulett, M. D., Hogarth, P. M., and Epa, V. C. (2000).Monoclonal antibodies and synthetic peptides define the active site of FcepsilonRI and a

potential receptor antagonist. Allergy 55, 609-619.

Rihet, P., Demeure, C. E., Bourgois, A., Prata, A., and Dessein, A. J. (1991). Evidence for

an association between human resistance to Schistosoma mansoni and high anti-larval IgElevels. Eur J Immunol 27, 2679-2686.

Riske, F., Hakimi, J., Mallamaci, M., Griffin, M., Pilson, B., Tobkes, N., Lin, P., Danho,

W., Kochan, J., and Chizzonite, R. (1991). High affinity human IgE receptor (Fc epsilonRI). Analysis of functional domains of the alpha-subunit with monoclonal antibodies. J

Biol Chem 266, 11245-11251.

Rivera, J., Kinet, J. P., Kim, J., Pucillo, C, and Metzger, H. (1988). Studies with a

monoclonal antibody to the beta subunit of the receptor with high affinity for

immunoglobulin E. Mol Immunol 25, 647-661.

Rook, G. A., and Stanford, J. L. (1998). Give us this day our daily germs. Immunol Today79, 113-116.

Schmitz, J., Thiel, A., Kuhn, R., Rajewsky, K., Muller, W., Assenmacher, M., and

Radbruch, A. (1994). Induction of interleukin 4 (IL-4) expression in T helper (Th) cells is

not dependent on IL-4 from non-Th cells. J Exp Med 779, 1349-1353.

Schüler, G., and Steinman, R. M. (1985). Murine epidermal Langerhans cells mature into

potent immunostimulatory dendritic cells in vitro. J Exp Med 161, 526-546.

Seder, R. A., Paul, W. E., Dvorak, A. M., Sharkis, S. J., Kagey-Sobotka, A., Niv, Y.,

Finkelman, F. D., Barbieri, S. A., Galli, S. J., and Plaut, M. (1991). Mouse splenic and bone

marrow cell populations that express high-affinity Fc epsilon receptors and produceinterleukin 4 are highly enriched in basophils. Proc Natl Acad Sei U S A 55, 2835-2839.

Sepp, E., Julge, K., Vasar, M., Naaber, P., Bjorksten, B., and Mikelsaar, M. (1997).Intestinal microflora of Estonian and Swedish infants. Acta Paediatr 86, 956-961.

Page 125: The Interplay of IgE and its High Affinity Receptor FceRI

122 REFERENCES

Shea-Donohue, T., and Urban, J. F., Jr. (2004). Gastrointestinal parasite and host

interactions. Curr Opin Gastroenterol 20, 3-9.

Shin, E. H., Osada, Y., Chai, J. Y., Matsumoto, N., Takatsu, K., and Kojima, S. (1997).Protective roles of eosinophils in Nippostrongylus brasiliensis infection. Int Arch AllergyImmunol 114 Suppl 1, 45-50.

Simister, N. E., and Mostov, K. E. (1989). An Fc receptor structurally related to MHC class

I antigens. Nature 337, 184-187.

Smith, D. H., Malone, D. C, Lawson, K. A., Okamoto, L. J., Battista, C, and Saunders, W.

B. (1997). A national estimate of the economic costs of asthma. Am J Respir Crit Care Med

156, 787-793.

Smurthwaite, L., Walker, S. N., Wilson, D. R., Birch, D. S., Merrett, T. G, Durham, S. R.,and Gould, H. J. (2001). Persistent IgE synthesis in the nasal mucosa of hay fever patients.Eur J Immunol 37, 3422-3431.

Soumelis, V., Reche, P. A., Kanzler, H., Yuan, W., Edward, G., Homey, B., Gilliet, M.,

Ho, S., Antonenko, S., Lauerma, A., etal. (2002). Human epithelial cells trigger dendritic

cell mediated allergic inflammation by producing TSLP. Nat Immunol 3, 673-680.

Sporik, R., Holgate, S. T., Platts-Mills, T. A., and Cogswell, J. J. (1990). Exposure to

house-dust mite allergen (Der p I) and the development of asthma in childhood. A

prospective study. N Engl J Med 323, 502-507.

Stewart, G. A., Thompson, P. J., and McWilliam, A. S. (1993). Biochemical properties of

aeroallergens: contributory factors in allergic sensitization? Pediatr Allergy Immunol 4,163-172.

Strachan, D. P. (1989). Hay fever, hygiene, and household size. Bmj 299, 1259-1260.

Sunyer, J., Anto, J. M., Tobias, A., and Burney, P. (1999). Generational increase of self-

reported first attack of asthma in fifteen industrialized countries. European Community

Respiratory Health Study (ECRHS). Eur Respir J 14, 885-891.

Sutton, B. J., and Gould, H. J. (1993). The human IgE network. Nature 366, 421-428.

Tada, T., Okumura, K., Platteau, B., Beckers, A., and Bazin, H. (1975). Half-lives of two

types of rat homocytotropic antibodies in circulation and in the skin. Int Arch Allergy ApplImmunoW5, 116-131.

Takai, T. (2002). Roles of Fc receptors in autoimmunity. Nat Rev Immunol 2, 580-592.

Takhar, P., Smurthwaite, L., Coker, H. A., Fear, D. J., Banfield, G. K., Carr, V. A.,

Durham, S. R., and Gould, H. J. (2005). Allergen drives class switching to IgE in the nasal

mucosa in allergic rhinitis. J Immunol 174, 5024-5032.

Tan, B. B., Weald, D., Strickland, I., and Friedmann, P. S. (1996). Double-blind controlled

trial of effect of housedust-mite allergen avoidance on atopic dermatitis. Lancet 347, 15-18.

Page 126: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 123

Tanaka, K., Okamoto, Y., Nagaya, Y., Nishimura, F., Takeoka, A., Hanada, S., Kohno, S.,and Kawai, M. (1988). A nasal allergy model developed in the guinea pig by intranasal

application of 2,4-toluene diisocyanate. Int Arch Allergy Appl Immunol 55, 392-397.

Tang, M. L., Kemp, A. S., Thorburn, J., and Hill, D. J. (1994). Reduced interferon-gammasecretion in neonates and subsequent atopy. Lancet 344, 983-985.

Toru, H., Kinashi, T., Ra, C, Nonoyama, S., Yata, J., and Nakahata, T. (1997). Interleukin-

4 induces homotypic aggregation of human mast cells by promoting LFA-l/ICAM-1

adhesion molecules. Blood 59, 3296-3302.

Turner, C. R., Andresen, C. J., Smith, W. B., and Watson, J. W. (1996). Characterization of

a primate model of asthma using anti-allergy/anti-asthma agents. Inflamm Res 45, 239-245.

Turner, H., and Kinet, J. P. (1999). Signalling through the high-affinity IgE receptor Fc

epsilonRI. Nature 402, B24-30.

Turner, K. J., Feddema, L., and Quinn, E. H. (1979). Non-specific potentiation of IgE by

parasitic infections in man. Int Arch Allergy Appl Immunol 55, 232-236.

Valent, P., Bevec, D., Maurer, D., Besemer, J., Di Padova, F., Butterfield, J. H., Speiser,W., Majdic, O., Lechner, K., and Bettelheim, P. (1991). Interleukin 4 promotes expressionof mast cell ICAM-1 antigen. Proc Natl Acad Sei U S A 55, 3339-3342.

Valentine, M. D. (1992). Anaphylaxis and stinging insect hypersensitivity. Jama 268, 2830-

2833.

van den Biggelaar, A. H., van Ree, R., Rodrigues, L. C, Lell, B., Deelder, A. M.,

Kremsner, P. G., and Yazdanbakhsh, M. (2000). Decreased atopy in children infected with

Schistosoma haematobium: a role for parasite-induced interleukin-10. Lancet 356, 1723-

1727.

Vercelli, D., Jabara, H. H., Arai, K., and Geha, R. S. (1989). Induction of human IgE

synthesis requires interleukin 4 and T/B cell interactions involving the T cell receptor/CD3

complex and MHC class II antigens. J Exp Med 169, 1295-1307.

Voehringer, D., Shinkai, K., and Locksley, R. M. (2004). Type 2 immunity reflects

orchestrated recruitment of cells committed to IL-4 production. Immunity 20, 267-277'.

von Bubnoff, D., Geiger, E., and Bieber, T. (2001). Antigen-presenting cells in allergy. J

Allergy Clin Immunol 705, 329-339.

von Mutius, E., Weiland, S. K., Fritzsch, C, Duhme, H., and Keil, U. (1998). Increasing

prevalence of hay fever and atopy among children in Leipzig, East Germany. Lancet 357,862-866.

Wan, T., Beavil, R. L., Fabiane, S. M., Beavil, A. J., Sohi, M. K., Keown, M., Young, R. J.,

Henry, A. J., Owens, R. J., Gould, H. J., and Sutton, B. J. (2002). The crystal structure of

IgE Fc reveals an asymmetrically bent conformation. Nat Immunol 3, 681-686.

Page 127: The Interplay of IgE and its High Affinity Receptor FceRI

124 REFERENCES

Wang, B., Rieger, A., Kilgus, O., Ochiai, K., Maurer, D., Fodinger, D., Kinet, J. P., and

Stingl, G. (1992). Epidermal Langerhans cells from normal human skin bind monomeric

IgE via Fc epsilon RI. J Exp Med 775, 1353-1365.

Wang, H. W., Tedla, N., Lloyd, A. R., Wakefield, D., and McNeil, P. H. (1998). Mast cell

activation and migration to lymph nodes during induction of an immune response in mice. J

Clin Invest 702, 1617-1626.

Wang, L. F., Lin, J. Y., Hsieh, K. H., and Lin, R. H. (1996). Epicutaneous exposure of

protein antigen induces a predominant Th2-like response with high IgE production in mice.

J Immunol 156, 4077-4082.

Warner, J. A., Jones, C. A., Jones, A. C, and Warner, J. O. (2000). Prenatal origins of

allergic disease. J Allergy Clin Immunol 705, S493-498.

Watanabe, N., Katakura, K., Kobayashi, A., Okumura, K., and Ovary, Z. (1988). Protective

immunity and eosinophilia in IgE-deficient SJA/9 mice infected with Nippostrongylusbrasiliensis and Trichinella spiralis. Proc Natl Acad Sei U S A 55, 4460-4462.

Wedemeyer, J., Tsai, M., and Galli, S. J. (2000). Roles of mast cells and basophils in innate

and acquired immunity. Curr Opin Immunol 72, 624-631.

Weisbroth, S. H., Friedman, S., and Scher, S. (1976). The parasitic ecology of the rodent

mite, Myobia musculi. III. Lesions in certain host strains. Lab Anim Sei 26, 725-735.

Wills-Karp, M., Santeliz, J., and Karp, C. L. (2001). The germless theory of allergicdisease: revisiting the hygiene hypothesis. Nat Rev Immunol 7, 69-75.

Wilson, D. R., Merrett, T. G., Varga, E. M., Smurthwaite, L., Gould, H. J., Kemp, M.,

Hooper, J., Till, S. J., and Durham, S. R. (2002). Increases in allergen-specific IgE in BAL

after segmental allergen challenge in atopic asthmatics. Am J Respir Crit Care Med 165,22-26.

Yamaguchi, M., Lantz, C. S., Oettgen, H. C, Katona, I. M., Fleming, T., Miyajima, I.,

Kinet, J. P., and Galli, S. J. (1997). IgE enhances mouse mast cell Fc(epsilon)RI expressionin vitro and in vivo: evidence for a novel amplification mechanism in IgE-dependentreactions. J Exp Med 755, 663-672.

Yanagihara, Y., Kajiwara, K., Basaki, Y., Ikizawa, K., Ebisawa, M., Ra, C, Tachimoto, H.,and Saito, H. (1998). Cultured basophils but not cultured mast cells induce human IgE

synthesis in B cells after immunologic stimulation. Clin Exp Immunol 777, 136-143.

Yazdanbakhsh, M., Kremsner, P. G., and van Ree, R. (2002). Allergy, parasites, and the

hygiene hypothesis. Science 296, 490-494.

Ying, S., Humbert, M., Barkans, J., Corrigan, C. J., Pfister, R., Menz, G., Lärche, M.,

Robinson, D. S., Durham, S. R., and Kay, A. B. (1997). Expression of IL-4 and IL-5

mRNA and protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in

bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics. J Immunol

755, 3539-3544.

Page 128: The Interplay of IgE and its High Affinity Receptor FceRI

REFERENCES 125

Yoshimoto, T., and Paul, W. E. (1994). CD4pos, NKl.lpos T cells promptly produceinterleukin 4 in response to in vivo challenge with anti-CD3. J Exp Med 779, 1285-1295.

Page 129: The Interplay of IgE and its High Affinity Receptor FceRI

126 REFERENCES

Page 130: The Interplay of IgE and its High Affinity Receptor FceRI

Acknowledgements 127

Acknowledgements

Als erstes möchte ich mich bei Hans Hengartner und Rolf Zinkernagel bedanken, die mich

in ihre Arbeitsgruppe aufgenommen haben und mir die Möglichkeit gaben, meine

Doktorarbeit in ihrem Institut durchzuführen. Ein besonderer Dank gilt meinem

Doktorvater, Hans Hengartner, der mir stets sein Vertrauen zu spüren gab.

Most of all I would like to thank my super-supervisor, Kathy McCoy, for her enormous

support, constant encouragements, and tireless smile during the entire time of my thesis. I

am extremely grateful for her invaluable help, be it in practical lab-work, project

discussions, or thesis proofreading. Kathy not only taught me a great deal about

immunology, but was also the most fun boss I could ever hope to work with!

I will never forget the glorious time of the Girls' Lab G43! I wish to thank the fabulous

girls Marianne Martinic, Kathy McCoy, Nicola Harris, and Nathalie Oetiker, who besides

being great lab-mates also became good friends. The coffee-breaks, dinners, parties, and

above all many, many laughs created a fantastic atmosphere, and I consider myself

extremely lucky for having worked in such a great environment.

Herzlichen Dank an meine PhD-mates Raphael Zellweger, Katja Fink und Philippe Krebs

für die gute Stimmung, die wir während der letzten vier Jahre untereinander hatten, sowie

für die genüsslichen Gourmet-PhD-Treffen. Un grand merci speziell an Rafi, der in der

strengen Abschlussphase dieser Doktorarbeit immer für gute Unterhaltung und Glacé-

Pausen zu haben war.

Ein grosser Dank gilt Sarah Hatak für die wertvolle Hilfe bei den unendlichen ELISAs

sowie gegen schlechte Haltung und verspannten Nacken.

Ich möchte mich herzlich bei Pascale Ohnsorg bedanken, weil sie mir erlaubte, meine

„Supervisor"-Fähigkeiten zu trainieren, und weil sie immer so wunderbar unkompliziert

war.

Page 131: The Interplay of IgE and its High Affinity Receptor FceRI

128 Acknowledgements

I wish to thank all the members of Expimm for keeping the institute together and making it

so diverse and special. I am grateful for the help and support from all of you, and for those

valuable interactions, which have made my time here an enriching experience. Many thanks

to the jogging-crew for such energy-reloading and entertaining breaks, and to the computer-

experts for countless IT-support.

Ganz besonders möchte ich mich bei meinen Eltern bedanken, für ihre grenzenlose

Unterstützung und enormes Vertrauen. Mamo i Tato dziekuje Warn bardzo!

Meinem Freund, Beat Sigrist, bin ich dafür, dass er mir stets Unterstützung, Kraft, Mut,

Freude und Liebe schenkt (und jeden Morgen das feine Müsli zubereitet!) zutiefst dankbar.

Page 132: The Interplay of IgE and its High Affinity Receptor FceRI

Curriculum Vitae

Curriculum Vitae

Personal

Name

Date of birth

Place of birth

Nationality

Marital status

Work address

Home address

Languages

Veronika Pochanke

07.09.1975

Bielsko-Biala, Poland

Swiss and Polish

single

Institute of Experimental Immunology

University Hospital Zürich

Schmelzbergstrasse 12

8091 Zürich, Switzerland

Phone:+41 (0)44 255 27 34

FAX:+41 (0)44 255 44 20

e-mail: [email protected]

Waffenplatzstrasse 51

8002 Zürich, Switzerland

German, English, French, Polish, Esperanto

Education

2001 to present

2000-2001

1996-2000

1998 - 1999

1991-1996

Doctoral thesis: "The Interplay of IgE and its High

Affinity Receptor FcsRI" at the Institute of

Experimental Immunology of the University Hospitaland the Swiss Federal Institute of Technology ETH,

Zürich, Switzerland, under the supervision of Prof. Dr.

Hans Hengartner and Dr. Kathy D. McCoy

Diploma thesis: "Analysis of Gene Silencing in StablyTransfected Cell Lines" at the Laboratory of Molecular

Biotechnology LBTM of the University of Lausanne,

Switzerland, under the supervision of Prof. Dr. Nicolas

Mermod - Faculty Award (Prix de Faculté)

Studies in Biology at the University of Lausanne,Switzerland

Exchange student in Biology at the University of South

Carolina, Columbia, USA - President's Honor List,Dean's Honor List

Gymnasium Typus C, Kantonsschule am Burggraben,St. Gallen, Switzerland