the role of complement in immunity to nippostrongylus

106
The Role of Complement in Immunity to Nippostrongylus brasiliensis A thesis submitted for the degree of DOCTOR OF PHILOSOPHY as a portfolio of publications by Paul Robert Giacomin Discipline of Microbiology and Immunology School of Molecular and Biomedical Science The University of Adelaide Australia September, 2007

Upload: others

Post on 22-Oct-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

The Role of Complement in Immunity to Nippostrongylus brasiliensis

A thesis submitted for the degree of

DOCTOR OF PHILOSOPHY

as a portfolio of publications

by

Paul Robert Giacomin

Discipline of Microbiology and Immunology School of Molecular and Biomedical Science

The University of Adelaide Australia

September, 2007

TABLE OF CONTENTS

ABSTRACT……………………………………………………………………………i

DECLARATION……………………………………………………………………...iii

ACKNOWLEDGEMENT OF ANY HELP………………………………………….. iv

STATEMENT OF AUTHORSHIP-CHAPTER 2……………………………………..v

STATEMENT OF AUTHORSHIP-CHAPTER 3…………………………………....vii

STATEMENT OF AUTHORSHIP-CHAPTER 4……………………………………..x

ACKNOWLEDGEMENTS………………………………………………………….xvi

PUBLICATIONS…………………………………………………………………..xviii

COMMONLY-USED ABBREVIATIONS………………………………………….xix

CHAPTER 1: INTRODUCTION AND REVIEW OF THE LITERATURE

1.1 HEALTH AND ECONOMIC CONSEQUENCES OF PARASITIC HELMINTH INFECTIONS ..........................................................................................1 1.2 IMMUNE RESPONSES TO HELMINTH INFECTION .......................................2

1.2.1 Immune recognition and antigen processing ...................................................3 1.2.2 Cytokine responses during helminth infection .................................................4 1.2.3 Immunological basis of gastrointestinal helminth expulsion ...........................6

1.3 LEUKOCYTE-MEDIATED KILLING OF HELMINTHS....................................9 1.3.1 Neutrophils........................................................................................................9 1.3.2 Macrophages...................................................................................................10 1.3.3 Eosinophils......................................................................................................10

1.3.3.1 IL-5 and eosinophils ................................................................................11 1.3.3.2 Eosinophil recruitment.............................................................................12 1.3.3.3 Eosinophil activation, secretion and degranulation .................................13 1.3.3.4 Role of eosinophils in disease..................................................................14 1.3.3.5 Role of eosinophils is killing helminths ..................................................15 1.3.3.6 Other roles for eosinophils during helminth infections ...........................17

1.4 THE COMPLEMENT SYSTEM ..........................................................................19 1.4.1 Function of the complement system ...............................................................19 1.4.2 Pathways to complement activation ...............................................................20 1.4.3 Complement-dependent immunity to pathogens ............................................21

1.5 ROLE OF COMPLEMENT IN IMMUNITY TO HELMINTHS.........................23 1.5.1 Complement activation by helminths .............................................................25 1.5.2 Complement-dependent leukocyte-mediated killing of helminths.................26

1.5.2.1 Recruitment of effector leukocytes..........................................................26 1.5.2.2 Adherence and activation of effector leukocytes.....................................27

1.5.3 Evasion of complement activation and leukocyte adherence .........................27 1.6 NIPPOSTRONGYLUS BRASILIENSIS AS A MODEL FOR STUDYING IMMUNITY TO HELMINTHS.............................................................29

1.6.1 Parasite life cycle ............................................................................................29 1.6.2 Immune responses to N. brasiliensis ..............................................................30

1.6.2.1 Cellular inflammatory responses .............................................................30 1.6.2.2 Cytokine responses ..................................................................................32

1.6.2.3 Role for eosinophils .................................................................................32

1.7 INTRODUCTION TO THIS STUDY...................................................................34 CHAPTER TWO: Quantitation of complement and leukocyte binding to a parasitic helminth species....…………………………………………… 36 LINKAGE TO CHAPTER TWO AND ARTICLE……………………………… 37 CHAPTER THREE: Loss of complement activation and leukocyte adherence as Nippostrongylus brasiliensis develops within the murine host ………. 39 LINKAGE TO CHAPTER THREE AND ARTICLE............................................40 CHAPTER FOUR: The role of complement in innate, adaptive and eosinophil-dependent immunity to the nematode Nippostrongylus brasiliensis.…….43 LINKAGE TO CHAPTER FOUR AND ARTICLE…………………………….. 44 CHAPTER FIVE: DISCUSSION AND CONCLUSION 5.1 GENERAL DISCUSSION ....................................................................................46

5.1.1 Summary of main findings .............................................................................46 5.1.2 Complement and eosinophil-dependent immunity to helminths ....................47

5.1.2.1 Eosinophil-dependent resistance to helminths.........................................47 5.1.2.2 Role of complement in vivo and in vitro..................................................49 5.1.2.3 Complement-dependent eosinophil recruitment to parasite-infected skin .......................................................................................51 5.1.2.4 Complement-independent eosinophil recruitment...................................51 5.1.2.5 Eosinophil versus neutrophil recruitment ................................................53 5.1.2.6 Complement-independent leukocyte adherence to helminths .................54 5.1.2.7 Eosinophil degranulation .........................................................................55 5.1.2.8 Larval aggregation ...................................................................................57

5.1.3 Evasion of complement activation by helminths............................................58 5.1.4 Pulmonary cellular responses following helminth infection ..........................59

5.1.4.1 Restricted early cellular inflammation in the lungs .................................59 5.1.4.2 Delayed cellular inflammation in the lungs.............................................60

5.1.5 Secondary immune response to helminth infection........................................61 5.1.6 Future directions for studies using complement-deficient/IL-5 Tg mice .......62 5.1.7 Issues for design of anthelmintic vaccines .....................................................62

5.2 CONCLUSION......................................................................................................63 REFERENCES…………………………………………………………………….....65

iABSTRACT

Approximately two billion people are infected with helminths worldwide. In order to develop

a vaccine against these pathogens, more needs to be known about the immune response to

helminths. Eosinophils are important for resistance to some helminth species and their

recruitment to infected tissues, attachment to parasites and degranulation may all be critical

processes for immunity. Complement may contribute to these processes via generation of

chemotactic factors (C3a and C5a) or opsonisation of the parasite with C3b/iC3b. The

importance of complement during helminth infection is unclear, though complement does

promote leukocyte-mediated killing of several helminth species in vitro. The aim of the

present study was to investigate the role of complement in immunity of mice to

Nippostrongylus brasiliensis, with a focus on whether complement facilitates eosinophil-

dependent resistance to this parasite. A new fluorescence-based method for quantifying in

vitro complement deposition and leukocyte adherence on N. brasiliensis was developed. C3

from human serum was deposited on infective-stage L3 via the classical or lectin complement

pathways. In contrast, the alternative complement pathway mediated binding of mouse C3

and eosinophil-rich mouse peritoneal leukocytes to L3. Interestingly, the ability of

complement and leukocytes to bind to the parasite changed as it matured. Larvae recovered

from the skin 30 min post-injection (p.i.) were coated with C3, however those harvested 150

min p.i. exhibited reduced C3 binding capacity. Binding of C3 and eosinophils to larvae

recovered from the lungs 24-48 h p.i. (L4) was also diminished compared to that seen on L3.

Adult intestinal worms bound C3 and leukocytes only when treated ex vivo with serum and

cells. Mice lacking in classical (C1q-deficient), alternative (factor B-deficient) or all

complement pathways (C3-deficient) were then employed to determine if complement was

important for resistance of mice to N. brasiliensis. IL-5 Tg mice deficient in individual

complement genes were generated to assess whether complement contributed to eosinophil-

dependent resistance to the parasite. Factor B-deficient mice exhibited impaired C3

deposition on larvae, eosinophil recruitment, eosinophil degranulation and larval aggregation

iiin the skin 30 min p.i. Eosinophil recruitment was similarly abolished by treatment of mice

with the C5aR inhibitor PMX53. However at 150 min p.i., larval aggregation, eosinophil and

neutrophil recruitment, leukocyte adherence and eosinophil degranulation were largely

complement-independent. Ablation of factor B or C3 caused minor but significant increases

in lung-larval burden during primary, but not in secondary, infections. Critically, a lack of C3

or factor B in IL-5 Tg mice failed to greatly impair the strong innate anti-parasite resistance

typical of these animals, suggesting that eosinophils can provide immunity to N. brasiliensis

infection in the absence of complement. This was unexpected, given the evidence from this

and previous studies which suggested that in vitro, complement is important for promoting

eosinophil-dependent killing of N. brasiliensis and other helminth species. The mechanism(s)

by which eosinophils kill N. brasiliensis remain unknown, but may involve the coordination

of the complement system with complement-independent factors that act in the early stages of

infection. Critically, the influence of complement is limited, because soon after entry into the

host, the parasite develops the ability to resist complement activation.

ivACKNOWLEDGEMENT OF ANY HELP

I acknowledge the help of:

All co-authors named on each of the published journal articles comprised in this thesis, for

evaluating manuscript drafts and suggesting changes during the revision process. In

particular, Dr. Lindsay Dent, who acted as co-author for all manuscripts, co-wrote and revised

drafts with myself before submission and critically read sections of my thesis.

Dr. Hui Wang, who contributed to the early development of techniques for measuring C3

deposition on helminths.

Ms. Michelle Knott, for technical assistance with large-scale animal experiments.

NOTE: Statements of authorship appear in the print copy of the thesis held in

the University of Adelaide Library.

xvi ACKNOWLEDGEMENTS

Firstly, I would like to thank my principal supervisor Dr. Lindsay Dent for his dedicated

support, guidance and encouragement throughout my Ph.D. studies. Your mentorship has

made my experience as a Ph.D. student very enjoyable and rewarding. To my co-supervisor,

Professor David Gordon, I also extend a thankyou for sharing your ideas and supporting my

goals.

I acknowledge the help and friendship of all Honours and Ph.D. students who have been part

of the Dent laboratory throughout my time here. In particular, I thank Michelle Knott and

Damon Tumes who have provided constant support and made the lab an enjoyable place to be

around. Also, I thank Hui Wang for her help with my project and technical assistance when I

first joined the lab.

I thank the student and staff members of the Discipline of Microbiology and Immunology and

the School of Molecular and Biomedical Science for making it such a good place to work. In

particular, I thank Nick Eyre, Francesca Bell, Wendy Parker, Georget Reaiche, The Friday

beer crew and the Wednesday soccer crew for making my time at Uni entertaining and

memorable.

To our national and international collaborators Marina Botto, Alex Loukas, Steve Taylor and

Mohamed Daha, I thank you for your assistance with my project.

Whilst conducting these studies I was supported by a University of Adelaide scholarship. I

also thank the School of Molecular and Biomedical Science for their support financially and

for other resources relating to my project.

xviiI thank all of my non-Uni friends for many years of great friendship and support.

I wholeheartedly thank my parents, for whom I am eternally grateful for supporting me with

whatever decisions I have made over the years, allowing me to achieve my goals. I also thank

the rest of my family (and extended family), especially my sister Amanda and all of my

grandparents.

Lastly, I thank my wife Michelle, who has been a loving and inspiring partner since I began

my Ph.D. studies and who I look forward to a spending long life with.

xviiiPUBLICATIONS

Within thesis:

1. Giacomin PR, Wang H, Gordon DL and Dent LA (2004). Quantitation of

complement and leukocyte binding to a parasitic helminth species. Journal of Immunological Methods 289 (1-2): 201-210

2. Giacomin PR, Wang H, Gordon DL, Botto M and Dent LA (2005). Loss of

complement activation and leukocyte adherence as Nippostrongylus brasiliensis develops within the murine host. Infection and Immunity 73 (11): 7442-7449

3. Giacomin PR, Gordon DL, Botto M, Daha MR, Sanderson SD, Taylor SM and

Dent LA (2007). Molecular Immunology 45 (2): 446-455

Other publication arising from Ph.D. studies:

1. Knott ML, Matthaei KI, Giacomin PR, Wang H, Foster PS, Dent LA (2007). Impaired resistance in early secondary Nippostrongylus brasiliensis infections in mice with defective eosinophilopoeisis. International Journal for Parasitology 37 (12): 1367-1378

Previous publications

1. Keating DJ, Rychkov GY, Giacomin P, Roberts ML (2005). Oxygen-sensing pathway for SK channels in the ovine adrenal medulla. Clinical and Experimental Pharmacology and Physiology, 32 (10): 882-887

2. McKay D, Brooker R, Giacomin P, Ridding M, Miles T (2002). Time course of

induction of increased human motor cortex excitability by nerve stimulation. Neuroreport, 13 (10): 1271-1273

Manuscripts in preparation

1. Giacomin PR, Gauld AD, Cava M, Iddewalla D, Gordon DL and Dent LA. Excretory/secretory proteins from Toxocara canis infective larvae reduce eosinophil-dependent innate resistance to Nippostrongylus brasiliensis infection

xixCOMMONLY-USED ABBREVIATIONS

Abbreviation full definition

AAM alternatively-activated macrophage

ADCC antibody-dependent cellular cytotoxicity

AMCase acidic mammalian chitinase

BAL bronchoalveolar lavage

CCR3 chemokine receptor 3

CR complement receptor

CVF cobra venom factor

DAF decay accelerating factor

ES excretory/secretory

EPO eosinophil peroxidase

Ig immunoglobulin

IL interleukin

i.p. intra-peritoneal

L3 third-stage larvae

L4 fourth-stage larvae

MAC membrane attack complex

MASP MBL-associated serine protease

MBL mannan binding lectin

MBP major basic protein

MPO myeloperoxidase

NK natural-killer

p.i. post-infection

PRR pattern recognition receptor

s.c sub-cutaneous

STAT6 signal transducer and activator of transcription 6

Th T-helper

Tg transgenic

VLA very-late antigen

WT wildtype

CHAPTER ONE

CHAPTER ONE: INTRODUCTION AND REVIEW OF THE

LITERATURE

CHAPTER ONE 1

1.1 HEALTH AND ECONOMIC CONSEQUENCES OF PARASITIC

HELMINTH INFECTIONS

Helminth infections cause enormous global health and economic problems. Recent estimates

indicate that approximately 2 billion people are infected worldwide, along with countless

agricultural and domesticated animals. Helminths are a diverse family of parasitic worms,

comprised of two main classes that are only distantly related, nematodes (roundworms) and

platyhelminths (flatworms). Within these two main groups there are many species of

helminths that exhibit complex life cycles, most commonly beginning with either ingestion of

parasite eggs or infective larva that enter the host. The parasite may undergo extensive

growth and maturation while migrating through the host, or in some cases through several

host species, to ultimately accomplish sexual reproduction. To achieve this, helminths must

acquire all nutrients from the host and this can cause significant host morbidity. This is

especially a problem for young host animals, since helminth infections may result in impaired

growth, development, cognitive function and pre-disposition to infection with other pathogens

(Finkelman et al. 1997). Some species of helminth migrate through multiple tissues as part of

the life cycle, causing mechanical damage to delicate tissues such as lung alveoli, the liver or

the eye. In addition to mechanical damage there is significant immunopathology associated

with inappropriate, excessive or chronic immune responses directed against the parasite

(Meeusen 1999). As the survival of the parasite is dependent on the survival of the host,

helminth infection does not typically result in host mortality. Nevertheless, life-threatening

illness and over 150,000 deaths do occur each year in developing countries (Crompton 1999).

Poor levels of nutrition and sanitation, tropical climate and a lack of availability of

anthelmintic drugs increases both the frequency of exposure to parasitic helminths and the

detrimental effects of infection. In developed countries, helminths are a major problem for

agricultural industries, where large losses in product yield and hence profit are seen due to

helminth infection of livestock.

CHAPTER ONE 2

Currently, anthelmintic drugs are the most effective treatment for helminth infections.

However, these drugs are expensive, do not protect against re-infection and there is evidence

of the emergence of drug-resistance in parasites of livestock (Besier and Love 2003; von

Samson-Himmelstjerna and Blackhall 2005). Hence, more effective preventative treatments

are needed to control helminth infections globally. Effective vaccines against human

helminth infections have not been developed. Recently some efficacious vaccines against

tapeworm species have been developed for use in livestock (Lightowlers et al. 2003), leading

to the possibility that a vaccine for preventing infection of humans with some parasite species

is close to fruition. To achieve this, more needs to be known about the nature of the host

immune response to helminth infection and how some helminths have evolved immune

evasion strategies.

1.2 IMMUNE RESPONSES TO HELMINTH INFECTION

Despite the diverse nature of the species of organisms that we classify as parasitic helminths,

in general, the immune responses directed toward elimination of these organisms are

surprisingly similar. The nature of these inflammatory responses are often similar to those

elicited by allergen challenge, resulting in the release of arrays of particular cytokines

(Finkelman et al. 1997), induction of polyclonal immunoglobulin E (IgE) and activation of

leukocytes such as eosinophils, neutrophils, basophils and mast cells (Kay et al. 1985).

Immune responses directed against helminths are complex by necessity, as diverse responses

must be elicited to parasites that have tissue-invasive and/or gastrointestinal lumen-dwelling

life cycles. Hence, the nature and magnitude of the immune response may change in different

infected tissues. Furthermore, most helminths possess the ability to actively modulate

immune responses to prevent immune-mediated destruction, allowing residence within a

single host for months, or even years as seen for some cestode and trematode species

CHAPTER ONE 3(Sandground 1936).

1.2.1 Immune recognition and antigen processing

In order for a host to elicit an effective innate or adaptive immune response against a specific

parasite, the parasite must first be recognised as non-self. This is made more complicated by

the fact that during the course of an infection with a helminth, the nature of the antigens

expressed by the parasite may change as it matures (Preston et al. 1986; Lightowlers and

Rickard 1988; Tkalcevic et al. 1996). Innate mechanisms of parasite recognition may aid in

elimination of the helminth upon initial exposure and also trigger adaptive immune responses

to protect against re-infection (Abraham et al. 2004; Knight et al. 2004; Pemberton et al.

2004; de Veer et al. 2007). Many species of helminth have been shown to activate the innate

immune system shortly after infection, but the mechanisms involved are not fully understood.

Expression of pattern-recognition receptors (PRRs) enable leukocytes to recognise surface

molecules that are highly conserved amongst pathogens of a given class (Applequist et al.

2002). Such molecules are termed pathogen-associated molecular patterns (PAMPs) and can

include carbohydrates, proteins, lipids and nucleic acids. While much research has

investigated the role of the Toll-like receptor (TLR) family in immunity to bacterial or fungal

pathogens (Netea et al. 2004), limited research has investigated the role of TLRs in eliciting

immune responses to helminths. There is some evidence that antigens produced by

schistosomes can activate TLR signalling (van der Kleij et al. 2002), though bacteria that

reside on or within some helminths may also be responsible for this activation (Brattig et al.

2004). Binding of host C-type lectins (CTLs) to the carbohydrate-rich parasite surface may

also be a mechanism of initiation of immune responses (McGuinness et al. 2003). Mannan-

binding lectin (MBL) is a collectin that has been shown to bind to helminths Schistosoma

mansoni (Klabunde et al. 2000) and Trichinella spiralis (Gruden-Movsesijan et al. 2003).

MBL binding can initiate complement activation on the parasite via the lectin pathway, which

may help leukocytes kill helminths (see section 1.5.2). Since complement can be rapidly and

CHAPTER ONE 4non-specifically activated by the presence of PAMPs, it may be a very important factor in the

recognition of helminths in the very early stages of infection. Many different host cell types

express receptors for complement factors (Gasque 2004), which can recognise pathogens

opsonised with complement molecules such as C3b, or alternatively act directly as PRRs for

lipopolysaccharide (LPS) and β-glycan on microbial surfaces (Ehlers 2000). A more detailed

description of the role of complement in immunity to parasitic helminths is provided in

section 1.5.

Dendritic cells are important for the development of adaptive immune responses to pathogens,

by processing and presenting foreign antigens to T-lymphocytes (Reis e Sousa 2001). The

ways in which helminth antigens are processed and presented are not completely understood.

As most helminths are too large to be internalised, the antigens presented by dendritic cells

may initially consist mostly of those factors excreted or secreted by the helminths

(Lightowlers and Rickard 1988; Balic et al. 2004). Presentation of parasite antigens to naïve

lymphocytes can result in the induction of a cytokine bias (see section 1.2.2) and can result in

the generation of parasite-specific and non-specific antibodies (Whelan et al. 2000).

Antibody that arises after previous exposure to a helminth can allow swift and specific

recognition of the parasite, enabling the rapid instigation of protective adaptive immune

responses.

1.2.2 Cytokine responses during helminth infection

Upon contact with a potentially harmful foreign body, the immune system must be able to

induce a specific set of effector mechanisms to eliminate the pathogen. Some of this

specificity is achieved by inducing the secretion of specific arrays of cytokines. CD4+ T-

lymphocytes are a major cytokine-producing and regulatory cell of the immune system,

though other leukocytes such as macrophages, natural killer (NK)-cells, basophils, mast cells,

eosinophils and CD8+ T cells also readily secrete cytokines upon stimulation. Mice that have

CHAPTER ONE 5impaired CD4-dependent cell function, after treatment with anti-CD4 antibody are less

resistant to infection with Heligomosomoides polygyrus (Urban et al. 1991) and Trichuris

muris (Koyama et al. 1995), with increased worm fecundity and impaired worm expulsion,

respectively. CD4+ T-cells differ in the types of cytokines that are expressed and at least 4

dominant cytokine patterns are commonly described. T-helper 1 (Th1) cells secrete

interleukin (IL)-2, interferon (IFN)-γ and tumour necrosis factor (TNF)-β (commonly termed

as Type-1 cytokines) (Mosmann and Coffman 1989). Th2 cells secrete IL-4, IL-5, IL-6, IL-9,

IL-10 and IL-13 (Type-2 cytokines) (Romagnani 2000). Both Th1 and Th2 cells may secrete

IL-3 and granulocyte-macrophage colony stimulating factor (GM-CSF). A third subset of

cytokine-producing CD4+ T-cells, the Th3 or Type 3 class, produce a more heterogeneous

cytokine profile, most notably transforming growth factor β (TGFβ) that functions mainly to

inhibit the actions of T-cells, macrophages and counteract the actions of other pro-

inflammatory cytokines. Cells within the Type 3 group are commonly known as regulatory

T-cells (T-regs) (Belkaid and Rouse 2005). Lastly, the recently identified Th17 cell lineage

represents a distinct group of CD4+ T-cells that produce, among other factors, the cytokine

IL-17 (Aggarwal et al. 2003; Harrington et al. 2006). Th17 cells are believed to be

particularly important for protection against extracellular bacteria through their ability to

stimulate inflammatory cell recruitment (Happel et al. 2005), but they have also been

implicated in a number of autoimmune diseases (Chen et al. 2006; Komiyama et al. 2006).

Some cytokines possess the ability to enhance production of other cytokines of the same

category, creating a positive feedback loop. This, along with the existence of cross

regulation, i.e. Type 1 cytokines blocking activity or production of Type 2 cytokines and vice

versa, leads to a pronounced cytokine bias in some disease states (Mosmann and Coffman

1989). Infection with several parasitic helminth species has been shown to be a potent

inducer of Type 2-cytokine biased immune responses (Grencis 1997). Exposing mice to

either proteins that are excreted or secreted (ES proteins) by the nematode Nippostrongylus

CHAPTER ONE 6brasiliensis (Holland et al. 2000) or the whole parasite (McKenzie et al. 1998; Min et al.

2004) is sufficient to cause a marked Type 2 cytokine response in mice. A Type 2-cytokine

bias has various effects on the immune status of the host, including IL-5-dependent

eosinophilia (Dent et al. 1990; Urban et al. 1992), IL-4-dependent class switching to promote

IgE production (Finkelman et al. 1988) and mastocytosis (Madden et al. 1991). In contrast,

induction of a Type 1 cytokine response in mice by administration of IL-12 has been shown

to delay clearance of N. brasiliensis and T. muris (Finkelman et al. 1994; Bancroft et al.

1997). The exact mechanism by which a Type 2 cytokine response is triggered during a

parasitic helminth infection is not completely understood. Early production of IL-4 is an

important step in the establishment and amplification of Type 2 immune responses, but how

cells are activated to secrete IL-4 is unclear. Recently, an IL-25-dependent non-B non T-cell

has been shown to be a major mediator of rapid IL-4, IL-5 and IL-13 production during N.

brasiliensis infection (Fallon et al. 2006). Additionally, basophils are an important early

source of IL-4 or IL-13 (Falcone et al. 2001; Min et al. 2004; Voehringer et al. 2004) and

these cells rapidly increase in number in the peripheral blood and infected tissues following

exposure to some helminths (Falcone et al. 2001). In this respect, basophils may be critical

for skewing the immune response in the early stages of helminth infection, in cooperation

with other cytokine-producing leukocytes such as eosinophils and the recently identified IL-

25-dependent non-B non T-cell population.

1.2.3 Immunological basis of gastrointestinal helminth expulsion

Efficient expulsion of intestine-dwelling helminths requires the instigation of physiological

changes to the gastrointestinal environment that act to “force” the parasite out of the intestine.

In particular, helminth infection is associated with increased intestinal smooth muscle cell

contractility. Mouse strains that produce more pronounced smooth muscle contractility (e.g.

NIH Swiss mice) expel T. muris more efficiently than other strains that exhibit lesser changes

in smooth muscle contractility (e.g. B10.BR mice) (Vallance et al. 1997). Helminth

CHAPTER ONE 7infections also result in increases in ion and water secretion, which act to “flush” the parasites

from the intestine (Mettrick et al. 1979). Goblet cells within the intestinal epithelium undergo

rapid proliferation (hyperplasia) at the time of expulsion of several species of helminth (Nawa

et al. 1994; Onah and Nawa 2000). The secretion of mucus by these cells may exclude and

trap worms in the gastrointestinal mucosa and promote expulsion by preventing their

attachment and feeding. Epithelial cells within the intestinal microenvironment have been

shown to produce an array of proteins that may be potent effector molecules against

helminths, such as intelectins and resistin-like molecules (RELMs) (Datta et al. 2005; Nair et

al. 2005), however the functions of these molecules during helminth infection are not fully

understood. An increase in the rate of intestinal epithelial turnover (Symons 1978) and rate of

migration from the base of the crypts to the extrusion zone have been associated with

elimination of T. muris (Cliffe et al. 2005). This may restrict the ability of helminths to attach

to the gut wall and hence aid in worm expulsion.

Mast cells have long been implicated as important for immunity to helminths. Their primary

role may be to promote the expulsion of gastrointestinal parasites, however they are also

prominent in the skin and lungs during infections with some helminth species (Bentley et al.

1981; Matsuda et al. 2001). Intestinal mastocytosis and an increase in tissue mast cell

protease levels coincide with expulsion of N. brasiliensis and T. spiralis (Miller and Jarrett

1971; Woodbury et al. 1984). Mice deficient in mast cells (W/Wv mice) or mice treated with

anti-c-kit antibody are slow to expel T. spiralis and Strongyloides ratti (Abe and Nawa 1987;

Donaldson et al. 1996). However, there is evidence that expulsion of some helminths, such as

N. brasiliensis, proceeds normally in the absence of mast cells (Uber et al. 1980). Mast cells

release an as yet unidentified factor by degranulation that may stimulate parasite expulsion

(Onah and Nawa 2000). In summary, the process of expulsion of gastrointestinal helminths is

complex and involves elements of both the innate and adaptive immune system, co-ordinated

with physiological changes to intestinal tissues and secretions.

CHAPTER ONE 8

It is now commonly believed that changes in intestinal physiology after helminth infection are

a direct result of immune activation. Several Type-2 cytokines have been implicated in the

instigation of mechanisms that promote expulsion of gastrointestinal helminths. In particular,

activation of signal transducer and activation of transcription-6 (STAT6), a transcription

factor activated after ligation of the IL-4R (Takeda et al. 1996), is important. Worm

expulsion is delayed in STAT6-deficient mice infected with N. brasiliensis or Strongyloides

venezuelensis (Urban et al. 1998; Negrao-Correa et al. 2006). Blockade of the IL-4R using

an anti-IL-4R antibody, which inhibits both IL-4 and IL-13 signalling, leads to chronic T.

muris infections in normally resistant mice (Else et al. 1994). Subsequent experiments with

IL-4 or IL-13-deficient mice supported this observation and suggest that both cytokines play a

role in expulsion of this parasite (Bancroft et al. 1997). The type-2 cytokine IL-9 is also

involved in the process of T. muris expulsion, since neutralisation of IL-9 in mice infected

with this parasite delays worm expulsion (Khan et al. 2003). Intestinal smooth muscle

contractility ex vivo is inhibited by anti-IL-9 antibody treatment, suggesting IL-9 may act to

stimulate physiological changes in the small intestine that are necessary for expulsion of

parasites. Elimination of N. brasiliensis is also dependent on IL-4R and STAT6 expression,

however IL-13 may be more important than IL-4 for expulsion this parasite (Urban et al.

1998). The IL-4R must be expressed by non bone marrow-derived cells (i.e. non immune

cells) for timely expulsion of N. brasiliensis (Urban et al. 2001). On the other hand, whilst T.

spiralis expulsion still requires IL-4R signalling, in this model it is essential that the receptor

is expressed by bone marrow-derived immune cells and that mast cells are present in order for

the parasite to be expelled from the host (Urban et al. 2001). In summary, Type 2 cytokines

have been shown to be important for promoting expulsion of worms from the gut. The exact

mechanism of expulsion varies depending on which parasite is present, however activation of

the IL-4/IL-13/STAT6 system appears to be a common element required for expulsion of

gastrointestinal worms.

CHAPTER ONE 9 1.3 LEUKOCYTE-MEDIATED KILLING OF HELMINTHS

While PRRs, complement and antibody may be important for recognition of tissue-invasive

helminths and cytokines are effective at initiating protective immune responses and causing

parasite expulsion, active killing of helminths is typically dependent on cell-mediated

immunity. Macrophages and neutrophils have the capacity to directly inflict damage on

helminths. However it is eosinophils, which are often generated in large numbers during

helminth infections, which may have evolved as key effectors of protective immunity against

some parasite species.

1.3.1 Neutrophils

Neutrophils are effective at killing bacterial or unicellular organisms but there is some

evidence that they can also be cytotoxic for larger helminthic parasites (Incani and McLaren

1981; Shaio et al. 1990). Recruitment of these cells is rapid and is stimulated by many factors

including IL-8, prostaglandins, platelet-activating factor (PAF), leukotriene (LT)B4, N-

formyl methyonyl-leucyl-phenylalanine (fMLP) and complement factors such as C3a and C5a

(Jagels and Hugli 1992). Neutrophils are often observed to be amongst the first leukocytes to

arrive in an infected tissue. Neutrophils express Fc receptors that allow specific recognition

of antibody bound to the parasite surface, which promotes antibody-dependent cell-mediated

cytotoxicity (ADCC). Neutrophils express complement receptors CR1, CR3 and CR4 that

also facilitate their attachment to the surface of a pathogen. Complement-mediated ADCC

has been shown to be effective in the killing of S. mansoni schistosomula (Incani and

McLaren 1981) and Angiostrongylus cantonensis (Shaio et al. 1990) in vitro. Neutrophils can

also kill Strongyloides stercoralis larvae in vivo (Ligas et al. 2003; Galioto et al. 2006). The

mechanism by which neutrophils kill helminths is not clear, but is likely to include the actions

of the enzyme myeloperoxidase (MPO) that generate reactive oxygen intermediates and

CHAPTER ONE 10hypochlorite ions that could damage parasites. Purified MPO can efficiently kill newborn T.

spiralis larvae in vitro (Buys et al. 1984) and co-culture of neutrophils with T. spiralis or N.

brasiliensis results in deposition of superoxide on parasites (MacKenzie et al. 1981). Despite

this, neutrophils are not effective at killing other helminth species such as Toxocara canis

(Huwer et al. 1989). Furthermore, neutrophil responses are typically transient and with time

these cells are often replaced by eosinophils or macrophages in parasite-infected tissues.

1.3.2 Macrophages

As with neutrophils, macrophages are also effective at phagocytosing small microorganisms

and have been reported to be able to kill several helminth species (Sher et al. 1982; Egwang

et al. 1984). Macrophages express Fc receptors that can facilitate ADCC, and CR1, CR3 and

CR4 that allow recognition of complement factors bound to helminths. Macrophages adhere

to and degranulate on N. brasiliensis larvae in the presence of complement or antibody

(MacKenzie et al. 1981) and can kill N. brasiliensis (Egwang et al. 1984) and S. mansoni

(Sher et al. 1982) larvae in vitro. Macrophages also produce MPO and express the enzyme

nitric oxide synthase (NOS), which can generate toxic nitric oxide or peroxynitrite, both of

which may damage helminths (Buys et al. 1984; James et al. 1998; Gupta et al. 2004).

1.3.3 Eosinophils

There is strong evidence to support a role for eosinophils in protection against some helminth

species. Eosinophils are terminally differentiated granulocytic leukocytes produced from

myeloid CD34+ precursors in the bone marrow (Warren and Moore 1988). The early stage of

their differentiation from these precursors is controlled primarily by the cytokines GM-CSF

and IL-3 (Warren and Moore 1988). In the later stage, IL-5 is the critical cytokine involved

in the terminal differentiation and maturation of eosinophils (Lopez et al. 1986; Dent et al.

1990).

CHAPTER ONE 11Eosinophil granules store proteins that are both toxic to invading pathogens but also

potentially damaging to host tissues in some disease states. In humans there are four main

eosinophil granule proteins; major basic protein (MBP), eosinophil peroxidase (EPO),

eosinophil-derived neurotoxin (EDN) and eosinophil cationic protein (ECP) (Hamann et al.

1991). MBP and EPO are also expressed by mouse eosinophils; however the ribonucleases

EDN and ECP are not. Instead, mouse eosinophils express at least 11 genes that encode

proteins with ribonuclease activity, termed eosinophil-associated ribonucleases (EARs)

(Cormier et al. 2001). MBP is located within the electron-dense crystalline core of eosinophil

granules, while the other proteins are located within the surrounding matrix (Gleich et al.

1976; Dvorak et al. 1994; Egesten et al. 1997). In addition to these granule proteins,

eosinophils also express an array of other proteins, including cytokines, chemokines, growth

factors and lipid mediators (Rothenberg and Hogan 2006).

1.3.3.1 IL-5 and eosinophils

IL-5 controls the differentiation, maturation and survival of eosinophils in many species

including humans (Warren and Sanderson 1985; Lopez et al. 1986) and the maturation of B-1

lymphocytes in mice (Kinashi et al. 1986). Raised levels of IL-5 are commonly seen in

animals infected with parasitic helminths (Coffman et al. 1989), under allergic and asthmatic

conditions (Sur et al. 1995; Nagai et al. 1996) and in some autoimmune diseases (Hellmich et

al. 2005). Basal eosinophil numbers in the peripheral blood are typically low in naïve

animals (<5% of total leukocytes) with most residing within the gastrointestinal tract and

more variably in the female reproductive tract (Weller 1991; Mishra et al. 1999). However,

raised levels of IL-5 can stimulate eosinophilopoiesis in the bone marrow and vast increases

in blood and tissue eosinophil numbers (Dent et al. 1990). Treatment of humans or animals

with anti-IL-5 antibody reduces peripheral blood eosinophilia during experimental helminth

infections (Coffman et al. 1989; Sher et al. 1990) and in asthmatic and hyper-eosinophilia

syndrome (HES) patients (Leckie et al. 2000; Garrett et al. 2004; Simon et al. 2005).

CHAPTER ONE 12

The relationship between IL-5 and eosinophils in vivo has been explored using mice over-

expressing IL-5 transgenes (IL-5 Tg mice) and mice genetically deficient in IL-5 (IL-5-/-). IL-

5 Tg mice have been developed by several strategies and in each case the mice express

constitutive eosinophilia (Dent et al. 1990; Vaux et al. 1990; Tominaga et al. 1991), however

the distribution of eosinophils in various tissues differs depending on the nature of the

transgene used. IL-5 Tg lines developed by ligation of the dominant controlling region of the

human CD2 gene to a murine genomic DNA IL-5 gene and flanking regions yields IL-5 Tg

expression that is largely T-cell specific (Dent et al. 1990). Eosinophilia in these mice is

most evident in the peripheral blood, bone marrow, spleen, intestinal wall and lungs. The

degree of eosinophilia is dependent on transgene copy number, such that the Tg(0IL5)C2 line

(abbreviated Tg5C2) with approximately 49 copies of the IL-5 transgene express more

pronounced eosinophilia that the Tg(0IL5)C1 line (abbreviated Tg5C1), which has

approximately 8 copies of the IL-5 transgene. Conversely, IL-5-/- mice fail to develop

eosinophilia when challenged with aeroallergens in a mouse model of asthma or when

infected with helminths (Foster et al. 1996; Kopf et al. 1996; Matthaei et al. 1997; Ovington

et al. 1998). However these mice do exhibit very low numbers of eosinophils that are

functionally and morphologically similar to those in wild type (WT) mice. Although

eosinophils can be generated in small numbers in the absence of IL-5, presumably due to the

actions of GM-CSF and IL-3 in early differentiation (Warren and Moore 1988), eosinophilia

is dependent on IL-5 (Dent et al. 1990; Foster et al. 1996; Yu et al. 2002).

1.3.3.2 Eosinophil recruitment

Rapid and specific recruitment of eosinophils to sites of infection may be critical for

immunity to pathogens and can be mediated by many different factors (Resnick and Weller

1993). After maturation in the bone marrow, eosinophils circulate in the blood and traffic to

the gastrointestinal tract, thymus, mammary glands or uterus under normal, uninfected

CHAPTER ONE 13conditions. This trafficking is dependent on the chemokine eotaxin-1 (Gouon-Evans et al.

2000; Rothenberg et al. 2001; Rothenberg and Hogan 2006), which is a major chemotactic

factor for eosinophils (Mould et al. 1997; Mishra et al. 1999). However, other cells have

been shown to express the eotaxin-1 receptor chemokine receptor 3 (CCR3) and eotaxin can

be chemotactic for other leukocytes (Menzies-Gow et al. 2002). Many different factors can

mediate eosinophil recruitment aside from eotaxin-1, including the chemokines regulated on

activation, normal T-cell expressed and secreted (RANTES), some cytokines, complement

factors C3a and C5a, PAF, leukotrienes, integrins and acidic mammalian chitinase

(AMCase)(reviewed in Resnick and Weller 1993; Rothenberg and Hogan 2006). Upon

stimulation, circulating eosinophils adhere to the endothelial wall via expression of leukocyte

function-associated antigen (LFA)-1, very late antigen (VLA)-4, P-selectin glycoprotein

ligand (PSGL)-1 and Mac-1, which interact with their corresponding ligands on endothelial

cells (Rothenberg and Hogan 2006). The cells then migrate through the endothelium,

localising at the source of the inflammatory signal.

1.3.3.3 Eosinophil activation, secretion and degranulation

Once recruited to the site of infection, tissue injury or allergen exposure, eosinophils can

respond to a variety of stimuli, becoming activated and selectively releasing a range of

products. Typically, eosinophils circulating in the peripheral blood are far less active than

those present in tissues (Nonaka et al. 1999), but become more activated during recruitment

and extravasation. Eosinophil activation, manifested as the ability to express and release

granule proteins, is enhanced by the presence of IL-5 (Fujisawa et al. 1990). However under

various conditions, eosinophil degranulation can be triggered by cytokines such as IL-3 and

GM-CSF, chemokines, complement factors (C3b, C3a, C5a), PAF, ligation of FcγR and

factors released by tissue-invasive pathogens (Abu-Ghazaleh et al. 1992; Rothenberg and

Hogan 2006). The exact mechanism by which eosinophils release their granule contents is

not fully understood. Regulated exocytosis of cytoplasmic vesicles is the most commonly

CHAPTER ONE 14described mechanism, where eosinophils selectively release specific granules in a piecemeal

fashion (Henderson and Chi 1985; Dvorak et al. 1992; Logan et al. 2003; Clark et al. 2004).

Selective release of granule proteins in response to varying stimuli may explain the diversity

of functions for eosinophils. Eosinophils have also been demonstrated to degranulate by

cytolysis, where entire cytoplasmic contents are released non-specifically and this may cause

significant tissue pathology (Erjefalt et al. 1998).

1.3.3.4 Role of eosinophils in disease

Eosinophils can be destructive to the host, particularly in allergic diseases. Eosinophils are

commonly detected in elevated numbers in asthmatic humans and levels of the toxic granule

protein MBP are raised in bronchoalveolar lavage (BAL) fluid from such patients (Gleich et

al. 1979). IL-5 has been thought of as a useful target molecule to alleviate the symptoms of

asthma. Administering antibodies that inhibit IL-5 to guinea pigs (Mauser et al. 1993) and

mice (Corry et al. 1996) prevents the establishment of allergen-induced eosinophilia but was

not observed to greatly affect airways hyper-reactivity. However, in studies with IL-5-/- mice

(Foster et al. 1996) or mice with a complete lack of eosinophils (Lee et al. 2004), more

encouraging results were obtained, with animals failing to develop airways pathology and

hyper-reactivity after allergen challenge. In humans, trials have used anti-IL-5 antibodies to

target eosinophilia in an attempt to alleviate symptoms for asthmatic patients (Leckie et al.

2000). This study yielded discouraging results, where peripheral blood eosinophilia was

successfully reduced but symptoms were not significantly improved. Anti-IL-5 therapy may

not completely prevent eosinophilopoeisis or recruitment into tissues (Flood-Page et al. 2003)

and sufficient numbers remain after therapy, contributing to lung pathology and loss of

function. There remains some controversy as to the role of eosinophils in allergic asthma, but

it is now becoming apparent that eosinophils also contribute to other diseases, notably several

diseases affecting the gastrointestinal tract (Rothenberg 2004).

CHAPTER ONE 151.3.3.5 Role of eosinophils in killing helminths

Evidence that eosinophils protect against helminth infection was initially derived from

observations that these cells were found located around dead larvae from tissues and that the

magnitude of eosinophilia was positively correlated with the degree of resistance to infection

(Taliaferro and Sarles 1939; Butterworth 1984; Hagan et al. 1985). The precise mechanisms

by which eosinophils kill helminths remain unknown, though adherence to the organism and

degranulation may be critical. Early in vitro studies demonstrated that eosinophils kill S.

mansoni schistosomula in the presence of antibody and a heat-labile component of serum

(Butterworth et al. 1975; David et al. 1980). Eosinophils were observed to adhere to the

schistosomula and degranulate, releasing MBP directly onto the surface of the parasite.

Eosinophils are also effective at killing larval stages of T. spiralis (MacKenzie et al. 1980;

MacKenzie et al. 1981; Venturiello et al. 1995), N. brasiliensis (McLaren et al. 1977;

MacKenzie et al. 1981; Shin et al. 2001), Haemonchus contortus (Rainbird et al. 1998), H.

polygyrus (Penttila et al. 1983) and some filarial nematodes (Chandrashekar et al. 1990;

Brattig et al. 1991). In these studies, antibody appears to be important for eosinophil

adherence, degranulation and killing, but other serum factors also contribute to these

processes. In particular, the complement system is postulated to play an important role in

both the presence and absence of antibody (Butterworth 1984).

Infecting mice that display constitutive eosinophilia (IL-5 Tg mice) with helminths has

provided important insight into the role of eosinophils in resistance to helminth infection. IL-

5 Tg mice infected with N. brasiliensis (Dent et al. 1997a; Dent et al. 1997b; Shin et al. 1997;

Daly et al. 1999), A. cantonensis (Sugaya et al. 1997), Angiostrongylus costaricensis (Sugaya

et al. 2002), Litomosoides sigmodontis (Martin et al. 2000) and S. stercoralis (Herbert et al.

2000) have much lower parasite burdens than similarly treated WT mice. This is associated

with rapid eosinophil recruitment to sites of infection and restriction of parasite migration and

development. These results demonstrate that if eosinophils are sufficiently numerous and/or

CHAPTER ONE 16activated at the time of the initial infection, robust resistance can be mounted against some

helminths. In contrast, IL-5 Tg mice infected with parasites such as T. canis (Sugane et al.

1996; Dent et al. 1997a; Dent et al. 1999), S. mansoni (Dent et al. 1997b) or T. spiralis (Dent

et al. 1997a; Hokibara et al. 1997) were either no more resistant than WT animals or carried

high parasite loads, suggesting these parasites may have evolved strategies to evade attack by

eosinophils and that tissue eosinophilia may interfere with the function of other elements of

the immune response.

Studies using anti-IL-5 antibodies or IL-5-/- mice to diminish eosinophilia during helminth

infection have yielded mixed results in terms of the effect on parasite load. IL-5-/- mice

infected with Brugia malayi and S. ratti display higher worm burdens than WT mice during a

primary infection, however a lack of IL-5 does not impair resistance to re-infection with the

same parasites (Ovington et al. 1998; Ramalingam et al. 2003; Simons et al. 2005). Similar

results were observed for S. stercoralis, where IL-5-/- mice and mice treated with anti-CCR3

antibody had impaired killing of larvae during primary but not secondary infections (Herbert

et al. 2000; Galioto et al. 2006). Transfer of eosinophils or neutrophils to IL-5-/- mice

restored their ability to kill this parasite during primary infections (Galioto et al. 2006).

Significantly increased parasite burdens after anti-IL-5 antibody treatment were also observed

for infections with A. cantonensis (Sasaki et al. 1993), Onchocerca lienalis (Folkard et al.

1996) and S. venezuelensis (Korenaga et al. 1991). Therefore, for these helminths,

eosinophils are important for parasite killing during primary infections but may not be

required during secondary infections.

IL-5 and eosinophils do not appear to play a significant role in resistance to other helminth

species. IL-5 plays only a very minor role in immunity to T. spiralis, where anti-IL-5

antibody treatment of mice or infection of IL-5-/- mice had no or little effect on parasite

persistence in the intestine (Herndon and Kayes 1992; Vallance et al. 2000). IL-5-/- mice, or

CHAPTER ONE 17mice treated with anti-IL-5 antibodies exhibit similar parasite burdens to WT mice infected

with S. mansoni (Sher et al. 1990), N. brasiliensis (Coffman et al. 1989), T. muris (Betts and

Else 1999) or T. canis (Takamoto et al. 1997) despite effective blockade of the development

of eosinophilia. However, IL-5-/- or eosinophil-deficient ∆dblGATA mice exhibit higher

parasite fecundity than in WT mice during primary infections with N. brasiliensis (Knott et

al. 2007).

Eosinophils do degranulate in parasite-infected tissues but whether this contributes to parasite

killing is not clear (Hsu et al. 1974; von Lichtenberg et al. 1977; Kephart et al. 1984; Daly et

al. 1999). Preparations of eosinophil granule proteins can kill or immobilise parasite larvae in

vitro (Butterworth et al. 1979; Buys et al. 1981; Jong et al. 1981; Hamann et al. 1990), hence

degranulation may be important for eosinophil-dependent parasite killing. Whether

adherence to the parasite is essential for triggering degranulation is unclear. Mice deficient in

the granule proteins MBP and EPO develop higher worm burdens than WT animals infected

with L. sigmodontis (Specht et al. 2006), but the role of the eosinophil granule proteins in

killing other helminths in vivo is unclear. Resistance of mice to S. stercoralis (Abraham et al.

2004) and B. malayi (Ramalingam et al. 2005) is eosinophil-dependent, but not affected by an

absence of EPO and for B. malayi, resistance is also unaffected by the absence of MBP. This

suggests that for these parasites, EPO or MBP alone are not essential for parasite killing and

eosinophils possess other factors in their arsenal that are capable of damaging helminths.

1.3.3.6 Other roles for eosinophils during helminth infections

While the primary function of eosinophils during helminth infection may be active killing via

the release of toxic mediators, they may also contribute in other ways. There is evidence that

eosinophils can present antigens to T-cells (Weller et al. 1993; MacKenzie et al. 2001),

including helminth antigens that induce a Type-2 cytokine response (Padigel et al. 2006).

Eosinophils can also be a major source of cytokines during helminth infection, in particular

CHAPTER ONE 18by producing IL-4, which is important for induction of a Type-2 cytokine bias (Shinkai et al.

2002; Voehringer et al. 2004). While not directly demonstrated during helminth infection,

eosinophils are thought to be associated with tissue remodelling and wound healing in other

models (Bassett et al. 1977; Gouon-Evans et al. 2000; Sferruzzi-Perri et al. 2003), potentially

by the release of TGF-β (Wong et al. 1991) and hence could conceivably aid in repairing

tissues damaged by helminths after resolution of infection.

To summarise, while the role of eosinophils in killing parasitic helminths remains

controversial, there is considerable evidence to suggest that these cells can be effective at

killing or damaging some species of parasite if present in adequate numbers and at the

appropriate time during the course of an infection. Eosinophils appear to be most effective at

killing tissue-invasive helminths, though there are notable exceptions including T. canis, T.

spiralis and S. mansoni. For those studies where eosinophils were not demonstrated to play a

major role, it is possible that eosinophils were either not available in sufficient numbers to

exert an effect, or may only be effective against a particular and perhaps immature stage of

maturation of the parasite. It is also likely that some helminth species have evolved immune

evasion mechanisms that suppress eosinophil function. Certainly, parasites capable of

residing in host tissues for long periods would have been under strong pressure to evolve

strategies to evade damage induced by eosinophils. Equally, parasites such as N. brasiliensis

that migrate rapidly through tissues and normally have a relatively short period of residence

in the host are inadequately protected if eosinophils are present in large numbers at the onset

of infection. Eosinophils may be most effective against helminths soon after entry into the

host, particularly if the portal of entry is the skin. Understanding how eosinophils kill

helminths at these times may be critical for the development of effective vaccines that can

target these infective larval stages and prevent progression of the infection. The mechanism

by which eosinophils can be recruited to sites of infection, adhere to the target and kill by

degranulation (or by other mechanisms) is an area that requires further investigation.

CHAPTER ONE 19 1.4 THE COMPLEMENT SYSTEM

1.4.1 Function of the complement system

The complement system is important for both innate and adaptive immune responses to

infection with a variety of pathogens, mediating processes such as immune cell recruitment,

adhesion to the target, host cell activation, phagocytosis and direct lysis of small microbial

pathogens. The complement system consists of more than 35 fluid phase and cell-bound

proteins, most of which circulate as inactive enzymes until initiation events trigger activation

by proteolytic cleavage. Ultimately, the complement cascade can result in formation of the

membrane attack complex (MAC) which creates pores in cell membranes and may lead to cell

lysis. This is of importance for some smaller membrane-bound pathogens such as bacteria

and viruses, however due to their size and structure, the MAC is unlikely to cause significant

damage to most tissue-invasive parasitic helminths. However, certain events and factors

generated prior to the formation of the MAC may be advantageous for triggering immune

responses to helminths. Activation of complement occurs by three distinct routes (see section

1.4.2), but in all cases results in cleavage of the C3 molecule. Figure 1.1a details the

consequences of C3 activation schematically. C3 cleavage leads to generation of the

anaphylotoxin C3a, which can act as a chemotactic factor for leukocytes, and C3b which can

covalently bind to the surface of the pathogen or remain soluble (Rother and Till 1988).

Surface-bound C3b can act to cleave the C5 molecule into the anaphylotoxin C5a, another

chemoattractant molecule, and C5b which forms the first component of the MAC (C5b-9).

This covalent tagging of foreign molecules with C3b has other important functions, such as

opsonisation that facilitates recognition by leukocytes via CR1 (CD35). CR1 is found on

lymphocytes, erythrocytes and granulocytes such as eosinophils and neutrophils, where it

promotes phagocytosis (Gasque 2004). C3b can be further converted into iC3b by factor I-

mediated cleavage, in the presence of cofactors such as C4b and factor H (Rother and Till

Fi

gure

1.1

a: C

onse

quen

ces o

f C3

clea

vage

Figu

re 1

.1b:

Pat

hway

s to

com

plem

ent a

ctiv

atio

n

CHAPTER ONE 201988). The iC3b molecule is an “inactive” form of C3 in that it does not cause amplification

of the complement cascade. However, CR3 (CD11c/CD18) found on monocytes, neutrophils,

eosinophils and NK cells has high affinity for iC3b, which is also important for cell adhesion

and phagocytosis (Fischer et al. 1986; Rother and Till 1988; Gasque 2004). Further cleavage

of iC3b yields the fragments C3c, C3dg and C3d, the latter two of which can be recognised by

CR2 (Cooper et al. 1990). Although the complement system may be important in providing

protection against invading pathogens, when activated inappropriately it can be extremely

damaging to host tissues, as is seen in age-related macular degeneration, rheumatoid arthritis,

asthma, inflammatory bowel disease, ischemia-reperfusion injury and some infectious

diseases. Host cells are normally protected from the potentially damaging effects of

complement activation by various inhibitory molecules such as factor H, C1-inhibitors, C4-

binding protein, decay accelerating factor (DAF) and factor I (Gasque 2004).

1.4.2 Pathways to complement activation

There are three main pathways of initiation of the complement cascade, the classical, lectin

and alternative pathways, detailed schematically in Figure 1.1b. Cleavage of the C3 molecule

is central to all pathways. The classical pathway of complement activation begins primarily

with C1q recognition of pathogen-bound antibody, though C1q can recognise other pathogen-

associated molecules such as bacterial lipopolysaccharide and C-reactive protein (Gewurz et

al. 1993). C1q complexes with C1r and C1s in a Ca2+-dependent manner to form an esterase

that cleaves both C4 and C2 in the presence of Mg2+ (Rother and Till 1988). The resultant

cleavage factors C4b and C2a join to form the membrane-bound classical pathway C3

convertase (C4b2a) which then cleaves C3. The lectin pathway is similar to the classical

pathway, but does not require C1q and involves Ca2+-dependent recognition of microbial

carbohydrates by fluid-phase pattern recognition molecules such as MBL or ficolins (Endo et

al. 2006). This recognition triggers activation of the MBL-associated serine proteases

(MASP-1, MASP-2 and MASP-3), where MASP-2 cleaves and activates C2 and C4 to

CHAPTER ONE 21produce the same C3 convertase (C4b2a) found in the classical pathway (Wallis et al. 2007).

In addition, MASP-1 and MASP-2 have the ability to cleave C3 directly (Ambrus et al.

2003). The alternative pathway of complement activation is initiated by interactions with any

surface, including host cells, as well as carbohydrate-rich structures such as bacterial, yeast or

parasite cell walls. Activation is triggered by spontaneous hydrolysis of C3, resulting in

formation of a “C3b-like” molecule C3(H2O) (Rother and Till 1988). This combines with

factor B, which is cleaved by factor D to form C3(H2O)Bb, a C3 convertase that cleaves C3 to

form C3b, which then binds to the surface of the target. Metastable C3b then interacts with

factor B in the presence of Mg2+ to form C3bBb which is a C3 convertase that acts to generate

more C3b, hence amplifying C3b opsonisation of the microbial surface (Rother and Till

1988). If this process remained unimpeded it would result in consumption of all serum C3,

but it is normally controlled by negative regulators such as DAF, which blocks the interaction

between factor B and C3b (Kinoshita et al. 1986), and by factors I and H, which accelerate

conversion of C3b to inactive iC3b (Brown et al. 1983).

1.4.3 Complement-dependent immunity to pathogens

The complement system is typically the first line of defence against a wide range of

pathogenic organisms. Because microbial activation of the classical pathway usually requires

the presence of antibody, it is the alternative or lectin pathways of complement that would

typically be most important for innate immunity to bacteria and viruses in humans and other

animals (Kolble and Reid 1993; Petersen et al. 2001). However, naturally-occurring

antibodies reactive with micro-organisms are common and could activate the classical

pathway even without prior exposure of the host to a specific pathogen (Kozel et al. 1996;

Mold et al. 2002). Antibody-independent activation of the classical pathway may also play a

role, as has been demonstrated for both Gram negative (Loos et al. 1978) and Gram positive

(Eads et al. 1982) bacteria and for some viruses (Bartholomew and Esser 1980).

CHAPTER ONE 22Many species of bacteria, fungi, parasites and viruses have been shown to activate

complement in vitro; though the role of complement in vivo for immunity to these pathogens

is less well defined. Several methods of studying the role of complement in vivo are used.

One is the pharmacological depletion of complement components (typically C3) by treatment

of animals with cobra venom factor (CVF) which acts to generate an alternative complement

pathway convertase that is resistant to factor I-dependent inhibition (Shin et al. 1969). CVF-

treated animals are more susceptible to infection with pathogens such as Streptococcus

pneumoniae (Winkelstein et al. 1975), Candida albicans (Gelfand et al. 1978) and

Leishmania amazonensis (Laurenti et al. 2004). Similarly, CVF-treated mice immunised with

Escherichia coli are as susceptible to secondary infection as naïve mice (Ahlstedt 1981) and

CVF-treated chickens are highly vulnerable to infection with fowlpox virus (Ohta et al.

1986). While CVF-treatment of animals is a useful tool, C3 depletion is not absolute, with

levels of C3 reduced to 5-15% of those found in untreated animals (Jones and Ogilvie 1971).

More complete ablation of complement activity can be seen in animals with natural genetic

deficiencies in particular complement components. The mechanisms of complement-

dependent resistance can be probed in greater detail through the study of the consequences of

genetic mutations in C2, C4, C5 and C6 molecules. Humans deficient in C5 have been shown

to be extremely susceptibile to Neisseria gonorrhoeae and Neisseria meningitidis infections

(reviewed in Guenther 1983; Gianella-Borradori et al. 1990) and C3-deficient humans are

highly susceptible to Haemophilus influenzae type B (Winkelstein and Moxon 1992) and

various pyogenic bacterial infections. Studies with spontaneous-mutant C5-deficient mice

have demonstrated that the terminal arm of the complement cascade has an important function

during infection and the influence of complement is not just a consequence of C3 deposition

(Lovchik and Lipscomb 1993; Ferreira et al. 2000; Mullick et al. 2004).

Lastly and most recently, more comprehensive analysis of the role of complement has been

achieved using mice genetically engineered to be deficient in specific complement factors.

CHAPTER ONE 23This has the advantage that the roles of multiple factors can be analysed in the same host

species and strain. A large range of complement-deficient mice are now available and these

have been used in many studies to illustrate the in vivo role of various complement proteins in

immunity to bacterial, fungal, parasitic and viral infections (Wessels et al. 1995; Fischer et al.

1996; Matsumoto et al. 1997; Botto et al. 1998; Taylor et al. 2001; Pickering et al. 2002;

Mehlhop and Diamond 2006; Yuste et al. 2006).

Complement can also contribute to other elements of the immune response. Complement

receptors have been identified on lymphocytes and follicular dendritic cells (Carroll 2004)

and deficiency in C3 induced by CVF treatment (Pepys 1974) or genetic C3 or C4 deficiency

(Fischer et al. 1996) leads to impaired antibody responses to T lymphocyte-dependent

antigens. Complement is important for the activation of naïve B cells via complement

receptors CD21/CD35 and for persistence of antibody secretion (Carroll 2004). Elements of

the complement system can also modulate the function of T lymphocytes (Carroll 2004),

though its relative importance for T cell biology is unclear. Hence, complement can function

at several levels to provide resistance to infection with micro-organisms, from inducing the

activation of innate effector leukocytes to modulating adaptive, lymphocyte-dependent

responses during infection.

1.5 ROLE OF COMPLEMENT IN IMMUNITY TO HELMINTHS

Relatively little is known about the role of complement in vivo in providing resistance to

helminth infections. S. mansoni has been the most widely studied helminth with regards to

the role of complement, with varying outcomes between studies. CVF-treatment of mice

impairs both primary (Santoro et al. 1982) and secondary (Tavares et al. 1978) resistance to

S. mansoni in some studies, but not in others (Sher et al. 1982; Vignali et al. 1988). These

discrepancies can be attributed to differences in the timing and efficacy of the de-

CHAPTER ONE 24complementation treatment, as susceptibility to attack by complement can change as the

parasite matures. Skin-stage schistosomula are vulnerable to lysis by normal rat serum (NRS)

in vitro, however lung-stage schistosomula, which are resistant to complement activation in

vitro, are highly susceptible to complement in vivo (Vignali et al. 1988). In contrast, C5 or

C3 deficiency in mice has no impact on liver or intestinal parasite loads or egg production in

either primary or secondary S. mansoni infections (Ruppel et al. 1982; Sher et al. 1982; La

Flamme et al. 2003), though the development of a long-term Type-2 cytokine response is

compromised in C3-deficient animals (La Flamme et al. 2003).

The role of complement in host resistance to other helminth species can be even more

uncertain. CVF treatment of naturally-resistant strains of mice reduces the extent of killing of

T. taeniaeformis in the liver (Davis and Hammerberg 1988) and comparable outcomes are

observed when C5-deficient are used (Davis and Hammerberg 1990). Similarly, C5-deficient

mice are more susceptible to the development of large cysts when chronically infected with E.

granulosus and this correlates with reduced eosinophil recruitment to infected sites,

implicating C5a as a chemotactic factor (Ferreira et al. 2000). In contrast, C5 does not play a

role in innate or adaptive resistance to S. stercoralis, as C5-deficient mice efficiently recruit

granulocytes and larval killing is similar to that seen in WT mice (Kerepesi et al. 2006). For

S. stercoralis, killing of larvae by neutrophils during primary and secondary infections is C3-

dependent, suggesting that larval killing requires C3-mediated neutrophil adherence rather

than C5a-mediated cell recruitment (Kerepesi et al. 2006). Killing of S. stercoralis larvae

during secondary infections is IgG-dependent and sensitive to CVF treatment, suggesting that

the classical complement pathway is required (Ligas et al. 2003). Together, these studies

suggest that the relative importance and role of complement varies depending on the species

and life cycle of the parasite, the tissue infected and the nature of the humoral or cellular

inflammatory response. The following sections address the mechanism(s) through which the

complement system may protect against helminth infection.

CHAPTER ONE 25 1.5.1 Complement activation by helminths

Complement activation by helminths in vitro has been described for a number of species.

Deposition of the central molecule of the complement system (C3) on the parasite surface is

most often studied but there is also evidence that proteins excreted or secreted by some

parasites also activate complement (Baeza et al. 1994; Irigoin et al. 1996). Binding of other

molecules such as C1q, C4 (Linder and Huldt 1983) and MBL (Klabunde et al. 2000) to

parasites has also been examined. The generation of soluble factors such as C5a has not been

explored in vitro for parasitic helminths. Components of the terminal end of the complement

cascade (e.g. C5-C9) are also deposited on some helminth species (Ruppel et al. 1984;

Kennedy and Kuo 1988), though few studies have demonstrated that the MAC directly

damages helminths. For example, the early stages of some helminths such as cercariae and

newly-transformed schistosomula of S. mansoni (Marikovsky et al. 1986) and Echinococcus

granulosus oncospheres (Heath et al. 1994) are susceptible to complement-mediated lysis.

The pathway to initiation of in vitro complement activation has been described for some

helminths. It is clear that the relative importance of each pathway varies for different parasite

species and the nature of the host serum used. In the presence of serum from naïve animals,

the alternative pathway dominates for various life stages of T. spiralis (Hong et al. 1992), H.

polygyrus (Prowse et al. 1979) and Dirofilaria immitis (Abraham et al. 1988). Different

stages of S. mansoni have been shown to activate all three pathways, with schistosomula and

adult worms capable of triggering the alternative (Santoro et al. 1979; Marikovsky et al.

1986) and classical pathways (Santoro et al. 1979; Linder and Huldt 1983) and cercariae and

adult worms activating the lectin pathway (Klabunde et al. 2000). T. spiralis larvae can also

activate the lectin pathway (Gruden-Movsesijan et al. 2003). These studies are based on

methods such as the determination of sensitivity to Ca2+ depletion and Mg2+ chelation, C4

binding, consumption of complement components, serum C4-depletion and the use of C4 or

CHAPTER ONE 26C1q-deficient serum. As yet, no study has comprehensively investigated the role of all three

complement pathways for complement activation on any helminth species. This is now

feasible given the recent availability of mice genetically engineered to be deficient in various

complement factors.

Very few studies have established that complement is activated in vivo by helminths during

the course of an infection, the first study to do so was by Befus 1977. S. stercoralis L3

recovered from diffusion chambers implanted in skin of naïve and immunised mice were

coated with C3 and the extent of complement activation increased over a 6-24 hr period

(Brigandi et al. 1996). Furthermore, immune mice had higher levels of C3 deposition,

consistent with classical pathway activation. Activation of complement during E. granulosus

infection has been indirectly examined by analysing complement consumption, i.e. observing

a reduction of biologically active serum C3 after infection (Marikovsky et al. 1990; Diaz et

al. 1995).

1.5.2 Complement-dependent leukocyte-mediated killing of helminths

1.5.2.1 Recruitment of effector leukocytes

Little work has been done on the role of complement in cellular inflammatory responses

during helminth infections. The rapid generation of chemotactic factors C3a and C5a makes

this system a prime candidate for mediating early cellular inflammatory responses during an

acute helminth infection. Generation of C3a and C5a can also mediate mast cell activation

(Johnson et al. 1975), which can stimulate the recruitment of other inflammatory leukocytes

by releasing leukotrienes and chemokines (Lukacs et al. 1998; Malaviya and Abraham 2000).

Natural deficiency of C5 impairs eosinophil, but not neutrophil recruitment to the peritoneal

cavity three days post-secondary infection with E. granulosus (Ferreira et al. 2000) but does

not affect eosinophil recruitment to liver six days post primary infection with Taenia

taeniaeformis (Davis and Hammerberg 1990). Similarly, absence of C3, C3aR or C5 does not

CHAPTER ONE 27impair neutrophil or eosinophil recruitment into sub-cutaneous diffusion chambers 1-3 days

after primary or secondary S. stercoralis infection (Kerepesi et al. 2006).

1.5.2.2 Adherence and activation of effector leukocytes

Complement may allow attachment and activation of effector leukocytes, by opsonising the

surface of the parasite with C3 and its proteolytic products. Leukocyte adherence to parasitic

helminths in vitro is mostly dependent on complement and antibody. In the absence of

specific antibody (i.e. in the presence of naïve serum), complement is most important

(MacKenzie et al. 1980; MacKenzie et al. 1981; Butterworth 1984; Badley et al. 1987;

Desakorn et al. 1987; Venturiello et al. 1995; Shin et al. 2001), though other mediators such

as adhesion molecules can also play a role (Brattig et al. 1995; Shin et al. 2001). Eosinophil

and neutrophil adherence to H. polygyrus (Prowse et al. 1979; Penttila et al. 1983) and A.

cantonensis (Shaio et al. 1990) is complement-dependent in the presence of normal serum,

but antibody plays a greater role in immune serum. Whereas newborn T. spiralis larvae are

killed by eosinophils in the presence of antibody (Venturiello et al. 1995), eosinophil

adherence to and killing of infective-stage larvae is partially complement-dependent

(MacKenzie et al. 1980). Adherence of eosinophils and macrophages to N. brasiliensis

(MacKenzie et al. 1980; Egwang et al. 1985; Shin et al. 2001) and eosinophils to H. contortus

(Rainbird et al. 1998) promotes larval killing and this is similarly complement-dependent.

The pathway of complement activation that promotes cell adherence and killing of helminths

is unknown. Cell adherence to helminths does occur in vivo (Wang and Bell 1988; Melo et

al. 1990), and eosinophil degranulation can be detected in close proximity to parasite larvae

(Daly et al. 1999), however the mechanism by which these processes occur has not been

described.

1.5.3 Evasion of complement activation and leukocyte adherence

The complement system has ancient origins. Homologues of complement proteins such as C3

CHAPTER ONE 28have been isolated from the horseshoe crab (Limulus polyphemus), a “living fossil” that has

existed for at least 550 million years (Zhu et al. 2005). Hence, to enhance the chance of

survival within a host, some parasites have needed to evolve strategies to either avoid or limit

complement activation. S. mansoni recovered from either murine skin (Ruppel et al. 1984) or

lungs (Pearce et al. 1990) do not have C3 bound on their surface even though non-injected

infective-stage schistosomula are strong activators of complement in vitro. Similarly, T.

spiralis larvae recovered from muscle fix little C3 in vivo, but strongly activate complement

in vitro (Stankiewicz et al. 1989). There is evidence that the ability of complement and

leukocytes to bind to helminths changes as parasites mature to different life stages. Newborn

T. spiralis larvae or S. mansoni schistosomula activate complement more than their respective

adult parasitic stages (Marikovsky et al. 1990; Hong et al. 1992). Complement-dependent

leukocyte adherence to fourth-stage larvae of D. immitis (Abraham et al. 1988) and

Onchocerca volvulus (Brattig et al. 1991) is less pronounced than on third-stage larvae. The

mechanisms through which these parasites evade complement after only a short time within a

host is not fully understood, but may involve surface expression (Deng et al. 2003) or active

secretion (Badley et al. 1987; Marikovsky et al. 1988) of complement-activating or inhibitory

proteins, or by acquisition of host complement inhibitory factors (Meri et al. 2002).

In conclusion, the role of complement in immunity to parasitic helminths requires further

investigation. Importantly, there is very little information on the relative roles of individual

pathways of complement activation during infection with any helminth species. Considering

the large amount of in vitro evidence that complement contributes to killing of helminths by

facilitating the adherence of eosinophils and other leukocytes, as well as its potential for

promoting inflammatory cell recruitment to sites of infection, there is a significant gap in the

knowledge as to whether complement contributes to eosinophil-dependent killing of

helminths in vivo.

CHAPTER ONE 29

1.6 NIPPOSTRONGYLUS BRASILIENSIS AS A MODEL FOR

STUDYING IMMUNITY TO HELMINTHS

1.6.1 Parasite life cycle

The nematode N. brasiliensis is a parasite of rodents and has been widely used in

experimental studies because the life cycle of the parasite and the immune responses elicited

resemble those seen with some of the tissue-invasive gastrointestinal helminths that infect

humans and domesticated animals (Ogilvie and Jones 1971). N. brasiliensis infections in

mice and rats are convenient because the parasite is not pathogenic for humans, can be easily

maintained in the laboratory, does not require an intermediate host and has a short life-cycle

in these species. Figure 1.2 details the life cycle of N. brasiliensis in mice. In the natural

scenario, the life cycle of the parasite begins with the infective third-stage larvae (L3)

entering the host by penetrating through intact skin. Larvae then migrate from the skin to the

alveoli of the lungs over a period of 1-4 days where they moult and mature into fourth-stage

larvae (L4) (Kassai 1982). Larvae then migrate to the small intestine via the trachea,

oesophagus and stomach. In the small intestine, the parasite undergoes its final moult to

develop to the adult stage, a process that begins approximately five days post-infection (p.i.).

Eggs begin to appear in the faeces by day 7 p.i. and can persist until final parasite expulsion,

9-14 days after infection. Eggs hatch in the external environment and mature through two

moults from first-stage larvae (L1) to L3 (Kassai 1982).

Generally, the life cycle of the parasite in rats and mice is similar, however the total length of

the infection is usually shorter in mice (Kassai 1982). As gene knockout technology is far

easier to achieve and more advanced in mice than in rats, more recently much of the work

investigating immune responses to N. brasiliensis has been done in mice. For experimental

purposes, rodents are usually infected by subcutaneous injection of L3 in a minimal volume

Figu

re 1

.2:

Sche

mat

ic re

pres

enta

tion

of th

e lif

e cy

cle

of N

. bra

silie

nsis

CHAPTER ONE 30of liquid, allowing for more accurate control of delivery and dose. The kinetics of parasite

migration can be monitored experimentally by assessing escape from the skin, determining

lung larval burden 1-2 days p.i., with peak L4 numbers at day 2. Parasite migration to the

small intestine can be examined from day 3 to 7 p.i. and mature egg-producing adult worms

are recoverable in peak numbers from days 6-8 p.i.

The precise route that N. brasiliensis larvae take from the site of inoculation to reach the

lungs is unknown, but may involve transportation through the lymphatic and circulatory

systems (Clarke 1967). Larvae that reach the lungs are passively trapped in arterioles of the

pulmonary parenchyma before actively passing through into the terminal bronchioles and in

the process, cause significant haemorrhage and oedema of lung tissue. This is associated with

increased lung weight, mucus production and tissue damage (Ramaswamy and Befus 1993).

Upon reaching the small intestine, the parasite resides within the gastrointestinal mucosa

where it feeds off nutrients from the gut wall (Ogilvie and Jones 1971; Kassai 1982), growing

very rapidly and undergoing a final moult before reaching sexual maturity. The intestinal

epithelium is damaged during feeding by digestive enzymes released from the parasite’s

oesophageal and excretory glands and cellular debris is taken up via the parasite’s mouth

(Bottjer and Bone 1985). Worms preferentially inhabit the anterior third of the small

intestine, aggregating with other worms (Brambell 1965), presumably to facilitate mating.

Significant changes in the distribution pattern of intestinal worms has been attributed to

immune-mediated damage to the parasite prior to or during the intestinal stage of infection

(Kassai 1982; Dent et al. 1999).

1.6.2 Immune responses to N. brasiliensis

1.6.2.1 Cellular inflammatory responses

The cellular inflammatory response to N. brasiliensis begins very soon after injection into

CHAPTER ONE 31skin. Subcutaneous injection of naïve WT mice typically results in a cellular inflammatory

response that peaks 2-8 h p.i. and is comprised mostly of neutrophils (Taliaferro and Sarles

1939; Daly et al. 1999). This cellular infiltrate subsides two days after infection, by which

time most larvae have migrated from the skin. In fact, in WT mice, relatively few larvae are

recoverable from skin as little as two hours after inoculation (Daly et al. 1999). Hence,

neutrophils that are recruited to the site of infection are largely ineffective at damaging or

killing N. brasiliensis larvae. Eosinophils, lymphocytes and macrophages are also recruited

to N. brasiliensis-infected skin during primary infections of WT mice, albeit in smaller

numbers. The factors which promote this early inflammatory response to N. brasiliensis

infection have not been examined.

Within 4 days of infection, eosinophil numbers in the bone marrow increase significantly

(Rennick et al. 1990) and substantial peripheral blood eosinophilia develops within 10-13

days p.i. (Coffman et al. 1989; Urban et al. 1993). An acute inflammatory response in the

lung peaks 6-9 days after initial infection (Ramaswamy and Befus 1993; Arizono et al. 1996;

Daly 1999; Voehringer et al. 2004), 2-5 days after the larvae had left this site, en route to the

small intestine. There is also evidence that this belated inflammatory response is biphasic,

such that a second substantial peak response occurs approximately 16 days after initial

infection (Ramaswamy and Befus 1989). Curiously, during the short period in which N.

brasiliensis resides in the lungs (1-2 days), the inflammatory response is very mild relative to

that in the skin (Daly 1999; McNeil et al. 2002). The reason for this is unclear and there is

also no obvious explanation for the development of a substantial but delayed and biphasic

inflammatory response after the parasite has left the lungs. Eosinophils are the most prevalent

cell type in this late inflammatory response (Voehringer et al. 2004), though there has been

much interest in IL-4-producing CD4+ T-cells found at 8-12 days p.i. (Shinkai et al. 2002).

The chemoattractant molecules promoting cell recruitment to skin may therefore not be

generated in the lung, or alternatively the process of cell recruitment may be inactivated by

CHAPTER ONE 32the parasite. The cellular immune response in the intestine following N. brasiliensis infection

involves the participation of mast cells and goblet cells, as described in section 1.2.3.

The rapid nature of the life cycle of the N. brasiliensis parasite in naïve mice ensures that it

typically escapes a tissue before the cellular inflammatory response mounted against it is able

to trap the parasite. This is certainly of benefit during primary infections as it means the

parasite is not extensively damaged in transition to the small intestine. However if sufficient

effector cells of the right type can be recruited, for example during secondary infections with

the parasite or perhaps if the host has a concurrent infection with another pathogen, a more

effective immune response can be elicited, resulting in reduced parasite migration, maturation

and egg production (Taliaferro and Sarles 1939; Kassai 1982).

1.6.2.2 Cytokine responses

The cytokine response to N. brasiliensis, along with a number of other helminthic parasites, is

a predominately Type-2 biased response (Kopf et al. 1993; Grencis 1997; McKenzie et al.

1998; Min et al. 2004). The type-2 cytokine response following N. brasiliensis infection is

described in detail in earlier sections, in particular the rapid generation of IL-4-producing

cells (see section 1.2.3), the importance of STAT6 and IL-13 in worm expulsion (see section

1.2.4) and the role of IL-5 in stimulating eosinophil responses (see section 1.3.3.1).

1.6.2.3 Role for eosinophils

While the role of eosinophils in killing some species of helminth remains controversial, the

evidence for these cells as effectors in immunity to N. brasiliensis is strong. Eosinophils have

been shown to adhere to L3 in vitro and release their granule contents (McLaren et al. 1977;

MacKenzie et al. 1981) and can damage the parasite (Shin et al. 2001), such that the larvae

have impaired ability to migrate when subsequently injected into a naïve murine host (Daly et

al. 2004). As discussed in section 1.3.3, the exact mechanisms by which eosinophils kill

CHAPTER ONE 33helminths, including N. brasiliensis in vitro are not completely understood, though adherence

of eosinophils to the parasite has been shown to be antibody and/or complement-dependent

(MacKenzie et al. 1980; Shin et al. 2001). It is not known whether adherence and/or

degranulation of these cells are absolutely required for parasite killing. Other leukocytes such

as macrophages, mast cells and neutrophils have been shown to adhere to N. brasiliensis in

vitro (MacKenzie et al. 1980; Egwang et al. 1984; Egwang et al. 1985), but only

macrophages and eosinophils have been demonstrated to be able to damage the parasite.

Studies with N. brasiliensis infections in IL-5 Tg mice clearly demonstrate that pre-existing

eosinophilia is associated with potent and early resistance, even in naïve animals (Dent et al.

1997a; Shin et al. 1997). These mice exhibit drastically reduced numbers of N. brasiliensis

lung larvae, intestinal worms and faecal eggs when compared to similarly-treated WT mice.

This resistance develops extremely rapidly (within hours) and occurs prior to the lung stage

and independently of adaptive immune mechanisms. Larvae are attacked by eosinophils in

the skin and may be trapped for up to 24-48 h (Daly et al. 1999). Eosinophils also provide

potent resistance to N. brasiliensis during secondary infections, as IL-5-/- and eosinophil-

deficient ∆dblGATA mice exhibit higher lung larval burdens than WT mice 48 h post-

secondary challenge (Knott et al. 2007).

The mechanism of eosinophil-dependent killing of N. brasiliensis in vivo is unknown. Rapid

recruitment of these cells to infected skin, recognition of the parasite and attachment followed

by release of toxic granule proteins may be critical processes. While multiple factors have

been shown to be important for eosinophil recruitment (see section 1.3.3.2) and adherence

(see section 1.3.3.5) in other models, the complement system may be most critical as it can

mediate both processes and potentially trigger degranulation (Takafuji et al. 1996; Egesten

and Malm 1998). Furthermore, the complement cascade can be triggered very rapidly and

independent of adaptive immunity and so it satisfies the criteria as a rapid innate effector

CHAPTER ONE 34mechanism. Complement activation in the very early stages of infection may therefore be

critical for eosinophil-dependent resistance to N. brasiliensis. Until recently, our knowledge

of interactions between the complement system and N. brasiliensis has largely been limited to

in vitro studies (MacKenzie et al. 1980; MacKenzie et al. 1981; Egwang et al. 1984; Shin et

al. 2001). However, in one study of CVF-treated rats, the rate of worm expulsion 15 days p.i.

was unaffected (Jones and Ogilvie 1971). The impact of complement depletion on earlier

stages of N. brasiliensis migration and inflammatory responses in the host has not been

reported.

1.7 INTRODUCTION TO THIS STUDY

The present studies will address the role of complement in the immune response of mice to

infection with the nematode N. brasiliensis. The N. brasiliensis model is a useful model in

which to study the role of complement. Despite the fact that naïve WT mice possess only

modest resistance to the parasite, potent innate immunity is seen in eosinophilic IL-5 Tg mice

and WT mice develop robust resistance to re-infection. The hypotheses underpinning this

thesis are that, during N. brasiliensis infection, complement:

1. stimulates recruitment of effector cells, such as eosinophils;

2. promotes interactions of these cells with the parasite surface;

3. enhances primary and/or secondary resistance of WT mice and

4. facilitates eosinophil-dependent primary resistance of IL-5 Tg mice.

The first aim was to comprehensively characterise the nature of the interactions between N.

brasiliensis, complement proteins and eosinophils. In particular, we were interested in the

specific pathway(s) that mediate complement activation on N. brasiliensis in vitro and if this

mediated the adherence of eosinophils to the parasite surface. To do this we first needed to

develop a new method for analysing and quantifying interactions between complement

CHAPTER ONE 35proteins, eosinophils and parasitic worms. Secondly, we aimed to establish the role of

complement in providing resistance of mice to N. brasiliensis infections. Our focus was on

the role of complement in eosinophil-dependent anti-parasite resistance in the early stages of

infection.

It is expected that this study will provide a detailed analysis of the function of the

complement system during infections with tissue-invasive parasitic helminths.

CHAPTER TWO 36

CHAPTER TWO: QUANTITATION OF COMPLEMENT AND

LEUKOCYTE BINDING TO A PARASITIC HELMINTH SPECIES

1Paul R. Giacomin, 1Hui Wang, 2David L. Gordon and 1Lindsay A. Dent

1School of Molecular and Biomedical Science, University of Adelaide, North Tce, South

Australia, Australia

2Department of Microbiology and Infectious Diseases, Flinders Medical Centre, University of

South Australia, Bedford Park, Adelaide, Australia.

Journal of Immunological Methods-2004 Jun, volume 289, issue 1-2, pages 201-210 doi:10.1016/j.jim.2004.04.024

CHAPTER TWO 37

LINKAGE TO CHAPTER TWO

Complement is the immune system’s first line of defense against invading pathogens and the

notion that it may promote protective immunity to parasitic helminths has been long-held.

The primary importance of complement in this context may be to aid in leukocyte-mediated

immunity, by mediating recruitment of these cells to infected tissues and their adherence to

the parasite. Helminths are strong activators of complement in vitro (Santoro et al. 1979) and

several leukocyte subsets can adhere to these parasites in both complement-dependent and

independent manners (MacKenzie et al. 1981). The importance of the complement system

may vary depending on the species of the helminth, the stage of parasite maturation and the

species of the host. Much of our understanding of the nature of the interactions between

complement proteins, helminths and leukocytes has been derived from in vitro studies. While

these studies gained valuable information regarding the importance of complement, the

methodologies employed had some major limitations, where they were often purely

qualitative, subjective and inefficient. Previously used methods for quantifying complement

activation and cell adherence on helminths are outlined in greater detail in the introduction to

Chapter Two.

The broad aim of this research project was to further our understanding of how complement

may promote resistance to parasitic helminth infection. Determining the specific pathway of

complement activation involved was a major focus, which initially required the development

of improved in vitro assays that allowed sensitive, automated and objective quantification of

complement deposition and cell adherence to helminths.

In summary, Chapter Two describes novel fluorescence-based methods for analysing and

quantifying both the activation of complement on a species of helminth (N. brasiliensis) and

the adherence of eosinophils to this same parasite. Importantly, the new techniques were used

CHAPTER TWO 38to establish the pathway involved in activation of human and mouse complement on N.

brasiliensis infective larvae and the role of complement in eosinophil adherence to the

nematode.

Giacomin, P.R., Wang, H., Gordon, D.L. and Dent, L.A. (2004) Quantitation of complement and leukocyte binding to a parasitic helminth species. Journal of Immunological Methods, v. 289, (1-2), pp. 201-210, June 2004

NOTE: This publication is included in the print copy of the thesis

held in the University of Adelaide Library.

It is also available online to authorised users at:

http://dx.doi.org/10.1016/j.jim.2004.04.024

CHAPTER THREE 39

CHAPTER THREE: LOSS OF COMPLEMENT ACTIVATION AND

LEUKOCYTE ADHERENCE AS NIPPOSTRONGYLUS BRASILIENSIS

DEVELOPS WITHIN THE MURINE HOST

1Paul R. Giacomin, 1Hui Wang, 2David L. Gordon, 3Marina Botto and 1Lindsay A. Dent

1School of Molecular and Biomedical Science, University of Adelaide, North Tce, South

Australia, Australia

2Department of Microbiology and Infectious Diseases, Flinders Medical Centre, University of

South Australia, Bedford Park, Adelaide, Australia.

3Molecular Genetics and Rheumatology Section, Faculty of Medicine, Imperial College,

London W12 0NN, England, UK.

Infection and Immunity-2005 Nov, volume 73, issue 11, pages 7442-7449 doi:10.1128/IAI.73.11.7442-7449.2005

CHAPTER THREE 40

LINKAGE TO CHAPTER THREE

Chapter Two outlined the development of new and improved methods for quantification of

the binding of complement proteins and leukocytes to parasitic helminths. Development of

these methodologies was critical for the studies comprised within Chapter Three.

Implementation of these methods generated important experimental data regarding the in vitro

interactions between the complement system, leukocytes and helminths. The mechanism of

deposition of both human and mouse C3 was determined and interestingly, the pathway by

which complement was activated differed depending on the species of serum used. Human

C3 was deposited either via the Ca2+-dependent classical or lectin pathways, we could not

conclusively determine the relative contribution of each individual pathway due to lack of

reagents. Activation of the classical pathway in naïve human serum would be unlikely to

have occurred due to the absence of parasite-specific antibody, whether antibody-independent

classical pathway activation occurred was not resolved. The fact that the Ca2+-independent

alternative pathway dominated for mouse C3 deposition was expected, as the alternative

pathway has been shown to be important for complement activation on a number of species of

helminth (Prowse et al. 1979; Santoro et al. 1979; Abraham et al. 1988; Hong et al. 1992).

As N. brasiliensis is a parasite that infects rodents but not humans, the primary focus for the

remainder of this study was on the role of the mouse complement system in immunity to the

parasite. While methods of Ca2+/Mg2+ depletion are useful for some purposes, they are not

very specific as EGTA can chelate Mg2+ at high concentrations. Hence, a more conclusive

demonstration of the relative roles of each complement pathway is required and this will

involve the use of serum taken from mice genetically engineered to be deficient in various

complement factors. The outcomes of these studies are included in the following chapter.

Chapter Two demonstrated that complement is the major contributor to the adherence of

eosinophil-rich mouse peritoneal leukocytes to infective N. brasiliensis larvae. This was in

CHAPTER THREE 41agreement with previous studies using the N. brasiliensis model as well as some other

helminths (MacKenzie et al. 1980; MacKenzie et al. 1981; Butterworth 1984; Badley et al.

1987; Desakorn et al. 1987; Venturiello et al. 1995; Shin et al. 2001). Nevertheless, some

degree of cell adherence did occur in the presence of serum depleted of C3 activity by heat

treatment, suggesting complement-independent factors can contribute, though this may be a

minor pathway. Methods for heat-inactivation of serum may not be 100% effective, hence

using serum deficient in various complement factors will clarify the influence of complement

on cell adherence to N. brasiliensis L3, while also determining the precise pathway involved.

This will represent the most definitive demonstration of the role of each individual

complement pathway in mediating eosinophil adherence to a parasitic helminth species; the

results of this study are included in Chapter Three.

As N. brasiliensis is a parasite that undergoes several stages of maturation as it migrates

through different tissues, the ability of the complement system and leukocytes to interact with

the parasite may change. This may be part of an immune evasion mechanism used by the

parasite to avoid complement recognition, inflammatory responses, eosinophil adherence and

eosinophil degranulation. The results from Chapter Two clearly indicate that infective-stage

L3 are sensitive to activation of the mouse alternative complement pathway and adherence of

eosinophils. The ability of complement and leukocytes to bind to other life stages of N.

brasiliensis, e.g. skin-recovered larvae, lung-stage L4 or adult intestinal worms is unknown.

Hence, the capacity for in vivo binding of host-derived complement as well binding of

exogenous C3 from NMS will be assessed for each of the aforementioned life-stages. The

specific pathway to complement activation will also be compared. Determining if and when

complement and eosinophils are effective at recognising N. brasiliensis will be important for

addressing our final goal of examining the role of complement in vivo.

CHAPTER THREE 42In summary, the following chapter compares the ability of mouse complement and

eosinophil-rich leukocytes to bind to infective-stage L3, skin-recovered larvae, lung-stage L4

and adult intestinal worms, with a particular focus on determining the most important

complement pathway involved using various complement-deficient sera.

Giacomin, P.R., Wang, H., Gordon, D.L., Botto, M. and Dent, L.A. (2005) Loss of complement activation and leukocyte adherence as Nippostrongylus brasiliensis develops within the murine host.Infection and Immunity, v. 73 (11), pp. 7442-7449, November 2005

NOTE: This publication is included in the print copy of the thesis

held in the University of Adelaide Library.

It is also available online to authorised users at:

http://dx.doi.org/10.1128/IAI.73.11.7442-7449.2005

CHAPTER FOUR 43

CHAPTER FOUR: THE ROLE OF COMPLEMENT IN INNATE,

ADAPTIVE AND EOSINOPHIL-DEPENDENT IMMUNITY TO THE

NEMATODE NIPPOSTRONGYLUS BRASILIENSIS

1Paul R. Giacomin, 2David L. Gordon, 3Marina Botto, 4Mohamed R Daha, 5Sam D.

Sanderson, 6Stephen M. Taylor and 1Lindsay A. Dent

1School of Molecular and Biomedical Science, University of Adelaide, North Tce, South

Australia, Australia

2Department of Microbiology and Infectious Diseases, Flinders Medical Centre, University of

South Australia, Bedford Park, Adelaide, Australia.

3Molecular Genetics and Rheumatology Section, Faculty of Medicine, Imperial College,

London, UK.

4Leiden University Medical Center, Department of Nephrology, Albinusdreef 2, Leiden, The

Netherlands

5University of Nebraska Medical Center, Omaha, Nebraska, USA

6School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia

Published online: Molecular Immunology doi:10.1016/j.molimm.2007.05.029

Full citation: Molecular Immunology-2008 Jan, volume 45, issue 2, pages 446-

455

CHAPTER FOUR 44

LINKAGE TO CHAPTER FOUR

Chapter Three described that the ability of complement and eosinophils to recognise N.

brasiliensis changes as the parasite matures. Infective-stage larvae were the most susceptible

to complement activation by the alternative pathway and cell adherence, though the lectin

pathway played a minor role in causing C3 deposition. Complement was activated in vivo on

larvae injected into skin air pouches; however parasites recovered from lungs or adult worms

taken from the intestine did not have complement bound on their surface. Furthermore, while

adult worms did bind C3 and leukocytes via alternative pathway activation after treatment

with exogenous serum and peritoneal cells, lung-stage larvae resisted complement activation

and cell adherence and the small amount of C3 that did bind was via lectin pathway

activation. These findings have important implications for the next part of the study which

aims to investigate the role of complement in immunity to N. brasiliensis in vivo. This will be

addressed using the various lines of complement-deficient mice whose sera we used in the

studies described in Chapter Three. Specifically, mice deficient in either the classical

pathway (C1qa-/-), alternative pathway (Bf-/-) or all complement pathways (C3-/-) were used.

To address the question of whether complement mediates eosinophil-dependent resistance to

N. brasiliensis, eosinophilic IL-5 Tg mouse strains deficient in C1q, factor B or C3 were

generated. We then used these mouse strains in the well-established N. brasiliensis model,

analysing immunity during both primary and secondary parasite infections.

Complement and eosinophils may be most effective in the very early stages of innate N.

brasiliensis infection, soon after parasite entry into the skin, as once the parasite reaches the

lungs their impact may be reduced (see Chapter Three). Hence, much of Chapter Four

focused on the early events of N. brasiliensis infection (0-150 min p.i.) using a skin air pouch

model. Parameters such as in vivo C3 deposition on the parasite, leukocyte adherence,

inflammatory cell recruitment and eosinophil degranulation were monitored. In addition, rate

CHAPTER FOUR 45of parasite migration from the skin to the lungs and the small intestine in normal and

complement-deficient mice with or without IL-5 transgene expression were assessed. Our

hypothesis, based on our findings from Chapter Three, was that the alternative pathway would

be most important for immunity to N. brasiliensis. Hence, absence of factor B in mice may

result in reduced inflammatory leukocyte recruitment to the skin, attachment of these cells to

the parasite and consequently the rate of parasite migration would be enhanced compared to

WT or single-mutant IL-5 Tg control mice.

In summary, the following chapter describes the role of various pathways of complement

activation in immunity of mice to N. brasiliensis infection with a focus on their role in

promoting eosinophil-dependent resistance to the parasite.

Paul R. Giacomin, P.R., Gordon, D.L., Botto, M., Daha, M.R., Sanderson, S.D., Taylor, S.M. and Dent, L.A. (2008) The role of complement in innate, adaptive and eosinophil-dependent immunity to the nematode Nippostrongylus brasiliensis. Molecular Immunology, v. 45 (2), pp. 446-455, January 2008

NOTE: This publication is included in the print copy of the thesis

held in the University of Adelaide Library.

It is also available online to authorised users at:

http://dx.doi.org/10.1016/j.molimm.2007.05.029

CHAPTER FIVE

CHAPTER FIVE: DISCUSSION AND CONCLUSION

CHAPTER FIVE 46

5.1 GENERAL DISCUSSION

Eosinophils equip the host immune system with a potent weapon against infection with

parasitic helminths. The precise mechanisms used by these cells to damage helminths are

unknown, though previous in vitro research has suggested that complement-mediated

adherence to the surface of the parasite may be important (MacKenzie et al. 1980;

Butterworth 1984; Shin et al. 2001). Furthermore, complement activation may be critical for

leukocyte recruitment to parasite-infected tissues. The aim of the present study was to

investigate the pathways to complement activation important for the in vitro interaction of

eosinophils with N. brasiliensis and also to determine if complement was important to provide

eosinophil-dependent resistance to helminth infections.

5.1.1 Summary of main findings

N. brasiliensis L3 activate human and mouse complement when exposed to serum in vitro and

we designed a novel assay that allowed rapid, automated and objective quantification of

complement activation and cell adherence to N. brasiliensis (Giacomin et al. 2004). The

assay was used to demonstrate that N. brasiliensis L3 activate human complement via the

classical or lectin pathways. In contrast, the alternative pathway mediated mouse C3

deposition on L3 and this was the primary mediator of the adherence of eosinophil-rich mouse

leukocytes in vitro (Giacomin et al. 2005). However, the roles of complement in vivo are

more complex. N. brasiliensis larvae did activate the alternative pathway of complement 30

min after injection into the skin of mice and this correlated with aggregation of larvae.

Unexpectedly, cell adherence to the parasite and larval aggregation at this site also occurred in

complement-deficient mice over a longer time frame of up to 2.5 h (Giacomin et al. 2008). In

the absence of the alternative complement pathway, eosinophil degranulation in the skin 30

min post-parasite injection was reduced, but eosinophils efficiently degranulated in the

absence of complement at a later time. The presence of the alternative complement pathway

CHAPTER FIVE 47 and the C5aR was critical for eosinophil recruitment to the skin 30 min p.i., though other

factors recruited eosinophils at later times. Interestingly, parasites recovered from the lungs

resisted complement activation and leukocyte adherence when exposed to serum and

eosinophil-rich leukocytes ex vivo and the small amount of complement that did bind was via

the lectin pathway of activation. Evasion of complement activation by the parasite began in

the first 2.5 h of infection, while the parasite resided within the skin and this may explain in

part why complement was of most significance in the first 30 min of infection. Intestinal

worms bound complement and leukocytes ex vivo via the alternative pathway of activation,

but not in vivo. Absence of the alternative complement pathway caused slight but significant

increases in lung larval burdens in normal and IL-5 Tg mice during a primary infection.

However, IL-5 Tg mice remained highly resistant to primary N. brasiliensis infection even in

the complete absence of complement, suggesting that eosinophils can provide resistance in a

complement-independent manner. Similarly, robust resistance of mice to secondary N.

brasiliensis infection was complement-independent. In conclusion, the alternative

complement pathway is important for mediating interactions of eosinophils with N.

brasiliensis in vitro and in the very early stages of a parasite infection. However evasion of

the complement system by the parasite and the participation of complement-independent

factors over time means its influence is reduced and that overall, complement does not

contribute greatly to immunity of mice to N. brasiliensis infection.

5.1.2 Complement and eosinophil-dependent immunity to helminths

5.1.2.1 Eosinophil-dependent resistance to helminths

While the role of eosinophils during helminth infection remains somewhat controversial

(Behm and Ovington 2000; Meeusen and Balic 2000), eosinophils are undoubtedly effective

mediators of resistance to N. brasiliensis in mice. One of the most conclusive pieces of

evidence for this was obtained using eosinophilic IL-5 Tg mice, where larvae were trapped at

the skin injection site within the first 2 h, with fewer larvae migrating to the lungs and small

CHAPTER FIVE 48 intestine than in WT mice (Dent et al. 1997a; Daly 1999; Daly et al. 1999). The results from

the present study similarly support a role for eosinophils in resistance to helminths. We

hypothesised that complement would be particularly important for mediating innate killing of

the parasite by eosinophils at the site of initial infection, in the skin. We therefore generated

IL-5 Tg mouse strains deficient in various complement factors. Our evidence clearly

demonstrates that in this model, and contrary to our initial hypothesis, complement-deficient

IL-5 Tg mice are almost as resistant to primary N. brasiliensis infection as IL-5 Tg animals.

Given that at 30 min p.i., eosinophil recruitment and degranulation, as well as larval

aggregation were reduced in the absence of factor B, we expected that eosinophils would have

impaired ability to restrict the migration of N. brasiliensis larvae. There was some evidence

that trapping of larvae in the skin of IL-5 Tg mice 150 min p.i. was partially dependent on the

alternative pathway. In the present study, the entrapment of larvae in the skin was not as

extensive as seen previously (Daly et al. 1999), perhaps due to differences in the strain of the

mice used. Nevertheless, overall resistance to N. brasiliensis was not impaired in the absence

of C3 or factor B, as parasite burdens in the lungs or small intestine were not greatly enhanced

compared to IL-5 Tg. This implies that eosinophils can efficiently limit N. brasiliensis

migration in the absence of complement and in our IL-5 Tg model, without specific antibody.

Damage to the parasite may be inflicted at the site of initial infection and possibly at other

sites in the pre-lung stage. The precise pathway that N. brasiliensis undertakes to get to the

lungs has not been conclusively established, though the parasite may pass through the

peripheral blood and lymphatic systems (Clarke 1967). Alternatively, damage incurred in the

skin may impair the ability of the parasite to colonise the small intestine, in particular. This

highlights one of the limitations of our “snapshot” method for analysing parasite migration

through the host, where mice are sacrificed at specific time points and only a limited number

of organs are examined for parasites. In the future, parasite migration in vivo should be

analysed using techniques such as whole-body imaging, where larvae labeled with a

fluorochrome or similar indicator might be tracked through a living rodent host, as has been

CHAPTER FIVE 49 done for green-fluorescent protein (GFP)-expressing bacteria (Zhao et al. 2001). Purified

eosinophils could then be labeled with a different colour dye and injected into the rodent so

that these cells could be tracked simultaneously to establish where they interact and co-

localise with the parasite.

5.1.2.2 Role of complement in vivo and in vitro

The present study included a comprehensive examination of the role of complement during

infection with a species of parasitic helminth, taking it from an in vitro setting through to its

role in vivo under various levels of immunity. These findings highlight that care must be

taken when trying to extrapolate in vitro observations to the in vivo situation. Our in vitro

work, and that done by others (MacKenzie et al. 1980; Shin et al. 2001), suggest that

eosinophils adhere to N. brasiliensis L3 via activated complement proteins and this is

important for parasite killing. However, while the alternative complement pathway did

mediate several processes in vivo that may be important for inflicting damage to the parasite

(C3 deposition, eosinophil and neutrophil recruitment, EPO release, larval aggregation), other

mechanisms compensated for a lack of complement and these may not be present in vitro.

Our results echo those from previous investigations on the role of complement in immunity to

other helminth species. For example, complement may contribute to some important immune

responses such as leukocyte recruitment (Ferreira et al. 2000; Giacomin et al. 2008), cytokine

responses (La Flamme et al. 2003) and cellular cytotoxicity against a particular larval stage or

within an individual infected tissue (Giacomin et al. 2004; Giacomin et al. 2005; Kerepesi et

al. 2006), but ultimately ablation of complement may not greatly affect parasite infection

kinetics or worm burdens. Complement can and probably does play an important role during

N. brasiliensis infection of normal mice, but there are compensating factors that may be

accentuated in complement-deficient mice. This is an inherent problem when working with

genetically modified mice. Mice surviving a gene deletion may only be able to do so if

capable of compensating for the deletion by up-regulating what may otherwise be relatively

minor homeostatic or defensive mechanisms. This, compounded with evasion mechanisms

CHAPTER FIVE 50 used by the parasite in vivo, may limit the efficacy of the complement system in anti-parasite

resistance.

We have classified the relative importance of the three pathways of complement activation

and established that in the mouse, the alternative pathway plays the greatest role in vivo and in

vitro. The lectin pathway does contribute to in vitro C3 deposition on L3 and L4 but we were

unable to directly determine the relative importance of the lectin pathway in vivo in this

infection model. This might be addressed by using C4-deficient mice, but these animals are

also deficient in the classical pathway (Fischer et al. 1996). Similarly, MBL-deficient lines of

mice have been produced, but the lectin pathway can be initiated by binding of a variety of

lectins other than MBL (Chan et al. 2006). In any case, the absence of C3 in mice caused no

greater defect than ablation of factor B for any of the parameters studied and so the lectin

pathway would seem to play little or no role in the overall immunity to N. brasiliensis.

Interestingly, the pathway to complement activation on L3 differed depending on whether

human or mouse sera were used (Giacomin et al. 2004), leading to the possibility that the role

of complement may vary depending on the species of the host. It is very common for the

immune response to helminth infection to be studied in mice using non-natural parasite

species, hence care must be taken when making conclusions from such studies. N.

brasiliensis is a parasite that naturally infects both rats and mice (Kassai 1982), but there is

evidence that immune responses to non-natural parasites can include enhanced non-specific

inflammation and parasite rejection (Meeusen and Balic 2000). Perhaps complement

activation is enhanced, or alternatively immune evasion mechanisms used by the parasite are

less effective during infection with non-natural helminth species. Indeed, studies with non-

natural parasites of mice have yielded more positive results regarding the role of complement

during helminth infection than we have in the present study (Santoro et al. 1982; Ferreira et

al. 2000; Kerepesi et al. 2006). Humans are accidental hosts for some species of helminth

CHAPTER FIVE 51 (e.g. T. canis) and can cause significant pathology, so understanding the immune responses to

both natural and non-natural helminths is important.

5.1.2.3 Complement-dependent eosinophil recruitment to parasite-infected skin

The present study has advanced our knowledge of how eosinophils are recruited to sites of

helminth infection. Activation of the alternative complement pathway played an important

role in the early C5aR-dependent recruitment of eosinophils to the skin during N. brasiliensis

infections. We did not directly test whether C5a was generated in the skin of mice during

infection, however there are ELISAs available that could be used to measure mouse C5a in air

pouch supernatants. One point to consider is that C5a (and C5a des-Arg) also bind to the

C5L2 receptor (Okinaga et al. 2003), which is not blocked by the C5aR inhibitor we used.

Hence it is conceivable that C5a could react with leukocytes via different pathways, though

C5L2 is considered to be a non-signalling C5a receptor with little biological role (Okinaga et

al. 2003). While the role of C3a has not been directly elucidated, C5a is clearly more

important in this model since C5aR inhibition completely blocked early eosinophil

recruitment to the same degree as in C3-/- mice, which are unable to generate C3a. Hence,

during the early stages of N. brasiliensis infection, C5a is generated by activation of the

alternative complement pathway and eosinophils that are exquisitely sensitive to C5a are

rapidly and preferentially recruited to the inoculation site.

5.1.2.4 Complement-independent eosinophil recruitment

Eosinophil recruitment to the skin was complement-independent within 2.5 h of infection.

Many factors may have mediated this recruitment, including eotaxin, PAF and AMCase

(Resnick and Weller 1993; Rothenberg and Hogan 2006). Data from our laboratory suggest

that eotaxin-1-deficient/IL-5 Tg mice recruit less eosinophils than single-mutant IL-5 Tg mice

2-4 h post-N. brasiliensis infection (Knott et al, unpublished data), consistent with the time

frame where complement-independent eosinophil recruitment is apparent. Similarly,

blockade of CCR3 with mAbs impairs blood and tissue eosinophil responses in N.

CHAPTER FIVE 52 brasiliensis-infected mice (Grimaldi et al. 1999). Eotaxin is a prominent eosinophil chemo-

attractant molecule during infections with other species of helminth (Simons et al. 2005;

Dixon et al. 2006). However, as with our observations in complement-deficient/IL-5 Tg

mice, absence of eotaxin in IL-5 Tg mice does not impair resistance to N. brasiliensis (Knott

et al, unpublished data). Factor B- and eotaxin-deficient/ IL-5 Tg triple-mutant mice could be

generated to test whether further reductions in eosinophil recruitment impair resistance to this

parasite, but the genetic backgrounds of the mutant animals currently available to our

laboratory are not compatible, hence extensive backcrossing may be required. A less time-

and labour-expensive strategy would be to treat eotaxin-deficient mice with the C5aR

inhibitor PMX53 and then monitor parasite infection kinetics and inflammatory responses.

Chitinase-like proteins, such as Ym1 and AMCase have been shown to be up-regulated during

helminth infection (Nair et al. 2005; Pesce et al. 2006) and these may directly or indirectly

promote inflammatory responses such as leukocyte recruitment (Owhashi et al. 2000; Zhu et

al. 2004). Recently, intranasal chitin administration was shown to induce recruitment of IL-4-

expressing eosinophils and neutrophils into the lungs of mice and also to activate AAMs

(Reese et al. 2007). Some species of helminth express chitin synthase (Harris et al. 2000),

though this has not been described for N. brasiliensis. Since the functional importance of

parasite-derived chitin and mammalian chitinase proteins during helminth infection is yet to

be fully defined, this represents an exciting new direction for the study of immunity to

helminths. Lastly, the number of eosinophils in the skin of IL-5 Tg mice under basal

conditions may have been sufficient to cause killing of N. brasiliensis, rendering the large

degree of eosinophil recruitment unnecessary. However, co-injection of peritoneal

eosinophils with N. brasiliensis L3 does not affect kinetics of parasite migration (Daly 1999).

Since those peritoneal cells may have been in a different state of activation to those residing

in the skin of IL-5 Tg mice, this issue remains unresolved. Further experiments could be

conducted to determine if leukocytes recruited into air pouches provide protection when

CHAPTER FIVE 53 transferred with larvae into WT or eosinophil-deficient ΔdblGATA mouse recipients. This

would address whether the process of recruitment activated eosinophils to a level sufficient

for anti-parasite immunity.

5.1.2.5 Eosinophil versus neutrophil recruitment

The recruitment of eosinophils in response to N. brasiliensis infection was extremely rapid

and consistent with that observed in experimental models of nasal and pulmonary allergic

inflammation (Tedeschi et al. 1994; Tiberio et al. 2003). To our knowledge, this is the most

rapid recruitment of eosinophils into parasite-infected tissues yet reported. In some infectious

diseases, neutrophils are the leukocytes most rapidly recruited and these cells are seen in large

numbers in the first 2 h of N. brasiliensis infection of WT mice (Daly et al. 1999). We have

shown that neutrophil recruitment to the skin following N. brasiliensis infection is partially

dependent on the alternative complement pathway and C5aR, however other factors also play

a role. Potential alternative neutrophil recruitment factors include PAF, LTB4, prostaglandins

and fMLP (Jagels and Hugli 1992). The role that neutrophils play in the skin is unclear

though they do not appear to greatly restrict larval migration. WT mice exhibit only a modest

level of resistance to infection, despite an extensive influx of neutrophils into the skin soon

after injection of larvae. Neutrophils have been shown to adhere to N. brasiliensis larvae both

in vitro (MacKenzie et al. 1981) and in the current study in the skin, however there is little

evidence of significant damage to L3. Although neutrophils may be non-specific and rapidly

recruited effector cells that specialize in phagocytosis, these cells have been shown to be able

to damage some helminths (Incani and McLaren 1981; Shaio et al. 1990). In particular, the

nematode S. stercoralis is susceptible to attack by neutrophils in the skin, though in these

studies larvae were mechanically trapped in diffusion chambers for days and hence exposed to

an ongoing barrage of neutrophils (Ligas et al. 2003; Galioto et al. 2006; Kerepesi et al.

2006). This is in contrast to what happens in the N. brasiliensis model, where larvae are free

to undergo their natural route of infection, limiting the opportunity of neutrophils to interact

CHAPTER FIVE 54 with the parasite.

5.1.2.6 Complement-independent leukocyte adherence to helminths

So, if complement activation is not essential, how do eosinophils recognize and kill N.

brasiliensis in vivo? Logically, eosinophils would need to make close contact with the

surface of the parasite to inflict damage, hence adherence may be a critical step. It is possible

that the same factors that mediate the relatively minor degree of complement-independent cell

adherence in vitro (Giacomin et al. 2004), also operate in vivo. The level of the fibronectin

receptor VLA-4 on eosinophils increases during infections with some helminth species

(Brattig et al. 1995). Both fibronectin and complement deposition on N. brasiliensis L3 have

been shown to occur in vitro (Shin et al. 2001). Each of these factors was demonstrated to be

important for the adherence of eosinophils, as treatment with either anti-VLA-4 or anti-CR3

antibodies both abolished cell adherence and the larval immobilization that followed. The

fact that blocking either fibronectin receptors or complement receptors completely inhibited

adherence was curious and the authors suggested, but did not prove, that simultaneous

engagement of both receptors may be essential. The in vitro data gained from the current

study does not refute the involvement of such a mechanism, as absence of C3 or heat-

treatment of serum almost completely blocked eosinophil adherence. However, our in vivo

data does not support such an hypothesis, as cells adhered effectively in vivo in C3-deficient

mice. Other factors may be mediating cell adherence in vivo, or perhaps the involvement of

the fibronectin pathway is enhanced in complement-deficient mice as a compensatory

mechanism

Clearly, more work needs to be done to establish the role of fibronectin and other adhesion

molecules in eosinophil-dependent killing of helminths. Firstly, deposition of fibronectin on

the surface of N. brasiliensis larvae in the skin of mice could be detected by

immunofluorescence. Secondly, anti-VLA-4 antibodies could be administered locally in skin

air pouches to attempt to block cell adherence in both WT and complement-deficient mice.

CHAPTER FIVE 55 There is evidence that other adhesion molecules can contribute to eosinophil-parasite

interactions, as anti-L-selectin antibodies reduce in vitro cytotoxicity against S. mansoni

schistosomula (Nutten et al. 1999). Curiously, the authors did not observe any differences in

cell adherence to larvae, hence this ligand may have other roles in leukocyte effector cell

function, such as activation or degranulation. Other factors such as natural antibodies may

also promote eosinophil adherence to helminths. TLRs are expressed by eosinophils (Plotz et

al. 2001) and can facilitate eosinophil recognition and activation during viral infections

(Phipps et al. 2007), though very little is known regarding the role of TLRs during infections

with helminths. Alternatively, eosinophils may not need to adhere to the parasite in order to

inflict damage. Other cell types, such as platelets, have been shown to be toxic to

schistosomula without adhering, most probably via the release of toxic oxygen free radicals

on the parasite surface at a distance (Joseph et al. 1985). Exploring such complement-

independent factors was beyond the scope of the current study. Future research should further

investigate how eosinophils adhere to parasites and focus on determining if it is necessary for

the immobilisation and/or killing of these pathogens.

5.1.2.7 Eosinophil degranulation

The precise role of eosinophil degranulation in killing parasitic helminths is unclear.

Furthermore, there is also debate as to whether mouse eosinophils readily degranulate in

models of asthma-like pulmonary eosinophilic inflammation (Persson et al. 1997; Lee and

Lee 2005). The present study has demonstrated that mouse eosinophils do degranulate under

physiological conditions during helminth infections and this is consistent with previous

studies with N. brasiliensis and other parasites (Daly et al. 1999; Herbert et al. 2000; Shinkai

et al. 2002; Simons et al. 2005). Furthermore, we demonstrated that EPO levels in the skin

30 min after N. brasiliensis infection were reduced in the absence of the alternative

complement pathway. We did not establish whether this reduction in EPO activity was

because fewer eosinophils were recruited into the skin at this time, or if complement

activation is important for triggering eosinophil degranulation. Certainly, eosinophil

CHAPTER FIVE 56 degranulation was restored to normal levels at a later time point (150 min p.i.) when

eosinophil recruitment was more pronounced. It is therefore possible that the process of

recruitment is sufficient to activate eosinophils, as tissue-dwelling eosinophils generally have

a greater ability to degranulate than those in the peripheral blood (Dvorak and Ishizaka 1994).

To determine if complement activation on helminths promotes eosinophil degranulation,

purified eosinophils and N. brasiliensis L3 could be co-injected into air pouches of WT or

factor B-deficient mice. Pouches could be lavaged 30 min after co-injection and cell-free

EPO levels could then be measured in the exudates. This experiment would assist in

determining if absence of the alternative complement pathway reduces the level of

degranulation of the transferred eosinophils.

While not demonstrated in this study, it is likely that eosinophil degranulation occurred within

the aggregates of larvae and cells formed in the skin. Closer inspection of these aggregates by

electron microscopy or staining for MBP using antibodies is required to determine if these

cells do indeed degranulate. A reduction in eosinophil degranulation and larval aggregation

in the very early stages of infection may have caused the increased rate of parasite migration

to the lungs that was observed in factor B-deficient animals. In addition, the technique for

measuring cell-free EPO activity may have underestimated the level of eosinophil

degranulation, as it is possible that eosinophil granule proteins were sequestered from the

aqueous phase by binding directly to the surface of the parasite. It is unclear whether

eosinophil degranulation is a mechanism by which eosinophils kill N. brasiliensis. Future

studies should assess N. brasiliensis infection kinetics in MBP or EPO-deficient mouse

strains. These mice have been used in other models of helminth infection, where the granule

proteins were shown either to be protective (Specht et al. 2006) or had no impact on the

progress of parasite infection (Abraham et al. 2004; Ramalingam et al. 2005). Similar studies

could be conducted using MBP-deficient/IL-5 Tg or EPO-deficient/IL-5 Tg mice, which

would have large numbers of eosinophils with more limited granule contents. The present

CHAPTER FIVE 57 study has built on important observations made by our group and by others regarding the

nature of eosinophil degranulation during helminth infection. The exact stimuli for eosinophil

degranulation remains unknown and the roles of individual granule proteins are yet to be

determined.

5.1.2.8 Larval aggregation

The aggregation of larvae after injection into mouse skin was an unexpected phenomenon that

has not been described elsewhere in the literature. It is interesting that the rate at which

aggregation occurred was dependent on the alternative complement pathway. In our in vitro

assay a small degree of larval aggregation did occur (Giacomin et al. 2004), but these weakly-

formed aggregates were easily disaggregated in the washing steps. Aggregates formed in vivo

were more difficult to break apart by pipetting. However, larvae have a startling ability to

escape from aggregates within a few hours, such that relatively few larvae were recoverable

by 2.5 h p.i. Furthermore, the formation of aggregates in WT mice did not greatly impede

migration to the lungs, as lung larval burdens in these mice were only slightly lower than in

factor B-deficient mice where aggregation did not occur until later. Hence, further research is

required to determine how this parasite escapes aggregates. It is feasible that ex-sheathment

and/or the release of proteolytic factors are key to this process.

We were unable to conclusively determine the identity of the fibrous material in the

aggregates that “tethers” larvae together. It is possible that it was fibrin, which is generated

via activation of the coagulation cascade after hydrolysis of fibrinogen by thrombin (Bouma

and Mosnier 2006). Binding of fibrin or prothrombin to the surface of a parasite could not

only facilitate aggregation of larvae but could also promote cell adherence (Kuijper et al.

1997) or the generation of chemotactic factors such as C5a (Huber-Lang et al. 2006). Why

complement activation on larvae would enhance activation of the coagulation system is

unclear, however there is evidence of functional overlap between both pathways, where

complement activation can induce tissue factor, which stimulates thrombin formation (Esmon

CHAPTER FIVE 58 2004). Hence, elements of the clotting system may make significant contributions to the

immune response to N. brasiliensis and may explain the limited role for complement in

resistance of IL-5 Tg mice to infection. Future studies should investigate this by examining

fibrin deposition and thrombin activation in vivo. Inhibitors of coagulation (e.g. antithrombin

III or Hirudin) could be used in experiments designed to assess larval aggregation, cell

adherence, leukocyte recruitment and resistance to parasite infection.

5.1.3 Evasion of complement activation by helminths

N. brasiliensis has the ability to resist complement activation after a short period of time

within the host. We initially reported that lung-stage larvae recovered 24-48 h p.i. do not bind

appreciable levels of C3 or leukocytes in vivo or ex vivo (Giacomin et al. 2005), but further

analysis revealed that the ability to resist C3 deposition begins while the parasite resides in

the skin (Giacomin et al. 2008). C3 deposited on the surface of the parasite within the first 30

min was reduced or was no longer detectable after 2.5 h. This was a surprise, as C3b

deposition is covalent and the molecule is not easily shed, though it may have been degraded

by factor I to a state that was unable to be detected by our anti-C3 antibodies, for example

C3d or C3dg. It is unlikely that reductions in C3 deposition were solely due to parasite ex-

sheathment, as chemically ex-sheathed larvae still bound C3. Acquisition of host complement

inhibitory proteins such as factor H may be one mechanism through which N. brasiliensis

evades complement, as has been described for O. volvulus (Meri et al. 2002). We have

demonstrated that N. brasiliensis L3 (Baker et al. 2004) and L4 (unpublished data) bind

human factor H after incubation with NHS. Immunofluorescence and Western blotting

should be used to determine if mouse factor H is taken up by N. brasiliensis L3 or L4 in vitro

or in vivo. This analysis should focus on the time required for factor H acquisition and the

tissues in which it occurs. Unfortunately, antibodies specific for mouse factor H are not

currently available. There are other host proteins that could be adsorbed by N. brasiliensis to

evade complement, including DAF that binds to schistosomula (Ramalho-Pinto 1987) and

CHAPTER FIVE 59 C4-binding protein. Alternatively, N. brasiliensis may excrete or secrete proteins that can

either directly or indirectly inhibit complement, as has been reported for other helminth

species (Badley et al. 1987; Suchitra and Joshi 2005; Garcia-Hernandez et al. 2007).

Proteomic technology could be used to detect differences in protein expression profiles by L3

and L4 and identify potential immune evasion proteins, some of which may be novel.

Activation of the complement system is a potent stimulator of inflammatory responses in a

variety of models. The present study has demonstrated this clearly for a model of helminth

infection, but excessive or prolonged activation of complement can also be damaging for the

host. Inappropriate complement activation can contribute to the pathogenesis of

inflammatory diseases such as asthma, neurodegenerative disorders and atherosclerosis

(Oksjoki et al. 2003; van Beek et al. 2003; Sarma et al. 2006). The fact that lung-stage larvae

of N. brasiliensis can inhibit complement activation means that it may be possible to identify

parasite-derived factors that can therapeutically limit excessive complement activation in

other diseases. To achieve this, the mechanism of complement evasion must be elucidated.

5.1.4 Pulmonary cellular responses following helminth infection

5.1.4.1 Restricted early cellular inflammation in the lung

In the present study, we focused on the prominent inflammatory response induced in the skin

by injection of N. brasiliensis L3. The nature of the inflammatory response in other tissues

such as the lungs has not been fully characterised. Leukocyte infiltration of the lungs whilst

larvae are in situ is very limited (Daly 1999; Daly et al. 1999; Knott et al. 2007), even though

large numbers of cells are recruited to this site in allergic inflammatory diseases, such as

asthma. The lack of inflammation in the lungs may be a consequence of poor activation of

complement on L4 and/or a failure to generate C5a. To test this, levels of C5a in BAL fluid

following N. brasiliensis infections should be compared with levels seen in lavage fluid from

skin air pouches. Alternatively, this could be assessed by comparing the inflammatory

CHAPTER FIVE 60 response induced by injection of L3 and L4 into the skin. However, recent studies in our

laboratory suggested that L4 induce a similar cellular inflammatory response to L3 when

injected into skin air pouches (Cava 2007). Hence it is possible that the ability of L4 to

inhibit inflammation may be tissue-specific. For example, the presence of anti-inflammatory

cells in the lung may restrict cell recruitment. Such cells may be alternatively-activated

macrophages (AAMs) which are known to be present in the lungs during N. brasiliensis

infection (Reece et al. 2006). The anti-inflammatory properties of these cells could be

determined by co-injecting lung leukocytes into the skin to see if they interfere with the

normally strong cellular inflammatory response induced by injection of L3. Should anti-

inflammatory activity be detected, these cells could be fractionated and their phenotype and

secretory products determined.

5.1.4.2 Delayed cellular inflammation into the lungs

It is not widely acknowledged that the inflammatory response in the lung does not develop

until days after the parasite has left the site. A substantial inflammatory response, including

eosinophils, develops 2-4 days after N. brasiliensis leaves the lungs (Voehringer et al. 2004).

There are several unknowns regarding this delayed inflammatory response, including the

stimulus for its eventual development. It is possible that the response is initiated by parasite

antigens or sheaths shed into the lung environment as the larvae depart for the gut. It could

also be a consequence of processes promoting tissue repair, a role which both AAMs and

eosinophils may play a role (Williams 2004; Reece et al. 2006). However despite the

presence of a strong inflammatory response from days 4-6, there is no evidence that parasite

killing occurs in the lungs during either primary or secondary infections (Knott et al. 2007).

The chemotactic factors that promote cell recruitment into the lungs are also unknown. The

late inflammatory response to N. brasiliensis infection should therefore be examined in mouse

strains deficient in factors that may recruit eosinophils, such as complement factors,

chemokines such as eotaxin-1 and cytokines such as IL-4, -5 and -13.

CHAPTER FIVE 61 5.1.5 Secondary immune response to helminth infection

The present study focused primarily on innate mechanisms of immunity to N. brasiliensis,

since we proposed that complement would play its greatest role upon initial exposure to the

parasite. However, there are still many questions to be answered regarding the nature of the

strong secondary resistance to N. brasiliensis infection. Absence of IL-5 or a complete lack

of eosinophils (in ΔdblGATA mice) is associated with impaired early resistance to N.

brasiliensis infection (Knott et al. 2007). Both of these mutant strains had higher lung larval

loads than WT mice during secondary infections. In the present study we have demonstrated

that complement-deficient mice are highly resistant to secondary infection, suggesting that

early eosinophil-dependent killing of N. brasiliensis during secondary infection is mediated

by other factors. Such factors may be the same as those that promote innate eosinophil-

dependent anti-larval immunity in the pre-lung stage of primary infections, or could be

parasite-specific antibody that mediates ADCC. Based on many other models of helminth

infection, it has long been thought that parasite-specific antibody protects against secondary

infection via neutrophil or eosinophil-dependent ADCC (MacKenzie et al. 1980; Incani and

McLaren 1981; Shaio et al. 1990; Venturiello et al. 1995). However, this study suggests that

the presence of parasite-specific antibody during secondary N. brasiliensis infection does not

promote classical pathway-dependent parasite killing, since C1q-deficient and C3-deficient

mice exhibit similar parasite burdens to WT animals in the lungs and small intestine. Hence,

complement is not required for anti-parasite resistance during secondary N. brasiliensis

infection.

The companion study of Knott et al (2007) indicates that eosinophils are also not essential for

expulsion of the adult intestinal stage of N. brasiliensis during secondary infections. Similar

results are seen during infections with other helminths, including B. malayi (Ramalingam et

al. 2003; Simons et al. 2005) and S. ratti (Ovington et al. 1998), suggesting that while

eosinophils may be protective in the early stages of secondary infections with some species of

CHAPTER FIVE 62 helminth, other elements of the immune response are sufficient to cause elimination of the

parasite from the host. It is likely that such mechanisms are dependent on STAT6 and IL-

4/IL-13, which may invoke physiological changes in the gastrointestinal environment that act

to “flush” or “force” helminths from the intestine (Mettrick et al. 1979; Nawa et al. 1994;

Bancroft et al. 1997; Urban et al. 1998; Cliffe et al. 2005).

5.1.6 Future directions for studies using complement-deficient/IL-5 Tg mice

We have investigated, using a unique mouse model, the relationship between IL-5,

eosinophils and the complement system during infection with a parasitic helminth. The

complement system plays an important role in regulating eosinophil trafficking and activation

during eosinophilic conditions. Hence, it may also play a role in other eosinophilic disease

models. The tools used and the data generated in this study could be applied to other disease

models, including infections with other parasite species. IL-5 Tg mice have been shown to be

resistant to infection with S. stercoralis, L. sigmodontis, A. cantonenesis and A. costaricensis

(Sugaya et al. 1997; Herbert et al. 2000; Martin et al. 2000; Sugaya et al. 2002). The

complement-deficient/IL-5 Tg mouse strains we developed as part of this study could be used

as hosts for these and other parasite species to establish if complement is more important for

eosinophil-dependent anti-parasite resistance. While the use of IL-5 Tg mice is essentially an

artificial system, the resultant eosinophilia is characteristic of a number of diseases. The role

of complement during eosinophilia could also be assessed in murine asthma models to

determine if the absence of specific pathways of complement activation lessens symptoms.

Similar approaches could be used in animal models of allergen-induced eosinophilic

oesophagitis, rhinitis, and gastroenteritis.

5.1.7 Issues for design of anthelmintic vaccines

Ultimately, the major goal for those studying immunity to helminths is the development of

vaccines to protect humans and domesticated animals from infection. The life cycles and

CHAPTER FIVE 63 pathology caused by the human pathogens S. stercoralis and the hookworms Necator

americanus and Ancylostoma duodenale are similar to the tissue-invasive stages of N.

brasiliensis in mice, albeit much longer in duration. Developing a vaccine that promotes

rapid eosinophil recruitment to sites of infection and limits the ability of the parasite to evade

complement may be a useful strategy. Such a vaccine would mirror what might occur in

natural and acquired resistance, restricting parasite migration and hence breaking the lifecycle

at an early stage within the host. Gastrointestinal helminths may become more difficult to

destroy as they mature and migrate through the host to the gut. Therefore, using the early

stages of parasite maturation as vaccine targets may be the most useful strategy, as it would

prevent much of the pathology caused by the helminth and stop parasite reproduction and

spreading of the disease.

Many studies using humans or animal subjects have investigated potential vaccine candidates,

including the use of attenuated larvae, native parasite ES proteins and individual recombinant

ES proteins (Schallig et al. 1997; Lightowlers et al. 2003; Vercauteren et al. 2004; Hewitson

et al. 2005). The present study has not conclusively demonstrated a mechanism by which

eosinophils might provide resistance to N. brasiliensis, but the process appears to be

dependent on multiple factors. Complement activation can rapidly trigger several important

immune responses, including eosinophil and neutrophil recruitment and it may also trigger

eosinophil degranulation. Evasion mechanisms employed by parasites mean that the

influence of complement is limited over time. Discovering the mechanisms used by

helminths to eventually avoid complement activation may identify new target molecules for

vaccination.

5.2 CONCLUSION

The present study has comprehensively examined the role of complement in immunity to the

nematode N. brasiliensis. Previously, most studies of the immune response to gastrointestinal

CHAPTER FIVE 64 helminth infections have focused on responses in the gut that promote parasite expulsion.

Our work is relatively unique in that we have concentrated on earlier events and especially

those that occur in the skin within the first few hours of an infection. Understanding how

parasites are killed at this early stage may be critical for developing strategies, including

vaccines, which will limit the progression an infection. Critically, eosinophils can be

important for anti-parasite resistance if available in sufficient numbers in the early stages of

infection (Daly et al. 1999; Dent et al. 1999). A crucial next step will be to determine if

similar protection is possible with some of the major nematode parasites of humans. In the

current study we have found that the infective stage of this parasite is exquisitely sensitive to

the alternative pathway of complement activation and to eosinophil attachment in vitro. We

have demonstrated that complement contributes to a number of important immune responses

in the first 30 min of sub-cutaneous injection. However the complement system becomes

largely redundant after just 2.5 h, most likely due to evasion strategies instituted by the

parasite and other factors within the immune system that can contribute to anti-parasite

immunity. The parasite can hence migrate through to the lungs and gut largely unaffected by

complement, improving its chances of maturation and reproduction. Hence, while the

interaction between eosinophils, complement and N. brasiliensis appears to be fairly straight-

forward in vitro, the in vivo setting is far more complex. This highlights that the mammalian

immune system is a complicated entity, where functional overlap between different elements

can lead to apparent redundancy of some components. Since parasites usually need to live in

close contact with the host’s immune system, in some cases for years, they have evolved

sophisticated strategies to limit potentially damaging arms of the immune response and

inhibition of the complement system may be a critical first stage in this.

REFERENCES 65Abe, T. and Y. Nawa (1987). Reconstitution of mucosal mast cells in W/Wv mice by adoptive transfer of bone marrow-derived cultured mast cells and its effect on the protective capacity to Strongyloides ratti infection. Parasite Immunol. 9: 31-38. Abraham, D., R. B. Grieve and M. Mika-Grieve (1988). Dirofilaria immitis: surface properties of third- and fourth-stage larvae. Exp Parasitol. 65: 157-167. Abraham, D., O. Leon, S. Schnyder-Candrian, C. C. Wang, A. M. Galioto, L. A. Kerepesi, J. J. Lee and S. Lustigman (2004). Immunoglobulin E and eosinophil-dependent protective immunity to larval Onchocerca volvulus in mice immunized with irradiated larvae. Infect Immun. 72: 810-817. Abu-Ghazaleh, R. I., H. Kita and G. J. Gleich (1992). Eosinophil activation and function in health and disease. Immunol Ser. 57: 137-167. Aggarwal, S., N. Ghilardi, M. H. Xie, F. J. de Sauvage and A. L. Gurney (2003). Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J Biol Chem. 278: 1910-1914. Ahlstedt, S. (1981). Experimental Escherichia coli 06 infection in mice. IV. On the relative importance of complement factors and antibodies for the host defence. Acta Pathol Microbiol Scand [C]. 89: 23-28. Ambrus, G., P. Gal, M. Kojima, K. Szilagyi, J. Balczer, J. Antal, L. Graf, A. Laich, B. E. Moffatt, W. Schwaeble, R. B. Sim and P. Zavodszky (2003). Natural substrates and inhibitors of mannan-binding lectin-associated serine protease-1 and -2: a study on recombinant catalytic fragments. J Immunol. 170: 1374-1382. Applequist, S. E., R. P. Wallin and H. G. Ljunggren (2002). Variable expression of Toll-like receptor in murine innate and adaptive immune cell lines. Int Immunol. 14: 1065-1074. Arizono, N., M. Nishida, R. Uchikawa, M. Yamada, S. Matsudea, T. Tegoshi, Y. Kitamura and M. Sasabe (1996). Lung granulomatous response induced by infection with the intestinal nematode Nippostrongylus brasiliensis is suppressed in mast cell-deficient Ws/Ws rats. Clin. Exp. Immunol. 160: 55-61. Badley, J. E., R. B. Grieve, J. H. Rockey and L. T. Glickman (1987). Immune-mediated adherence of eosinophils to Toxocara canis infective larvae: the role of excretory-secretory antigens. Parasite Immunol. 9: 133-143. Baeza, E., I. Poitou, C. Villejoubert and C. Boulard (1994). Complement depletion in rats infected with Fasciola hepatica: in vivo and in vitro studies. Vet Parasitol. 51: 219-230. Baker, S., H. Wang, P. R. Giacomin, L. A. Dent and D. L. Gordon (2004). Binding of complement factor H to the nematode Nippostrongylus brasiliensis. Mol Immunol. 41: 205-206. Meeting Abstract. Balic, A., Y. Harcus, M. J. Holland and R. M. Maizels (2004). Selective maturation of dendritic cells by Nippostrongylus brasiliensis-secreted proteins drives Th2 immune responses. Eur J Immunol. 34: 3047-3059. Bancroft, A. J., K. J. Else, J. P. Sypek and R. K. Grencis (1997). Interleukin-12 promotes a chronic intestinal nematode infection. Eur J Immunol. 27: 866-870.

REFERENCES 66Bartholomew, R. M. and A. F. Esser (1980). Mechanism of antibody-independent activation of the first component of complement (Cl) on retrovirus membranes. Biochemistry. 19: 2847-2853. Bassett, E. G., J. R. Baker and P. DeSouza (1977). A light microscopical study of healing incised dermal wounds in rats, with special reference to eosinophilic leukocytes and to the collagenous fibres of the periwound areas. Br. J. Exp. Pathol. 58: 581-605. Befus, A. D. (1977). Hymenolepis diminuta and H. microstoma: mouse immunoglobulins binding to the tegumental surface. Exp Parasitol. 41: 242-251. Behm, C. A. and K. S. Ovington (2000). The role of eosinophils in parasitic helminth infections: insights from genetically modified mice. Parasitol Today. 16: 202-209. Belkaid, Y. and B. T. Rouse (2005). Natural regulatory T cells in infectious disease. Nat Immunol. 6: 353-360. Bentley, A. G., A. S. Carlisle and S. M. Phillips (1981). Ultrastructural analysis of the cellular response to Schistosoma mansoni. II. Inflammatory response in rodent skin. Am J Trop Med Hyg. 30: 815-824. Besier, R. B. and S. C. J. Love (2003). Anthelminthic resistance in sheep nematodes in Australia: the need for new approaches. Aust J Exp Agric. 43: 1383-1391. Betts, C. J. and K. J. Else (1999). Mast cells, eosinophils and antibody-mediated cellular cytotoxicity are not critical in resistance to Trichuris muris. Parasite Immunol. 21: 45-52. Bottjer, K. P. and L. W. Bone (1985). Nippostrongylus brasiliensis: feeding activity in the mouse. Int J Parasitol. 15: 9-14. Botto, M., C. Dell'Agnola, A. E. Bygrave, E. M. Thompson, H. T. Cook, F. Petry, M. Loos, P. P. Pandolfi and M. J. Walport (1998). Homozygous C1q deficiency causes glomerulonephritis associated with multiple apoptotic bodies. Nat Genet. 19: 56-59. Bouma, B. N. and L. O. Mosnier (2006). Thrombin activatable fibrinolysis inhibitor (TAFI)--how does thrombin regulate fibrinolysis? Ann Med. 38: 378-388. Brambell, M. R. (1965). The distribution of a primary infestation of Nippostrongylus brasiliensis in the small intestine of laboratory rats. Parasitol. 55: 313-324. Brattig, N. W., A. Z. Abakar, F. Geisinger and T. F. Kruppa (1995). Cell-adhesion molecules expressed by activated eosinophils in Onchocerca volvulus infection. Parasitol Res. 81: 398-402. Brattig, N. W., C. Bazzocchi, C. J. Kirschning, N. Reiling, D. W. Buttner, F. Ceciliani, F. Geisinger, H. Hochrein, M. Ernst, H. Wagner, C. Bandi and A. Hoerauf (2004). The major surface protein of Wolbachia endosymbionts in filarial nematodes elicits immune responses through TLR2 and TLR4. J Immunol. 173: 437-445. Brattig, N. W., F. W. Tischendorf, G. Strote and C. E. Medina-de la Garza (1991). Eosinophil-larval-interaction in onchocerciasis: heterogeneity of in vitro adherence of eosinophils to infective third and fourth stage larvae and microfilariae of Onchocerca volvulus. Parasite Immunol. 13: 13-22.

REFERENCES 67Brigandi, R. A., H. L. Rotman, W. Yutanawiboonchai, O. Leon, T. J. Nolan, G. A. Schad and D. Abraham (1996). Strongyloides stercoralis: role of antibody and complement in immunity to the third stage of larvae in BALB/cByJ mice. Exp Parasitol. 82: 279-289. Brown, E. J., K. A. Joiner, T. A. Gaither, C. H. Hammer and M. M. Frank (1983). The interaction of C3b bound to pneumococci with factor H (beta 1H globulin), factor I (C3b/C4b inactivator), and properdin factor B of the human complement system. J Immunol. 131: 409-415. Butterworth, A. E. (1984). Cell-mediated damage to helminths. Adv Parasitol. 23: 143-235. Butterworth, A. E., R. F. Sturrock, V. Houba, A. A. Mahmoud, A. Sher and P. H. Rees (1975). Eosinophils as mediators of antibody-dependent damage to schistosomula. Nature. 256: 727-729. Butterworth, A. E., D. L. Wassom, G. J. Gleich, D. A. Loegering and J. R. David (1979). Damage to schistosomula of Schistosoma mansoni induced directly by eosinophil major basic protein. J Immunol. 122: 221-229. Buys, J., R. Wever and E. J. Ruitenberg (1984). Myeloperoxidase is more efficient than eosinophil peroxidase in the in vitro killing of newborn larvae of Trichinella spiralis. Immunology. 51: 601-607. Buys, J., R. Wever, R. van Stigt and E. J. Ruitenberg (1981). The killing of newborn larvae of Trichinella spiralis by eosinophil peroxidase in vitro. Eur J Immunol. 11: 843-845. Carroll, M. C. (2004). The complement system in B cell regulation. Mol Immunol. 41: 141-146. Carroll, M. C. (2004). The complement system in regulation of adaptive immunity. Nat Immunol. 5: 981-986. Cava, M. (2007). Parasite evasion of host immune responses: Immunomodulatory properties of parasite proteins derived from Necator americanus, Ancylostoma caninum and Toxocara canis. Honours Thesis. School of Molecular and Biomedical Science. Adelaide, University of Adelaide. Chan, R. K., S. I. Ibrahim, K. Takahashi, E. Kwon, M. McCormack, A. Ezekowitz, M. C. Carroll, F. D. Moore, Jr. and W. G. Austen, Jr. (2006). The differing roles of the classical and mannose-binding lectin complement pathways in the events following skeletal muscle ischemia-reperfusion. J Immunol. 177: 8080-8085. Chandrashekar, R., U. R. Rao and D. Subrahmanyam (1990). Antibody-mediated cytotoxic effects in vitro and in vivo of rat cells on infective larvae of Brugia malayi. Int J Parasitol. 20: 725-730. Chen, Y., C. L. Langrish, B. McKenzie, B. Joyce-Shaikh, J. S. Stumhofer, T. McClanahan, W. Blumenschein, T. Churakovsa, J. Low, L. Presta, C. A. Hunter, R. A. Kastelein and D. J. Cua (2006). Anti-IL-23 therapy inhibits multiple inflammatory pathways and ameliorates autoimmune encephalomyelitis. J Clin Invest. 116: 1317-1326. Clark, K., L. Simson, N. Newcombe, A. M. Koskinen, J. Mattes, N. A. Lee, J. J. Lee, L. A. Dent, K. I. Matthaei and P. S. Foster (2004). Eosinophil degranulation in the allergic lung of mice primarily occurs in the airway lumen. J Leukoc Biol. 75: 1001-1009.

REFERENCES 68 Clarke, K. R. (1967). The migration route of the third stage larvae of Nippostrongylus brasiliensis (Travassos, 1914). J Helminthol. 41: 285-290. Cliffe, L. J., N. E. Humphreys, T. E. Lane, C. S. Potten, C. Booth and R. K. Grencis (2005). Accelerated intestinal epithelial cell turnover: a new mechanism of parasite expulsion. Science. 308: 1463-1465. Coffman, R. L., B. W. Seymour, S. Hudak, J. Jackson and D. Rennick (1989). Antibody to interleukin-5 inhibits helminth-induced eosinophilia in mice. Science. 245: 308-310. Cooper, N. R., B. M. Bradt, J. S. Rhim and G. R. Nemerow (1990). CR2 complement receptor. J Invest Dermatol. 94: 112S-117S. Cormier, S. A., K. A. Larson, S. Yuan, T. L. Mitchell, K. Lindenberger, P. Carrigan, N. A. Lee and J. J. Lee (2001). Mouse eosinophil-associated ribonucleases: a unique subfamily expressed during hematopoiesis. Mamm Genome. 12: 352-361. Corry, D. B., H. G. Folkesson, M. L. Warnock, D. J. Erle, M. A. Matthay, J. P. Wiener-Kronish and R. M. Locksley (1996). Interleukin 4, but not interleukin 5 or eosinophils, is required in a murine model of acute airway hyperreactivity. J Exp Med. 183: 109-117. Crompton, D. W. (1999). How much human helminthiasis is there in the world? J Parasitol. 85: 397-403. Daly, C. M. (1999). Immune responses to Nippostrongylus brasiliensis in Interleukin-5 Transgenic Mice. Ph.D. thesis. Microbiology and Immunology. Adelaide, University of Adelaide. Daly, C. M., B. Li, J. Forbes, P. R. Giacomin and L. A. Dent (2004). Eosinophils as mediators of early resistance in mice to the nematode Nippostrongylus brasiliensis. Clin. Investigative Med. 27: 99D. Meeting abstract. Daly, C. M., G. Mayrhofer and L. A. Dent (1999). Trapping and immobilization of Nippostrongylus brasiliensis larvae at the site of inoculation in primary infections of interleukin-5 transgenic mice. Infect Immun. 67: 5315-5323. Datta, R., M. L. deSchoolmeester, C. Hedeler, N. W. Paton, A. M. Brass and K. J. Else (2005). Identification of novel genes in intestinal tissue that are regulated after infection with an intestinal nematode parasite. Infect Immun. 73: 4025-4033. David, J. R., A. E. Butterworth and M. A. Vadas (1980). Mechanism of the interaction mediating killing of Schistosoma mansoni by human eosinophils. Am J Trop Med Hyg. 29: 842-848. Davis, S. W. and B. Hammerberg (1988). Activation of the alternative pathway of complement by larval Taenia taeniaeformis in resistant and susceptible strains of mice. Int J Parasitol. 18: 591-597. Davis, S. W. and B. Hammerberg (1990). Taenia taeniaeformis: evasion of complement-mediated lysis by early larval stages following activation of the alternative pathway. Int J Parasitol. 20: 587-593.

REFERENCES 69de Veer, M. J., J. M. Kemp and E. N. Meeusen (2007). The innate host defence against nematode parasites. Parasite Immunol. 29: 1-9. Deng, J., D. Gold, P. T. LoVerde and Z. Fishelson (2003). Inhibition of the complement membrane attack complex by Schistosoma mansoni paramyosin. Infect Immun. 71: 6402-6410. Dent, L. A., C. Daly, A. Geddes, J. Cormie, D. A. Finlay, L. Bignold, P. Hagan, R. M. Parkhouse, T. Garate, J. Parsons and G. Mayrhofer (1997a). Immune responses of IL-5 transgenic mice to parasites and aeroallergens. Mem Inst Oswaldo Cruz. 92: 45-54. Dent, L. A., C. M. Daly, G. Mayrhofer, T. Zimmerman, A. Hallett, L. P. Bignold, J. Creaney and J. C. Parsons (1999). Interleukin-5 transgenic mice show enhanced resistance to primary infections with Nippostrongylus brasiliensis but not primary infections with Toxocara canis. Infect Immun. 67: 989-993. Dent, L. A., G. H. Munro, K. P. Piper, C. J. Sanderson, D. A. Finlay, R. K. Dempster, L. P. Bignold, D. G. Harkin and P. Hagan (1997b). Eosinophilic interleukin 5 (IL-5) transgenic mice: eosinophil activity and impaired clearance of Schistosoma mansoni. Parasite Immunol. 19: 291-300. Dent, L. A., M. Strath, A. L. Mellor and C. J. Sanderson (1990). Eosinophilia in transgenic mice expressing interleukin 5. J Exp Med. 172: 1425-1431. Desakorn, V., P. Suntharasamai, S. Pukrittayakamee, S. Migasena and D. Bunnag (1987). Adherence of human eosinophils to infective filariform larvae of Necator americanus in vitro. Southeast Asian J Trop Med Public Health. 18: 66-72. Diaz, A., A. M. Ferreira and A. Nieto (1995). Echinococcus granulosus: interactions with host complement in secondary infection in mice. Exp Parasitol. 80: 473-482. Dixon, H., C. Blanchard, M. L. Deschoolmeester, N. C. Yuill, J. W. Christie, M. E. Rothenberg and K. J. Else (2006). The role of Th2 cytokines, chemokines and parasite products in eosinophil recruitment to the gastrointestinal mucosa during helminth infection. Eur J Immunol. 36: 1753-1763. Donaldson, L. E., E. Scmitt, J. F. Huntley, G. F. Newlands and R. K. Grencis (1996). A critical role for stem cell factor and c-kit in host protective immunity to an intestinal helminth. Int. Immunol. 8: 559-567. Dvorak, A. M., S. J. Ackerman, T. Furitsu, P. Estrella, L. Letourneau and T. Ishizaka (1992). Mature eosinophils stimulated to develop in human-cord blood mononuclear cell cultures supplemented with recombinant human interleukin-5. II. Vesicular transport of specific granule matrix peroxidase, a mechanism for effecting piecemeal degranulation. Am J Pathol. 140: 795-807. Dvorak, A. M., T. Furitsu, P. Estrella, L. Letourneau, T. Ishizaka and S. J. Ackerman (1994). Ultrastructural localization of major basic protein in the human eosinophil lineage in vitro. J Histochem Cytochem. 42: 1443-1451. Dvorak, A. M. and T. Ishizaka (1994). Human eosinophils in vitro. An ultrastructural morphology primer. Histol Histopathol. 9: 339-374.

REFERENCES 70Eads, M. E., N. J. Levy, D. L. Kasper, C. J. Baker and A. Nicholson-Weller (1982). Antibody-independent activation of C1 by type Ia group B streptococci. J Infect Dis. 146: 665-672. Egesten, A., J. Calafat, P. F. Weller, E. F. Knol, H. Janssen, T. M. Walz and I. Olsson (1997). Localization of granule proteins in human eosinophil bone marrow progenitors. Int Arch Allergy Immunol. 114: 130-138. Egesten, A. and J. Malm (1998). Eosinophil degranulation in response to serum-opsonised beads: C5a and PAF-activating factors enhance ECp release, with roles for protein kinase A and C. Eur. J. Allergy Clin. Immunol. 53: 1066-1073. Egwang, T. G., A. D. Befus and J. Gauldie (1985). Activation of alveolar macrophages following infection with the parasitic nematode Nippostrongylus brasiliensis. Immunol. 54: 581. Egwang, T. G., J. Gauldie and D. Befus (1984). Complement-dependent killing of Nippostrongylus brasiliensis infective larvae by rat alveolar macrophages. Clin. Exp. Immunol. 55: 149-156. Ehlers, M. R. (2000). CR3: a general purpose adhesion-recognition receptor essential for innate immunity. Microbes Infect. 2: 289-294. Else, K. J., F. D. Finkelman, C. R. Maliszewski and R. K. Grencis (1994). Cytokine-mediated regulation of chronic intestinal helminth infection. J Exp Med. 179: 347-351. Endo, Y., M. Takahashi and T. Fujita (2006). Lectin complement system and pattern recognition. Immunobiology. 211: 283-293. Erjefalt, J. S., M. Andersson, L. Greiff, M. Korsgren, M. Gizycki, P. K. Jeffery and G. A. Persson (1998). Cytolysis and piecemeal degranulation as distinct modes of activation of airway mucosal eosinophils. J Allergy Clin Immunol. 102: 286-294. Esmon, C. T. (2004). The impact of the inflammatory response on coagulation. Thromb Res. 114: 321-327. Falcone, F. H., D. I. Pritchard and B. F. Gibbs (2001). Do basophils play a role in immunity against parasites? Trends Parasitol. 17: 126-129. Fallon, P. G., S. J. Ballantyne, N. E. Mangan, J. L. Barlow, A. Dasvarma, D. R. Hewett, A. McIlgorm, H. E. Jolin and A. N. McKenzie (2006). Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion. J Exp Med. 203: 1105-1116. Ferreira, A. M., M. Breijo, R. B. Sim and A. Nieto (2000). Contribution of C5-mediated mechanisms to host defence against Echinococcus granulosus hydatid infection. Parasite Immunol. 22: 445-453. Finkelman, F. D., I. M. Katona, J. F. Urban, Jr., J. Holmes, J. Ohara, A. S. Tung, J. V. Sample and W. E. Paul (1988). IL-4 is required to generate and sustain in vivo IgE responses. J Immunol. 141: 2335-2341. Finkelman, F. D., K. B. Madden, A. W. Cheever, I. M. Katona, S. C. Morris, M. K. Gately, B. R. Hubbard, W. C. Gause and J. F. Urban, Jr. (1994). Effects of interleukin 12 on immune

REFERENCES 71responses and host protection in mice infected with intestinal nematode parasites. J Exp Med. 179: 1563-1572. Finkelman, F. D., T. Shea-Donohue, J. Goldhill, C. A. Sullivan, S. C. Morris, K. B. Madden, W. C. Gause and J. F. Urban, Jr. (1997). Cytokine regulation of host defense against parasitic gastrointestinal nematodes: lessons from studies with rodent models. Annu Rev Immunol. 15: 505-533. Fischer, E., M. Capron, L. Prin, J. P. Kusnierz and M. D. Kazatchkine (1986). Human eosinophils express CR1 and CR3 complement receptors for cleavage fragments of C3. Cell Immunol. 97: 297-306. Fischer, M. B., M. Ma, S. Goerg, X. Zhou, J. Xia, O. Finco, S. Han, G. Kelsoe, R. G. Howard, T. L. Rothstein, E. Kremmer, F. S. Rosen and M. C. Carroll (1996). Regulation of the B cell response to T-dependent antigens by classical pathway complement. J Immunol. 157: 549-556. Flood-Page, P., A. Menzies-Gow, S. Phipps, S. Ying, A. Wangoo, M. S. Ludwig, N. Barnes, D. Robinson and A. B. Kay (2003). Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 112: 1029-1036. Folkard, S. G., P. J. Hogarth, M. J. Taylor and A. E. Bianco (1996). Eosinophils are the major effector cells of immunity to microfilariae in a mouse model of onchocerciasis. Parasitology. 112 ( Pt 3): 323-329. Foster, P. S., S. P. Hogan, A. J. Ramsay, K. I. Matthaei and I. G. Young (1996). Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J. Exp. Med. 183: 195-201. Foster, P. S., S. P. Hogan, A. J. Ramsay, K. I. Matthaei and I. G. Young (1996). Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J Exp Med. 183: 195-201. Fujisawa, T., R. Abu-Ghazaleh, H. Kita, C. J. Sanderson and G. J. Gleich (1990). Regulatory effect of cytokines on eosinophil degranulation. J Immunol. 144: 642-646. Galioto, A. M., J. A. Hess, T. J. Nolan, G. A. Schad, J. J. Lee and D. Abraham (2006). Role of eosinophils and neutrophils in innate and adaptive protective immunity to larval Strongyloides stercoralis in mice. Infect Immun. 74: 5730-5738. Garcia-Hernandez, P., M. Rodero and C. Cuellar (2007). Anisakis simplex: the activity of larval products on the complement system. Exp Parasitol. 115: 1-8. Garrett, J. K., S. C. Jameson, B. Thomson, M. H. Collins, L. E. Wagoner, D. K. Freese, L. A. Beck, J. A. Boyce, A. H. Filipovich, J. M. Villanueva, S. A. Sutton, A. H. Assa'ad and M. E. Rothenberg (2004). Anti-interleukin-5 (mepolizumab) therapy for hypereosinophilic syndromes. J Allergy Clin Immunol. 113: 115-119. Gasque, P. (2004). Complement: a unique innate immune sensor for danger signals. Mol Immunol. 41: 1089-1098. Gelfand, J. A., D. L. Hurley, A. S. Fauci and M. M. Frank (1978). Role of complement in host defense against experimental disseminated candidiasis. J Infect Dis. 138: 9-16.

REFERENCES 72 Gewurz, H., S. C. Ying, H. Jiang and T. F. Lint (1993). Nonimmune activation of the classical complement pathway. Behring Inst Mitt. 138-147. Giacomin, P. R., D. L. Gordon, M. Botto, M. R. Daha, S. D. Sanderson, S. M. Taylor and L. A. Dent (2008). The role of complement in innate, adaptive and eosinophil-dependent immunity to the nematode Nippostrongylus brasiliensis. Mol Immunol. 45: 446-455. Giacomin, P. R., H. Wang, D. L. Gordon, M. Botto and L. A. Dent (2005). Loss of complement activation and leukocyte adherence as Nippostrongylus brasiliensis develops within the murine host. Infect Immun. 73: 7442-7449. Giacomin, P. R., H. Wang, D. L. Gordon and L. A. Dent (2004). Quantitation of complement and leukocyte binding to a parasitic helminth species. J Immunol Methods. 289: 201-210. Gianella-Borradori, A., L. Borradori, P. M. Schneider, E. Gautier and P. J. Spath (1990). Combined complete C5 and partial C4 deficiency in humans: clinical consequences and complement-mediated functions in vitro. Clin Immunol Immunopathol. 55: 41-55. Gleich, G. J., E. Frias, D. A. Loegering, D. L. Wassom and D. Steinmuller (1979). Cytotoxic properties of the eosinophil major basic protein. J. Immunol. 123: 2925-2927. Gleich, G. J., D. A. Loegering, K. G. Mann and J. E. Maldonado (1976). Comparative properties of the Charcot-Leyden crystal protein and the major basic protein from human eosinophils. J Clin Invest. 57: 633-640. Gouon-Evans, V., M. E. Rothenberg and J. W. Pollard (2000). Postnatal mammary gland development requires macrophages and eosinophils. Development. 127: 2269-2282. Grencis, R. K. (1997). Th2-mediated host protective immunity to intestinal nematode infections. Philos Trans R Soc Lond B Biol Sci. 352: 1377-1384. Grimaldi, J. C., N. X. Yu, G. Grunig, B. W. Seymour, F. Cottrez, D. S. Robinson, N. Hosken, W. G. Ferlin, X. Wu, H. Soto, A. O'Garra, M. C. Howard and R. L. Coffman (1999). Depletion of eosinophils in mice through the use of antibodies specific for C-C chemokine receptor 3 (CCR3). J Leukoc Biol. 65: 846-853. Gruden-Movsesijan, A., M. Petrovic and L. Sofronic-Milosavljevic (2003). Interaction of mannan-binding lectin with Trichinella spiralis glycoproteins, a possible innate immune mechanism. Parasite Immunol. 25: 545-552. Guenther, L. C. (1983). Inherited disorders of complement. J Am Acad Dermatol. 9: 815-839. Gupta, R., P. Bajpai, L. M. Tripathi, V. M. Srivastava, S. K. Jain and S. Misra-Bhattacharya (2004). Macrophages in the development of protective immunity against experimental Brugia malayi infection. Parasitology. 129: 311-323. Hagan, P., H. A. Wilkins, U. J. Blumenthal, R. J. Hayes and B. M. Greenwood (1985). Eosinophilia and resistance to Schistosoma haematobium in man. Parasite Immunol. 7: 625-632. Hamann, K. J., R. L. Barker, R. M. Ten and G. J. Gleich (1991). The molecular biology of eosinophil granule proteins. Int Arch Allergy Appl Immunol. 94: 202-209.

REFERENCES 73Hamann, K. J., G. J. Gleich, J. L. Checkel, D. A. Loegering, J. W. McCall and R. L. Barker (1990). In vitro killing of microfilariae of Brugia pahangi and Brugia malayi by eosinophil granule proteins. J Immunol. 144: 3166-3173. Happel, K. I., P. J. Dubin, M. Zheng, N. Ghilardi, C. Lockhart, L. J. Quinton, A. R. Odden, J. E. Shellito, G. J. Bagby, S. Nelson and J. K. Kolls (2005). Divergent roles of IL-23 and IL-12 in host defense against Klebsiella pneumoniae. J Exp Med. 202: 761-769. Harrington, L. E., P. R. Mangan and C. T. Weaver (2006). Expanding the effector CD4 T-cell repertoire: the Th17 lineage. Curr Opin Immunol. 18: 349-356. Harris, M. T., K. Lai, K. Arnold, H. F. Martinez, C. A. Specht and J. A. Fuhrman (2000). Chitin synthase in the filarial parasite, Brugia malayi. Mol Biochem Parasitol. 111: 351-362. Heath, D. D., B. Holcman and R. J. Shaw (1994). Echinococcus granulosus: the mechanism of oncosphere lysis by sheep complement and antibody. Int J Parasitol. 24: 929-935. Hellmich, B., E. Csernok and W. L. Gross (2005). Proinflammatory cytokines and autoimmunity in Churg-Strauss syndrome. Ann N Y Acad Sci. 1051: 121-131. Henderson, W. R. and E. Y. Chi (1985). Ultrastructural characterization and morphometric analysis of human eosinophil degranulation. J Cell Sci. 73: 33-48. Herbert, D. R., J. J. Lee, N. A. Lee, T. J. Nolan, G. A. Schad and D. Abraham (2000). Role of IL-5 in innate and adaptive immunity to larval Strongyloides stercoralis in mice. J Immunol. 165: 4544-4551. Herndon, F. J. and S. G. Kayes (1992). Depletion of eosinophils by anti-IL-5 monoclonal antibody treatment of mice infected with Trichinella spiralis does not alter parasite burden or immunologic resistance to reinfection. J Immunol. 149: 3642-3647. Hewitson, J. P., P. A. Hamblin and A. P. Mountford (2005). Immunity induced by the radiation-attenuated schistosome vaccine. Parasite Immunol. 27: 271-280. Hokibara, S., M. Takamoto, A. Tominaga, K. Takatsu and K. Sugane (1997). Marked eosinophilia in interleukin-5 transgenic mice fails to prevent Trichinella spiralis infection. J Parasitol. 83: 1186-1189. Holland, M. J., Y. M. Harcus, P. L. Riches and R. M. Maizels (2000). Proteins secreted by the parasitic nematode Nippostrongylus brasiliensis act as adjuvants for Th2 responses. Eur J Immunol. 30: 1977-1987. Hong, Y., C. W. Kim and B. Ghebrehiwet (1992). Trichinella spiralis: activation of complement by infective larvae, adults, and newborn larvae. Exp Parasitol. 74: 290-299. Hsu, S. Y. L., H. F. Hsu, G. E. Penicle, G. L. Lust and J. W. Osborne (1974). Dermal hypersensitivity to schistosome cercariae in rhesus monkeys during immunisation and challenge. I. Complex hypersensitivity reactions of a well immunised monkey during challenge. J. Allergy. Clin. Immunol. 54: 339. Huber-Lang, M., J. V. Sarma, F. S. Zetoune, D. Rittirsch, T. A. Neff, S. R. McGuire, J. D. Lambris, R. L. Warner, M. A. Flierl, L. M. Hoesel, F. Gebhard, J. G. Younger, S. M. Drouin, R. A. Wetsel and P. A. Ward (2006). Generation of C5a in the absence of C3: a new complement activation pathway. Nat Med. 12: 682-687.

REFERENCES 74 Huwer, M., S. Sanft and J. S. Ahmed (1989). Enhancement of neutrophil adherence to Toxocara canis larvae by the C3 component of complement and IgG antibodies. Zentralbl Bakteriol Mikrobiol Hyg [A]. 270: 418-423. Incani, R. N. and D. J. McLaren (1981). Neutrophil-mediated cytotoxicity to schistosomula of Schistosoma mansoni in vitro: studies on the kinetics of complement and/or antibody-dependent adherence and killing. Parasite Immunol. 3: 107-126. Irigoin, F., R. Wurzner, R. B. Sim and A. M. Ferreira (1996). Comparison of complement activation in vitro by different Echinococcus granulosus extracts. Parasite Immunol. 18: 371-375. Jagels, M. A. and T. E. Hugli (1992). Neutrophil chemotactic factors promote leukocytosis. A common mechanism for cellular recruitment from bone marrow. J Immunol. 148: 1119-1128. James, S. L., A. W. Cheever, P. Caspar and T. A. Wynn (1998). Inducible nitric oxide synthase-deficient mice develop enhanced type 1 cytokine-associated cellular and humoral immune responses after vaccination with attenuated Schistosoma mansoni cercariae but display partially reduced resistance. Infect Immun. 66: 3510-3518. Johnson, A. R., T. E. Hugli and H. J. Muller-Eberhard (1975). Release of histamine from rat mast cells by the complement peptides C3a and C5a. Immunology. 28: 1067. Jones, V. E. and B. M. Ogilvie (1971). Protective immunity to Nippostrongylus brasiliensis: the sequence of events which expels worms from the rat intestine. Immunol. 20: 549. Jong, E. C., A. A. Mahmoud and S. J. Klebanoff (1981). Peroxidase-mediated toxicity to schistosomula of Schistosoma mansoni. J Immunol. 126: 468-471. Joseph, M., C. Auriault, M. Capron, J. C. Ameisen, V. Pancre, G. Torpier, J. P. Kusnierz, G. Ovlaque and A. Capron (1985). IgE-dependent platelet cytotoxicity against helminths. Adv Exp Med Biol. 184: 23-33. Kassai, T. (1982). Handbook of Nippostrongylus Brasiliensis (Nematode). Budapest, Commonwealth Agricultural Bureau, Slough, UK. Kay, A. B., R. Moqbel, S. R. Durham, A. J. MacDonald, G. M. Walsh, R. J. Shaw, O. Cromwell and J. Mackay (1985). Leucocyte activation initiated by IgE-dependent mechanisms in relation to helminthic parasitic disease and clinical models of asthma. Int Arch Allergy Appl Immunol. 77: 69-72. Kennedy, M. W. and Y. M. Kuo (1988). The surfaces of the parasitic nematodes Trichinella spiralis and Toxocara canis differ in the binding of post-C3 components of human complement by the alternative pathway. Parasite Immunol. 10: 459-463. Kephart, G. M., G. J. Gleich, D. H. Connor, D. W. Gibson and S. J. Ackerman (1984). Deposition of eosinophil granule major basic protein onto microfilariae of Onchocerca volvulus in the skin of patients treated with diethylcarbamazine. Lab Invest. 50: 51-61. Kerepesi, L. A., J. A. Hess, T. J. Nolan, G. A. Schad and D. Abraham (2006). Complement component C3 is required for protective innate and adaptive immunity to larval Strongyloides stercoralis in mice. J Immunol. 176: 4315-4322.

REFERENCES 75Khan, W. I., M. Richard, H. Akiho, P. A. Blennerhasset, N. E. Humphreys, R. K. Grencis, J. Van Snick and S. M. Collins (2003). Modulation of intestinal muscle contraction by interleukin-9 (IL-9) or IL-9 neutralization: correlation with worm expulsion in murine nematode infections. Infect Immun. 71: 2430-2438. Kinashi, T., N. Harada, E. Severinson, T. Tanabe, P. Sideras, M. Konishi, C. Azuma, A. Tominaga, S. Bergstedt-Lindqvist, M. Takahashi and et al. (1986). Cloning of complementary DNA encoding T-cell replacing factor and identity with B-cell growth factor II. Nature. 324: 70-73. Kinoshita, T., M. E. Medof and V. Nussenzweig (1986). Endogenous association of decay-accelerating factor (DAF) with C4b and C3b on cell membranes. J Immunol. 136: 3390-3395. Klabunde, J., J. Berger, J. C. Jensenius, M. Q. Klinkert, U. E. Zelck, P. G. Kremsner and J. F. Kun (2000). Schistosoma mansoni: adhesion of mannan-binding lectin to surface glycoproteins of cercariae and adult worms. Exp Parasitol. 95: 231-239. Knight, P. A., A. D. Pemberton, K. A. Robertson, D. J. Roy, S. H. Wright and H. R. Miller (2004). Expression profiling reveals novel innate and inflammatory responses in the jejunal epithelial compartment during infection with Trichinella spiralis. Infect Immun. 72: 6076-6086. Knott, M. L., K. I. Matthaei, P. R. Giacomin, H. Wang, P. S. Foster and L. A. Dent (2007). Impaired resistance in early secondary Nippostrongylus brasiliensis infections in mice with defective eosinophilopoeisis. Int J Parasitol. 37: 1367-1378. Kolble, K. and K. B. Reid (1993). Genetic deficiencies of the complement system and association with disease-early components. Int Rev Immunol. 10: 17-36. Komiyama, Y., S. Nakae, T. Matsuki, A. Nambu, H. Ishigame, S. Kakuta, K. Sudo and Y. Iwakura (2006). IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol. 177: 566-573. Kopf, M., F. Brombacher, P. D. Hodgkin, A. J. Ramsay, E. A. Milbourne, W. J. Dai, K. S. Ovington, C. A. Behm, G. Kohler, I. G. Young and K. I. Matthaei (1996). IL-5-deficient mice have a developmental defect in CD5+ B-1 cells and lack eosinophilia but have normal antibody and cytotoxic T cell responses. Immunity. 4: 15-24. Kopf, M., G. Le Gros, M. Bachmann, M. C. Lamers, H. Bluethmann and G. Kohler (1993). Disruption of the murine IL-4 gene blocks Th2 cytokine responses. Nature. 362: 245-248. Korenaga, M., Y. Hitoshi, N. Yamaguchi, Y. Sato, K. Takatsu and I. Tada (1991). The role of interleukin-5 in protective immunity to Strongyloides venezuelensis infection in mice. Immunology. 72: 502-507. Koyama, K., H. Tamauchi and Y. Ito (1995). The role of CD4+ and CD8+ T cells in protective immunity to the murine nematode parasite Trichuris muris. Parasite Immunol. 17: 161-165. Kozel, T. R., L. C. Weinhold and D. M. Lupan (1996). Distinct characteristics of initiation of the classical and alternative complement pathways by Candida albicans. Infect Immun. 64: 3360-3368.

REFERENCES 76Kuijper, P. H., H. I. Gallardo Torres, J. A. van der Linden, J. W. Lammers, J. J. Sixma, J. J. Zwaginga and L. Koenderman (1997). Neutrophil adhesion to fibrinogen and fibrin under flow conditions is diminished by activation and L-selectin shedding. Blood. 89: 2131-2138. La Flamme, A. C., A. S. MacDonald, C. R. Huxtable, M. Carroll and E. J. Pearce (2003). Lack of C3 affects Th2 response development and the sequelae of chemotherapy in schistosomiasis. J Immunol. 170: 470-476. Laurenti, M. D., A. Orn, I. L. Sinhorini and C. E. Corbett (2004). The role of complement in the early phase of Leishmania (Leishmania) amazonensis infection in BALB/c mice. Braz J Med Biol Res. 37: 427-434. Leckie, M. J., A. ten Brinke, J. Khan, Z. Diamant, B. J. O'Connor, C. M. Walls, A. K. Mathur, H. C. Cowley, K. F. Chung, R. Djukanovic, T. T. Hansel, S. T. Holgate, P. J. Sterk and P. J. Barnes (2000). Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 356: 2144-2148. Lee, J. J., D. Dimina, M. P. Macias, S. I. Ochkur, M. P. McGarry, K. R. O'Neill, C. Protheroe, R. Pero, T. Nguyen, S. A. Cormier, E. Lenkiewicz, D. Colbert, L. Rinaldi, S. J. Ackerman, C. G. Irvin and N. A. Lee (2004). Defining a link with asthma in mice congenitally deficient in eosinophils. Science. 305: 1773-1776. Lee, J. J. and N. A. Lee (2005). Eosinophil degranulation: an evolutionary vestige or a universally destructive effector function? Clin Exp Allergy. 35: 986-994. Ligas, J. A., L. A. Kerepesi, A. M. Galioto, S. Lustigman, T. J. Nolan, G. A. Schad and D. Abraham (2003). Specificity and mechanism of immunoglobulin M (IgM)- and IgG-dependent protective immunity to larval Strongyloides stercoralis in mice. Infect Immun. 71: 6835-6843. Lightowlers, M. W., A. L. Colebrook, C. G. Gauci, S. M. Gauci, C. T. Kyngdon, J. L. Monkhouse, C. Vallejo Rodriquez, A. J. Read, R. A. Rolfe and C. Sato (2003). Vaccination against cestode parasites: anti-helminth vaccines that work and why. Vet Parasitol. 115: 83-123. Lightowlers, M. W. and M. D. Rickard (1988). Excretory-secretory products of helminth parasites: effects on host immune responses. Parasitology. 96: S123-166. Linder, E. and G. Huldt (1983). Antibody-independent binding and activation of complement by Schistosoma mansoni adult worms. Parasite Immunol. 5: 183-194. Logan, M. R., S. O. Odemuyiwa and R. Moqbel (2003). Understanding exocytosis in immune and inflammatory cells: the molecular basis of mediator secretion. J Allergy Clin Immunol. 111: 923-932; quiz 933. Loos, M., B. Wellek, R. Thesen and W. Opferkuch (1978). Antibody-independent interaction of the first component of complement with Gram-negative bacteria. Infect Immun. 22: 5-9. Lopez, A. F., C. G. Begley, D. J. Williamson, D. J. Warren, M. A. Vadas and C. J. Sanderson (1986). Murine eosinophil differentiation factor. An eosinophil-specific colony-stimulating factor with activity for human cells. J Exp Med. 163: 1085-1099.

REFERENCES 77Lovchik, J. A. and M. F. Lipscomb (1993). Role for C5 and neutrophils in the pulmonary intravascular clearance of circulating Cryptococcus neoformans. Am J Respir Cell Mol Biol. 9: 617-627. Lukacs, N. W., C. M. Hogaboam, S. L. Kunkel, S. W. Chensue, M. D. Burdick, H. L. Evanoff and R. M. Strieter (1998). Mast cells produce ENA-78, which can function as a potent neutrophil chemoattractant during allergic airway inflammation. J Leukoc Biol. 63: 746-751. MacKenzie, C. D., M. Jungery, P. M. Taylor and B. M. Ogilvie (1980). Activation of complement, the induction of antibodies to the surface of nematodes and the effect of these factors and cells on worm survival in vitro. Eur J Immunol. 10: 594-601. MacKenzie, C. D., M. Jungery, P. M. Taylor and B. M. Ogilvie (1981). The in-vitro interaction of eosinophils, neutrophils, macrophages and mast cells with nematode surfaces in the presence of complement or antibodies. J Pathol. 133: 161-175. MacKenzie, J. R., J. Mattes, L. A. Dent and P. S. Foster (2001). Eosinophils promote allergic disease of the lung by regulating CD4(+) Th2 lymphocyte function. J Immunol. 167: 3146-3155. Madden, K. B., J. F. Urban, Jr., H. J. Ziltener, J. W. Schrader, F. D. Finkelman and I. M. Katona (1991). Antibodies to IL-3 and IL-4 suppress helminth-induced intestinal mastocytosis. J Immunol. 147: 1387-1391. Malaviya, R. and S. N. Abraham (2000). Role of mast cell leukotrienes in neutrophil recruitment and bacterial clearance in infectious peritonitis. J Leukoc Biol. 67: 841-846. Marikovsky, M., R. Arnon and Z. Fishelson (1988). Proteases secreted by transforming schistosomula of Schistosoma mansoni promote resistance to killing by complement. J Immunol. 141: 273-278. Marikovsky, M., F. Levi-Schaffer, R. Arnon and Z. Fishelson (1986). Schistosoma mansoni: killing of transformed schistosomula by the alternative pathway of human complement. Exp Parasitol. 61: 86-94. Marikovsky, M., M. Parizade, R. Arnon and Z. Fishelson (1990). Complement regulation on the surface of cultured schistosomula and adult worms of Schistosoma mansoni. Eur J Immunol. 20: 221-227. Martin, C., L. Le Goff, M. N. Ungeheuer, P. N. Vuong and O. Bain (2000). Drastic reduction of a filarial infection in eosinophilic interleukin-5 transgenic mice. Infect Immun. 68: 3651-3656. Matsuda, S., Y. Tani, M. Yamada, K. Yoshimura and N. Arizono (2001). Type 2-biased expression of cytokine genes in lung granulomatous lesions induced by Nippostrongylus brasiliensis infection. Parasite Immunol. 23: 219-226. Matsumoto, M., W. Fukuda, A. Circolo, J. Goellner, J. Strauss-Schoenberger, X. Wang, S. Fujita, T. Hidvegi, D. D. Chaplin and H. R. Colten (1997). Abrogation of the alternative complement pathway by targeted deletion of murine factor B. Proc Natl Acad Sci U S A. 94: 8720-8725. Matthaei, K. I., P. Foster and I. G. Young (1997). The role of interleukin-5 (IL-5) in vivo: studies with IL-5 deficient mice. Mem Inst Oswaldo Cruz. 92 Suppl 2: 63-68.

REFERENCES 78 Mauser, P. J., A. Pitman, A. Witt, X. Fernandez, J. Zurcher, T. Kung, H. Jones, A. S. Watnick, R. W. Egan, W. Kreutner and et al. (1993). Inhibitory effect of the TRFK-5 anti-IL-5 antibody in a guinea pig model of asthma. Am Rev Respir Dis. 148: 1623-1627. McGuinness, D. H., P. K. Dehal and R. J. Pleass (2003). Pattern recognition molecules and innate immunity to parasites. Trends Parasitol. 19: 312-319. McKenzie, G. J., C. L. Emson, S. E. Bell, S. Anderson, P. Fallon, G. Zurawski, R. Murray, R. Grencis and A. N. McKenzie (1998). Impaired development of Th2 cells in IL-13-deficient mice. Immunity. 9: 423-432. McLaren, D. J., C. D. Mackenzie and F. J. Ramalho-Pinto (1977). Ultrastructural observations on the in vitro interaction between rat eosinophils and some parasitic helminths (Schistosoma mansoni, Trichinella spiralis and Nippostrongylus brasiliensis). Clin Exp Immunol. 30: 105-118. McNeil, K. S., D. P. Knox and L. Proudfoot (2002). Anti-inflammatory responses and oxidative stress in Nippostrongylus brasiliensis-induced pulmonary inflammation. Parasite Immunol. 24: 15-22. Meeusen, E. N. (1999). Immunology of helminth infections, with special reference to immunopathology. Vet Parasitol. 84: 259-273. Meeusen, E. N. and A. Balic (2000). Do eosinophils have a role in the killing of helminth parasites? Parasitol Today. 16: 95-101. Mehlhop, E. and M. S. Diamond (2006). Protective immune responses against West Nile virus are primed by distinct complement activation pathways. J Exp Med. 203: 1371-1381. Melo, A. L., C. R. Machado and L. H. Pereira (1990). Differences between in vitro and in vivo obtained schistosomules. Rev Inst Med Trop Sao Paulo. 32: 84-85. Menzies-Gow, A., S. Ying, I. Sabroe, V. L. Stubbs, D. Soler, T. J. Williams and A. B. Kay (2002). Eotaxin (CCL11) and eotaxin-2 (CCL24) induce recruitment of eosinophils, basophils, neutrophils, and macrophages as well as features of early- and late-phase allergic reactions following cutaneous injection in human atopic and nonatopic volunteers. J Immunol. 169: 2712-2718. Meri, T., T. S. Jokiranta, J. Hellwage, A. Bialonski, P. F. Zipfel and S. Meri (2002). Onchocerca volvulus microfilariae avoid complement attack by direct binding of factor H. J Infect Dis. 185: 1786-1793. Mettrick, D. F., M. E. Budziakowski and R. B. Podesta (1979). Net fluxes of electrolytes in the rat intestine infected with Moniliformis dubius (Acanthocephala). Can J Physiol Pharmacol. 57: 882-886. Miller, H. R. and W. F. Jarrett (1971). Immune reactions in mucous membranes. I. Intestinal mast cell response during helminth expulsion in the rat. Immunology. 20: 277-288. Min, B., M. Prout, J. Hu-Li, J. Zhu, D. Jankovic, E. S. Morgan, J. F. Urban, Jr., A. M. Dvorak, F. D. Finkelman, G. LeGros and W. E. Paul (2004). Basophils produce IL-4 and accumulate in tissues after infection with a Th2-inducing parasite. J Exp Med. 200: 507-517.

REFERENCES 79Mishra, A., S. P. Hogan, J. J. Lee, P. S. Foster and M. E. Rothenberg (1999). Fundamental signals that regulate eosinophil homing to the gastrointestinal tract. J Clin Invest. 103: 1719-1727. Mishra, A., S. P. Hogan, J. J. Lee, P. S. Foster and M. E. Rothenberg (1999). Fundamental signals that regulate eosinophil homing to the gastrointestinal tract. J Clin Invest. 103: 1719-1727. Mold, C., B. Rodic-Polic and T. W. Du Clos (2002). Protection from Streptococcus pneumoniae infection by C-reactive protein and natural antibody requires complement but not Fc gamma receptors. J Immunol. 168: 6375-6381. Mosmann, T. R. and R. L. Coffman (1989). TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol. 7: 145-173. Mould, A. W., K. I. Matthaei, I. G. Young and P. S. Foster (1997). Relationship between interleukin-5 and eotaxin in regulating blood and tissue eosinophilia in mice. J Clin Invest. 99: 1064-1071. Mullick, A., M. Elias, S. Picard, L. Bourget, O. Jovcevski, S. Gauthier, A. Tuite, P. Harakidas, C. Bihun, B. Massie and P. Gros (2004). Dysregulated inflammatory response to Candida albicans in a C5-deficient mouse strain. Infect Immun. 72: 5868-5876. Nagai, H., S. Yamaguchi, H. Tanaka, S. Sur, G. J. Gleich, M. C. Swanson, K. R. Bartemes and D. H. Broide (1996). The role of interleukin-5 (IL-5) in allergic airway hyperresponsiveness in mice. Ann N Y Acad Sci. 796: 91-96. Nair, M. G., I. J. Gallagher, M. D. Taylor, P. Loke, P. S. Coulson, R. A. Wilson, R. M. Maizels and J. E. Allen (2005). Chitinase and Fizz family members are a generalized feature of nematode infection with selective upregulation of Ym1 and Fizz1 by antigen-presenting cells. Infect Immun. 73: 385-394. Nawa, Y., N. Ishikawa, K. Tsuchiya, Y. Horii, T. Abe, A. I. Khan, S. Bing, H. Itoh, H. Ide and F. Uchiyama (1994). Selective effector mechanisms for the expulsion of intestinal helminths. Parasite Immunol. 16: 333-338. Negrao-Correa, D., V. Pinho, D. G. Souza, A. T. Pereira, A. Fernandes, K. Scheuermann, A. L. Souza and M. M. Teixeira (2006). Expression of IL-4 receptor on non-bone marrow-derived cells is necessary for the timely elimination of Strongyloides venezuelensis in mice, but not for intestinal IL-4 production. Int J Parasitol. 36: 1185-1195. Netea, M. G., C. van der Graaf, J. W. Van der Meer and B. J. Kullberg (2004). Toll-like receptors and the host defense against microbial pathogens: bringing specificity to the innate-immune system. J Leukoc Biol. 75: 749-755. Nonaka, R., M. Nonaka, S. Takanashi, M. Jordana and J. Dolovich (1999). Eosinophil activation in the tissue: synthetic steroid, budesonide, effectively inhibits the survival of eosinophils isolated from peripheral blood but not nasal polyp tissues. J Clin Lab Immunol. 51: 39-53. Nutten, S., J. P. Papin, G. Woerly, D. W. Dunne, J. MacGregor, F. Trottein and M. Capron (1999). Selectin and Lewis(x) are required as co-receptors in antibody-dependent cell-mediated cytotoxicity of human eosinophils to Schistosoma mansoni schistosomula. Eur J Immunol. 29: 799-808.

REFERENCES 80 Ogilvie, B. M. and V. E. Jones (1971). Nippostrongylus brasiliensis: A review of immunity and the host/parasite relationship in the rat. Exp. Parasitology. 29: 138-177. Ogilvie, B. M. and V. E. Jones (1971). Parasitological review. Nippostrongylus brasiliensis: a review of immunity and host-parasite relationship in the rat. Exp Parasitol. 29: 138-177. Ohta, H., Y. Yoshikawa, C. Kai, K. Yamanouchi, H. Taniguchi, K. Komine, Y. Ishijima and H. Okada (1986). Effect of complement depletion by cobra venom factor on fowlpox virus infection in chickens and chicken embryos. J Virol. 57: 670-673. Okinaga, S., D. Slattery, A. Humbles, Z. Zsengeller, O. Morteau, M. B. Kinrade, R. M. Brodbeck, J. E. Krause, H. R. Choe, N. P. Gerard and C. Gerard (2003). C5L2, a nonsignaling C5A binding protein. Biochemistry. 42: 9406-9415. Oksjoki, R., P. T. Kovanen and M. O. Pentikainen (2003). Role of complement activation in atherosclerosis. Curr Opin Lipidol. 14: 477-482. Onah, D. N. and Y. Nawa (2000). Mucosal immunity against parasitic gastrointestinal nematodes. Korean J Parasitol. 38: 209-236. Ovington, K. S., K. McKie, K. I. Matthaei, I. G. Young and C. A. Behm (1998). Regulation of primary Strongyloides ratti infections in mice: a role for interleukin-5. Immunology. 95: 488-493. Owhashi, M., H. Arita and N. Hayai (2000). Identification of a novel eosinophil chemotactic cytokine (ECF-L) as a chitinase family protein. J Biol Chem. 275: 1279-1286. Padigel, U. M., J. J. Lee, T. J. Nolan, G. A. Schad and D. Abraham (2006). Eosinophils can function as antigen-presenting cells to induce primary and secondary immune responses to Strongyloides stercoralis. Infect Immun. 74: 3232-3238. Pearce, E. J., B. F. Hall and A. Sher (1990). Host-specific evasion of the alternative complement pathway by schistosomes correlates with the presence of a phospholipase C-sensitive surface molecule resembling human decay accelerating factor. J Immunol. 144: 2751-2756. Pemberton, A. D., P. A. Knight, J. Gamble, W. H. Colledge, J. K. Lee, M. Pierce and H. R. Miller (2004). Innate BALB/c enteric epithelial responses to Trichinella spiralis: inducible expression of a novel goblet cell lectin, intelectin-2, and its natural deletion in C57BL/10 mice. J Immunol. 173: 1894-1901. Penttila, I. A., P. L. Ey, C. R. Jenkin, F. Santoro, P. J. Lachmann, A. Capron, M. Capron, A. R. Anwar, S. R. Smithers and A. B. Kay (1983). Adherence of murine peripheral blood eosinophils and neutrophils to the different parasitic stages of Nematospiroides dubius. Aust J Exp Biol Med Sci. 61: 617-627. Pepys, M. B. (1974). Role of complement in induction of antibody production in vivo. Effect of cobra factor and other C3-reactive agents on thymus-dependent and thymus-independent antibody responses. J Exp Med. 140: 126-145. Persson, C. G., J. S. Erjefalt, M. Korsgren and F. Sundler (1997). The mouse trap. Trends Pharmacol Sci. 18: 465-467.

REFERENCES 81Pesce, J., M. Kaviratne, T. R. Ramalingam, R. W. Thompson, J. F. Urban, Jr., A. W. Cheever, D. A. Young, M. Collins, M. J. Grusby and T. A. Wynn (2006). The IL-21 receptor augments Th2 effector function and alternative macrophage activation. J Clin Invest. 116: 2044-2055. Petersen, S. V., S. Thiel and J. C. Jensenius (2001). The mannan-binding lectin pathway of complement activation: biology and disease association. Mol Immunol. 38: 133-149. Phipps, S., C. E. Lam, S. Mahalingam, M. Newhouse, R. Ramirez, H. F. Rosenberg, P. S. Foster and K. I. Matthaei (2007). Eosinophils contribute to innate anti-viral immunity and promote clearance of respiratory syncytial virus. Blood. Pickering, M. C., H. T. Cook, J. Warren, A. E. Bygrave, J. Moss, M. J. Walport and M. Botto (2002). Uncontrolled C3 activation causes membranoproliferative glomerulonephritis in mice deficient in complement factor H. Nat Genet. 31: 424-428. Plotz, S. G., A. Lentschat, H. Behrendt, W. Plotz, L. Hamann, J. Ring, E. T. Rietschel, H. D. Flad and A. J. Ulmer (2001). The interaction of human peripheral blood eosinophils with bacterial lipopolysaccharide is CD14 dependent. Blood. 97: 235-241. Preston, C. M., T. Jenkins and D. J. McLaren (1986). Surface properties of developing stages of Trichuris muris. Parasite Immunol. 8: 597-611. Prowse, S. J., P. L. Ey and C. R. Jenkin (1979). Alternative pathway activation of complement by a murine parasitic nematode (Nematospiroides dubius). Aust J Exp Biol Med Sci. 57: 459-466. Rainbird, M. A., D. Macmillan and E. N. Meeusen (1998). Eosinophil-mediated killing of Haemonchus contortus larvae: effect of eosinophil activation and role of antibody, complement and interleukin-5. Parasite Immunol. 20: 93-103. Ramalho-Pinto, F. J. (1987). Decay accelerating factor (DAF) as the host antigen with protective activity to complement killing of schistosomula. Mem Inst Oswaldo Cruz. 82 Suppl 4: 213-216. Ramalingam, T., L. Ganley-Leal, P. Porte and T. V. Rajan (2003). Impaired clearance of primary but not secondary Brugia infections in IL-5 deficient mice. Exp Parasitol. 105: 131-139. Ramalingam, T., P. Porte, J. Lee and T. V. Rajan (2005). Eosinophils, but not eosinophil peroxidase or major basic protein, are important for host protection in experimental Brugia pahangi infection. Infect Immun. 73: 8442-8443. Ramaswamy, K. and D. Befus (1989). Pulmonary inflammation in parasitic infection: immunoglobulins in bronchoalveolar washings of rats infected with Nippostrongylus brasiliensis. Parasite Immunol. 11: 655-665. Ramaswamy, K. and D. Befus (1993). Pulmonary immune responses to Nippostrongylus brasiliensis: isotype-specific antibodies in bronchoalvelor lavage fluids of rats. Parasite Immunol. 15: 573-582. Reece, J. J., M. C. Siracusa and A. L. Scott (2006). Innate immune responses to lung-stage helminth infection induce alternatively activated alveolar macrophages. Infect Immun. 74: 4970-4981.

REFERENCES 82Reese, T. A., H. E. Liang, A. M. Tager, A. D. Luster, N. Van Rooijen, D. Voehringer and R. M. Locksley (2007). Chitin induces accumulation in tissue of innate immune cells associated with allergy. Nature. 447: 92-96. Reis e Sousa, C. (2001). Dendritic cells as sensors of infection. Immunity. 14: 495-498. Rennick, D. M., L. Thompson-Snipes, R. L. Coffman, B. W. Seymour, J. D. Jackson and S. Hudak (1990). In vivo administration of antibody to interleukin-5 inhibits increased generation of eosinophils and their progenitors in bone marrow of parasitized mice. Blood. 76: 312-316. Resnick, M. B. and P. F. Weller (1993). Mechanisms of eosinophil recruitment. Am J Respir Cell Mol Biol. 8: 349-355. Romagnani, S. (2000). T-cell subsets (Th1 versus Th2). Ann Allergy Asthma Immunol. 85: 9-18; quiz 18, 21. Rothenberg, M. E. (2004). Eosinophilic gastrointestinal disorders (EGID). J Allergy Clin Immunol. 113: 11-28; quiz 29. Rothenberg, M. E. and S. P. Hogan (2006). The eosinophil. Annu Rev Immunol. 24: 147-174. Rothenberg, M. E., A. Mishra, E. B. Brandt and S. P. Hogan (2001). Gastrointestinal eosinophils in health and disease. Adv Immunol. 78: 291-328. Rother, K. and G. O. Till (1988). The Complement System. Heidelberg, Springer-Verlag. Ruppel, A., D. J. McLaren, H. J. Diesfeld and U. Rother (1984). Schistosoma mansoni: escape from complement-mediated parasiticidal mechanisms following percutaneous primary infection. Eur J Immunol. 14: 702-708. Ruppel, A., U. Rother and H. J. Diesfeld (1982). Schistosoma mansoni: development of primary infections in mice genetically deficient or intact in the fifth component of complement. Parasitology. 85 (Pt 2): 315-323. Ruppel, A., U. Rother and H. J. Diesfeld (1984). Schistosoma mansoni: loss of the ability of schistosomula to bind mouse complement following intravenous injection into mice. Tropenmed Parasitol. 35: 23-28. Sandground, J. H. (1936). On the Potential Longevity of Various Helminths with a Record for a Species of Trichostrongylus in Man. J. Parasitol. 22: 464-470. Santoro, F., P. J. Lachmann, A. Capron and M. Capron (1979). Activation of complement by Schistosoma mansoni schistosomula: killing of parasites by the alternative pathway and requirement of IgG for classical pathway activation. J Immunol. 123: 1551-1557. Santoro, F., B. Vandemeulebroucke, M. C. Liebart and A. Capron (1982). Schistosoma mansoni: role in vivo of complement in primary infection of mice. Exp Parasitol. 54: 40-46. Sarma, V. J., M. Huber-Lang and P. A. Ward (2006). Complement in lung disease. Autoimmunity. 39: 387-394.

REFERENCES 83Sasaki, O., H. Sugaya, K. Ishida and K. Yoshimura (1993). Ablation of eosinophils with anti-IL-5 antibody enhances the survival of intracranial worms of Angiostrongylus cantonensis in the mouse. Parasite Immunol. 15: 349-354. Schallig, H. D., M. A. van Leeuwen and A. W. Cornelissen (1997). Protective immunity induced by vaccination with two Haemonchus contortus excretory secretory proteins in sheep. Parasite Immunol. 19: 447-453. Sferruzzi-Perri, A. N., S. A. Robertson and L. A. Dent (2003). Interleukin-5 transgene expression and eosinophilia are associated with retarded mammary gland development in mice. Biol Reprod. 69: 224-233. Shaio, M. F., S. C. Hou, J. G. Chen, C. C. Wu, K. D. Yang and F. Y. Chang (1990). Immunoglobulin G-dependent classical complement pathway activation in neutrophil-mediated cytotoxicity to infective larvae of Angiostrongylus cantonensis. Ann Trop Med Parasitol. 84: 185-191. Sher, A., R. L. Coffman, S. Hieny and A. W. Cheever (1990). Ablation of eosinophil and IgE responses with anti-IL-5 or anti-IL-4 antibodies fails to affect immunity against Schistosoma mansoni in the mouse. J Immunol. 145: 3911-3916. Sher, A., S. Hieny, S. L. James and R. Asofsky (1982). Mechanisms of protective immunity against Schistosoma mansoni infection in mice vaccinated with irradiated cercariae. II. Analysis of immunity in hosts deficient in T lymphocytes, B lymphocytes, or complement. J Immunol. 128: 1880-1884. Sher, A., S. L. James, A. J. Simpson, J. K. Lazdins and M. S. Meltzer (1982). Macrophages as effector cells of protective immunity in murine schistosomiasis. III. Loss of susceptibility to macrophage-mediated killing during maturation of S. mansoni schistosomula from the skin to the lung stage. J Immunol. 128: 1876-1879. Shin, E. H., Y. Osada, J. Y. Chai, N. Matsumoto, K. Takatsu and S. Kojima (1997). Protective roles of eosinophils in Nippostrongylus brasiliensis infection. Int Arch Allergy Immunol. 114: 45-50. Shin, E. H., Y. Osada, H. Sagara, K. Takatsu and S. Kojima (2001). Involvement of complement and fibronectin in eosinophil-mediated damage to Nippostrongylus brasiliensis larvae. Parasite Immunol. 23: 27-37. Shin, H. S., H. Gewurz and R. Snyderman (1969). Reaction of a cobra venom factor with guinea pig complement and generation of an activity chemotactic for polymorphonuclear leukocytes. Proc Soc Exp Biol Med. 131: 203-207. Shinkai, K., M. Mohrs and R. M. Locksley (2002). Helper T cells regulate type-2 innate immunity in vivo. Nature. 420: 825-829. Simon, D., L. R. Braathen and H. U. Simon (2005). Anti-interleukin-5 antibody therapy in eosinophilic diseases. Pathobiology. 72: 287-292. Simons, J. E., M. E. Rothenberg and R. A. Lawrence (2005). Eotaxin-1-regulated eosinophils have a critical role in innate immunity against experimental Brugia malayi infection. Eur J Immunol. 35: 189-197.

REFERENCES 84Specht, S., M. Saeftel, M. Arndt, E. Endl, B. Dubben, N. A. Lee, J. J. Lee and A. Hoerauf (2006). Lack of Eosinophil Peroxidase or Major Basic Protein Impairs Defense against Murine Filarial Infection. Infect Immun. 74: 5236-5243. Stankiewicz, M., G. Sokolska, A. Pilarczyk, D. Sadowska and E. L. Jeska (1989). Lack of correlation between in vitro and in vivo activation of complement in mice by infective Trichinella spiralis larvae. J Parasitol. 75: 647-649. Suchitra, S. and P. Joshi (2005). Characterization of Haemonchus contortus calreticulin suggests its role in feeding and immune evasion by the parasite. Biochim Biophys Acta. 1722: 293-303. Sugane, K., Y. Kusama, M. Takamoto, A. Tominaga and K. Takatsu (1996). Eosinophilia, IL-5 level and recovery of larvae in IL-5 transgenic mice infected with Toxocara canis. J Helminthol. 70: 153-158. Sugaya, H., M. Aoki, T. Yoshida, K. Takatsu and K. Yoshimura (1997). Eosinophilia and intracranial worm recovery in interleukin-5 transgenic and interleukin-5 receptor alpha chain-knockout mice infected with Angiostrongylus cantonensis. Parasitol Res. 83: 583-590. Sugaya, H., C. Graeff-Teixeira, K. Ishida, S. Matsuda, K. Katahira and K. Yoshimura (2002). Interleukin-5 transgenic mice show augmented resistance to Angiostrongylus costaricensis infection. Parasitol Res. 88: 350-355. Sur, S., G. J. Gleich, M. C. Swanson, K. R. Bartemes and D. H. Broide (1995). Eosinophilic inflammation is associated with elevation of interleukin-5 in the airways of patients with spontaneous symptomatic asthma. J Allergy Clin Immunol. 96: 661-668. Symons, L. E. (1978). Epithelia cell mitosis and morphology in worm-free regions of the intestines of the rat infected by Nippostrongylus brasiliensis. J Parasitol. 64: 958-959. Takafuji, S., K. Tadokoro and K. Ito (1996). Effects of interleukin (IL)-3 and IL-5 on human eosinophil degranulation induced by complement components C3a and C5a. Allergy. 51: 563-568. Takamoto, M., K. S. Ovington, C. A. Behm, K. Sugane, I. G. Young and K. I. Matthaei (1997). Eosinophilia, parasite burden and lung damage in Toxocara canis infection in C57Bl/6 mice genetically deficient in IL-5. Immunology. 90: 511-517. Takeda, K., T. Tanaka, W. Shi, M. Matsumoto, M. Minami, S. Kashiwamura, K. Nakanishi, N. Yoshida, T. Kishimoto and S. Akira (1996). Essential role of Stat6 in IL-4 signalling. Nature. 380: 627-630. Taliaferro, W. H. and M. P. Sarles (1939). The cellular reactions in the skin, lungs and intestine of normal and immune rats after infection with Nippostrongylus muris. J. Infect. Dis. 64: 157-192. Tavares, C. A., G. Gazzinelli, T. A. Mota-Santos and W. Dias Da Silva (1978). Schistosoma mansoni: complement-mediated cytotoxic activity in vitro and effect of decomplementation on acquired immunity in mice. Exp Parasitol. 46: 145-151. Taylor, P. R., E. Seixas, M. J. Walport, J. Langhorne and M. Botto (2001). Complement contributes to protective immunity against reinfection by Plasmodium chabaudi chabaudi parasites. Infect Immun. 69: 3853-3859.

REFERENCES 85 Tedeschi, A., G. Palumbo, N. Milazzo and A. Miadonna (1994). Nasal neutrophilia and eosinophilia induced by challenge with platelet activating factor. J Allergy Clin Immunol. 93: 526-533. Tiberio, I. F., E. A. Leick-Maldonado, L. Miyahara, D. I. Kasahara, G. M. Spilborghs, M. A. Martins and P. H. Saldiva (2003). Effects of neurokinins on airway and alveolar eosinophil recruitment. Exp Lung Res. 29: 165-177. Tkalcevic, J., M. R. Brandon and E. N. Meeusen (1996). Fasciola hepatica: rapid switching of stage-specific antigen expression after infection. Parasite Immunol. 18: 139-147. Tominaga, A., S. Takaki, N. Koyama, S. Katoh, R. Matsumoto, M. Migita, Y. Hitoshi, Y. Hosoya, S. Yamauchi, Y. Kanai and et al. (1991). Transgenic mice expressing a B cell growth and differentiation factor gene (interleukin 5) develop eosinophilia and autoantibody production. J Exp Med. 173: 429-437. Uber, C. L., R. L. Roth and D. A. Levy (1980). Expulsion of Nippostrongylus brasiliensis by mice deficient in mast cells. Nature. 287: 226-228. Urban, J. F., Jr., I. M. Katona and F. D. Finkelman (1991). Heligmosomoides polygyrus: CD4+ but not CD8+ T cells regulate the IgE response and protective immunity in mice. Exp Parasitol. 73: 500-511. Urban, J. F., Jr., K. B. Madden, A. W. Cheever, P. P. Trotta, I. M. Katona and F. D. Finkelman (1993). IFN inhibits inflammatory responses and protective immunity in mice infected with the nematode parasite, Nippostrongylus brasiliensis. J Immunol. 151: 7086-7094. Urban, J. F., Jr., K. B. Madden, A. Svetic, A. Cheever, P. P. Trotta, W. C. Gause, I. M. Katona and F. D. Finkelman (1992). The importance of Th2 cytokines in protective immunity to nematodes. Immunol Rev. 127: 205-220. Urban, J. F., Jr., N. Noben-Trauth, D. D. Donaldson, K. B. Madden, S. C. Morris, M. Collins and F. D. Finkelman (1998). IL-13, IL-4Ralpha, and Stat6 are required for the expulsion of the gastrointestinal nematode parasite Nippostrongylus brasiliensis. Immunity. 8: 255-264. Urban, J. F., Jr., N. Noben-Trauth, L. Schopf, K. B. Madden and F. D. Finkelman (2001). Cutting edge: IL-4 receptor expression by non-bone marrow-derived cells is required to expel gastrointestinal nematode parasites. J Immunol. 167: 6078-6081. Vallance, B. A., P. A. Blennerhassett and S. M. Collins (1997). Increased intestinal muscle contractility and worm expulsion in nematode-infected mice. Am J Physiol. 272: G321-327. Vallance, B. A., K. I. Matthaei, S. Sanovic, I. G. Young and S. M. Collins (2000). Interleukin-5 deficient mice exhibit impaired host defence against challenge Trichinella spiralis infections. Parasite Immunol. 22: 487-492. van Beek, J., K. Elward and P. Gasque (2003). Activation of complement in the central nervous system: roles in neurodegeneration and neuroprotection. Ann N Y Acad Sci. 992: 56-71. van der Kleij, D., E. Latz, J. F. Brouwers, Y. C. Kruize, M. Schmitz, E. A. Kurt-Jones, T. Espevik, E. C. de Jong, M. L. Kapsenberg, D. T. Golenbock, A. G. Tielens and M.

REFERENCES 86Yazdanbakhsh (2002). A novel host-parasite lipid cross-talk. Schistosomal lyso-phosphatidylserine activates toll-like receptor 2 and affects immune polarization. J Biol Chem. 277: 48122-48129. Vaux, D. L., P. A. Lalor, S. Cory and G. R. Johnson (1990). In vivo expression of interleukin 5 induces an eosinophilia and expanded Ly-1B lineage populations. Int Immunol. 2: 965-971. Venturiello, S. M., G. H. Giambartolomei and S. N. Costantino (1995). Immune cytotoxic activity of human eosinophils against Trichinella spiralis newborn larvae. Parasite Immunol. 17: 555-559. Vercauteren, I., P. Geldhof, J. Vercruysse, I. Peelaers, W. van den Broeck, K. Gevaert and E. Claerebout (2004). Vaccination with an Ostertagia ostertagi polyprotein allergen protects calves against homologous challenge infection. Infect Immun. 72: 2995-3001. Vignali, D. A., Q. D. Bickle, M. G. Taylor, G. Tennent and M. B. Pepys (1988). Comparison of the role of complement in immunity to Schistosoma mansoni in rats and mice. Immunology. 63: 55-61. Voehringer, D., K. Shinkai and R. M. Locksley (2004). Type 2 immunity reflects orchestrated recruitment of cells committed to IL-4 production. Immunity. 20: 267-277. von Lichtenberg, F., A. Sher and S. McIntyre (1977). A lung model of schistosome immunity in mice. Am J Pathol. 87: 105-123. von Samson-Himmelstjerna, G. and W. Blackhall (2005). Will technology provide solutions for drug resistance in veterinary helminths? Vet Parasitol. 132: 223-239. Wallis, R., A. W. Dodds, D. A. Mitchell, R. B. Sim, K. B. Reid and W. J. Schwaeble (2007). Molecular interactions between MASP-2, C4, and C2 and their activation fragments leading to complement activation via the lectin pathway. J Biol Chem. 282: 7844-7851. Wang, C. H. and R. G. Bell (1988). Antibody-mediated in-vivo cytotoxicity to Trichinella spiralis newborn larvae in immune rats. Parasite Immunol. 10: 293-308. Warren, D. J. and M. A. Moore (1988). Synergism among interleukin 1, interleukin 3, and interleukin 5 in the production of eosinophils from primitive hemopoietic stem cells. J Immunol. 140: 94-99. Warren, D. J. and C. J. Sanderson (1985). Production of a T-cell hybrid producing a lymphokine stimulating eosinophil differentiation. Immunology. 54: 615-623. Weller, P. F. (1991). The immunobiology of eosinophils. N Engl J Med. 324: 1110-1118. Weller, P. F., T. H. Rand, T. Barrett, A. Elovic, D. T. Wong and R. W. Finberg (1993). Accessory cell function of human eosinophils. HLA-DR-dependent, MHC-restricted antigen-presentation and IL-1 alpha expression. J Immunol. 150: 2554-2562. Wessels, M. R., P. Butko, M. Ma, H. B. Warren, A. L. Lage and M. C. Carroll (1995). Studies of group B streptococcal infection in mice deficient in complement component C3 or C4 demonstrate an essential role for complement in both innate and acquired immunity. Proc Natl Acad Sci U S A. 92: 11490-11494.

REFERENCES 87Whelan, M., M. M. Harnett, K. M. Houston, V. Patel, W. Harnett and K. P. Rigley (2000). A filarial nematode-secreted product signals dendritic cells to acquire a phenotype that drives development of Th2 cells. J Immunol. 164: 6453-6460. Williams, T. J. (2004). The eosinophil enigma. J Clin Invest. 113: 507-509. Winkelstein, J. A. and E. R. Moxon (1992). The role of complement in the host's defense against Haemophilus influenzae. J Infect Dis. 165 Suppl 1: S62-65. Winkelstein, J. A., M. R. Smith and H. S. Shin (1975). The role of C3 as an opsonin in the early stages of infection. Proc Soc Exp Biol Med. 149: 397-401. Wong, D. T. W., A. Elovic, K. Matossian, N. Nagura, J. McBride, M. Y. Chou, J. R. Gordon, T. H. Rand, S. J. Galli and P. F. Weller (1991). Eosinophils from patients with blood eosinophilia express transforming growth factor- β1. Blood. 78: 2702-2707. Woodbury, R. G., H. R. P. Miller, J. F. Huntley, H. G. F. J. Newlands, A. C. Palliser and C. Wakelin (1984). Mucosal mast cells are functionally active during spontaneous expulsion of intestinal nematode infections in the rat. Nature. 32: 450-452. Yu, C., A. B. Cantor, H. Yang, C. Browne, R. A. Wells, Y. Fujiwara and S. H. Orkin (2002). Targeted deletion of a high-affinity GATA-binding site in the GATA-1 promoter leads to selective loss of the eosinophil lineage in vivo. J Exp Med. 195: 1387-1395. Yuste, J., S. Ali, S. Sriskandan, C. Hyams, M. Botto and J. S. Brown (2006). Roles of the alternative complement pathway and C1q during innate immunity to Streptococcus pyogenes. J Immunol. 176: 6112-6120. Zhao, M., M. Yang, E. Baranov, X. Wang, S. Penman, A. R. Moossa and R. M. Hoffman (2001). Spatial-temporal imaging of bacterial infection and antibiotic response in intact animals. Proc Natl Acad Sci U S A. 98: 9814-9818. Zhu, Y., S. Thangamani, B. Ho and J. L. Ding (2005). The ancient origin of the complement system. Embo J. 24: 382-394. Zhu, Z., T. Zheng, R. J. Homer, Y. K. Kim, N. Y. Chen, L. Cohn, Q. Hamid and J. A. Elias (2004). Acidic mammalian chitinase in asthmatic Th2 inflammation and IL-13 pathway activation. Science. 304: 1678-1682.