Download as pdf or txt
Download as pdf or txt
You are on page 1of 36

Avian Immunology 3rd Edition Bernd

Kaspers
Visit to download the full and correct content document:
https://ebookmass.com/product/avian-immunology-3rd-edition-bernd-kaspers/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

Immunology for medical students 3rd edition Edition


Helbert

https://ebookmass.com/product/immunology-for-medical-
students-3rd-edition-edition-helbert/

Avian Medicine, 3e Third Edition Jaime Samour

https://ebookmass.com/product/avian-medicine-3e-third-edition-
jaime-samour/

The Grammar of Interactives Bernd Heine

https://ebookmass.com/product/the-grammar-of-interactives-bernd-
heine/

Fundamental Immunology Seventh Edition

https://ebookmass.com/product/fundamental-immunology-seventh-
edition/
Immunology Ramesh

https://ebookmass.com/product/immunology-ramesh/

Valkyrie: The Avian Clone Chronicles Cheryl Brooks

https://ebookmass.com/product/valkyrie-the-avian-clone-
chronicles-cheryl-brooks/

Immunology , 9th Edition Male David

https://ebookmass.com/product/immunology-9th-edition-male-david/

Cellular and Molecular Immunology 9th Edition Abul K.


Abbas

https://ebookmass.com/product/cellular-and-molecular-
immunology-9th-edition-abul-k-abbas/

Clinical Immunology Nima Rezaei

https://ebookmass.com/product/clinical-immunology-nima-rezaei/
Avian Immunology
Avian Immunology
Third Edition

Edited by
Bernd Kaspers
Department for Veterinary Sciences, Veterinary Immunology Study Group,
University of Munich, Munich, Germany

Karel A. Schat
Department of Microbiology and Immunology, College of Veterinary Medicine,
Cornell University, Ithaca, NY, United States

Thomas W. Göbel
Department for Veterinary Sciences, Veterinary Immunology Study Group,
University of Munich, Munich, Germany

Lonneke Vervelde
Division Infection and Immunity, The Roslin Institute and R(D)SVS,
The University of Edinburgh, Midlothian, United Kingdom
Academic Press is an imprint of Elsevier
125 London Wall, London EC2Y 5AS, United Kingdom
525 B Street, Suite 1650, San Diego, CA 92101, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
Copyright © 2022 Elsevier Ltd. All rights reserved.
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical,
including photocopying, recording, or any information storage and retrieval system, without permission in writing from the
publisher. Details on how to seek permission, further information about the Publisher’s permissions policies and our
arrangements with organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at
our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may
be noted herein).
Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding,
changes in research methods, professional practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using any
information, methods, compounds, or experiments described herein. In using such information or methods they should be
mindful of their own safety and the safety of others, including parties for whom they have a professional responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any
injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or
operation of any methods, products, instructions, or ideas contained in the material herein.

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library
Library of Congress Cataloging-in-Publication Data
A catalog record for this book is available from the Library of Congress

ISBN: 978-0-12-818708-1

For Information on all Academic Press publications


visit our website at https://www.elsevier.com/books-and-journals

Publisher: Andre Gerhard Wolff


Acquisitions Editor: Linda Versteeg-buschman
Editorial Project Manager: Susan Ikeda
Production Project Manager: Niranjan Bhaskaran
Cover Designer: Christian J. Bilbow
Back Cover Images by ‘Karel “Ton” A. Schat’
Typeset by MPS Limited, Chennai, India
Dedication

Dedicated to

Professor Pete Kaiser 1964 2016


Coeditor Second Edition Avian Immunology
Valued Avian Immunologist and Sorely Missed by the Editors of the
Third Edition of Avian Immunology.
Contents

List of contributors xvii 2.3.11 Germinal center of the


Foreword xix peripheral lymphoid organs 21
Acknowledgments xxi 2.4 The spleen 22
2.4.1 Origin and anatomy 22
1. The importance of the avian immune 2.4.2 Periarteriolar lymphoid sheath 25
system and its unique features 1 2.4.3 Ellipsoids and periellipsoid
white pulp 25
Fred Davison 2.4.4 The marginal-zone equivalent
1.1 Introduction 1 and antigen handling 27
1.2 The contribution from avian 2.5 Gut-associated lymphoid tissue 28
lymphocytes 1 2.5.1 Follicle-associated epithelium or
1.3 Contribution of the bursa of Fabricius 2 lymphoepithelium 30
1.3.1 Gene conversion and the bursa 4 2.5.2 Esophageal and pyloric tonsils 30
1.4 The contribution of the chicken MHC 5 2.5.3 Peyer’s patches 31
1.5 Contributions to vaccinology 6 2.5.4 Meckel’s diverticulum 31
1.5.1 Embryonic (in ovo) vaccination 7 2.5.5 Cecal tonsils 32
1.6 Conclusion 8 2.6 Harderian gland 33
References 8 2.7 Mural lymph node 34
2.8 Ectopic lymphatic tissue and pineal
2. Structure of the avian lymphoid gland 36
system 11 2.9 Bone marrow 37
2.10 Blood 38
Nándor Nagy, Imre Oláh and
References 39
Lonneke Vervelde
2.1 Introduction 11
2.2 The thymus 12 3. Development of the avian
2.2.1 Anatomy and histological hematopoietic and immune
organization 12 systems 45
2.2.2 Thymic cortex 13
Laurent Yvernogeau, Nándor Nagy,
2.2.3 Thymic medulla 13
Dominique Dunon, Catherine Robin and
2.3 The bursa of Fabricius 15 Thierry Jaffredo
2.3.1 Anatomy and histology 15 3.1 Introduction 45
2.3.2 Bursal surface epithelium 16 3.2 Origins and migration routes of
2.3.3 Bursal follicle 17 hematopoietic cells using quail/chicken
2.3.4 Medulla 18 complementary chimeras 45
2.3.5 Bursal medullary epithelial 3.2.1 Looking for the source of
cells 18 hematopoeietic cells during
2.3.6 Bursal secretory dendritic cells 19 development 45
2.3.7 Bursal macrophages 19 3.2.2 Macrophage production by the
2.3.8 Bursal lymphocytes 21 yolk sac 46
2.3.9 Cortex 21 3.2.3 The aortic region produces HSCs 46
2.3.10 Peripheral lymphoid tissue of 3.3 Aortic clusters as the intraembryonic
the bursa of Fabricius 21 source of definitive hematopoiesis 46

vii
viii Contents

3.3.1 Cellular and molecular 4. B cells, the bursa of Fabricius, and the
identification of the clusters 46 generation of antibody repertoires 71
3.3.2 The paraaortic foci 46
3.3.3 Tracing the origins and fates Michael J.H. Ratcliffe and Sonja Härtle
of the aortic clusters 46 4.1 Introduction 71
3.4 Formation of the aorta: a dorsal 4.2 The generation of avian antibody
angioblastic lineage and a ventral repertoires 71
hemangioblasts lineage 48 4.2.1 Immunoglobulin light chains 71
3.4.1 Two endothelial lineages form the 4.2.2 Immunoglobulin heavy chains 72
vascular network of the embryo 48 4.2.3 Generation of Ig molecules by
3.4.2 Chimeric origin of the aortic V(D)J recombination 74
endothelial cells 48 4.2.4 Generation of Ig diversity by
3.5 Developing an in vitro model of somatic gene conversion 75
hemogenic endothelium commitment 4.2.5 Implications of gene conversion
and endothelial-to-hematopoietic for allelic exclusion 77
transition 50 4.3 The development of avian B cells 77
3.6 Spatiotemporal emergence and 4.3.1 Prebursal B cell development 77
organization of the chicken IAHCs 52 4.3.2 Colonization of the bursa by
3.7 Ecs of the late fetus/young adult bone B cell progenitors 78
marrow harbor hemogenic potential 4.3.3 Colonization of lymphoid
and generate multilineage follicles in the bursa 79
hematopoiesis 54 4.3.4 Growth of bursal B cells in
3.8 Spatial transcriptomics in the chicken bursal follicles 82
embryo reveals regulators of 4.3.5 Development of the bursa after
hematopoiesis 55 hatch 83
3.9 The avian thymus and T-cell 4.3.6 Role of cell adhesion molecules
development 57 and chemokines in bursal cell
3.9.1 Thymic development 57 development 85
3.9.2 Colonization of the thymus 57 4.3.7 Development of peripheral
3.9.3 T-cell differentiation 57 B cell populations 87
3.9.4 TCR rearrangement 58 4.3.8 Activation of peripheral B cells 89
3.10 The bursa of Fabricius, B-cell ontogeny, 4.3.9 Plasma cell development 90
and immunoglobulins 58 4.3.10 Cytokines in chicken B cell
3.10.1 Bursal development 58 development and activation 91
3.10.2 Formation of the bursal 4.3.11 Application of B cell cultures 92
epithelial anlage 58 References 93
3.10.3 Hematopoietic colonization
of the bursal rudiment and 5. Structure and evolution of avian
follicle bud formation 60
3.10.4 Development of the follicle-
immunoglobulins 101
associated epithelium and the Sonja Härtle, Katharine E. Magor,
follicular cortex 62 Thomas W. Göbel, Fred Davison and
3.10.5 Immunoglobulins 63 Bernd Kaspers
3.11 Lymphocyte-differentiating 5.1 The basic structure of immunoglobulins 101
hormones 63 5.2 Avian immunoglobulins 102
3.12 Development of the immune 5.2.1 Avian IgM 102
responses 64 5.2.2 Avian IgY (IgG) 103
3.12.1 Early immune responses 64 5.2.3 Avian IgA 104
3.12.2 Antibody isotype switching 5.2.4 Avian homologues of IgD
and hypersensitivity reaction 64 and IgE 105
3.12.3 Allograft rejection 64 5.2.5 L chains 105
3.13 Conclusion 64 5.2.6 Genomic organization of the
Acknowledgments 65 IgH and IgL locus 105
References 65 5.3 Ig half-life 107
Contents ix

5.4 Natural antibodies 107 7.7 Gene coevolution in the chicken


5.5 Maternal antibodies 108 major histocompatibility complex 142
5.6 Fc receptors 109 7.8 Other chicken genes important for
5.6.1 Chicken polymeric Ig receptor 109 the major histocompatibility
5.6.2 Chicken FcRn homologue 110 complex 144
5.6.3 Chicken Fc receptor cluster 110 7.9 Polymorphism and typing chicken
5.6.4 ggFcR 110 major histocompatibility complex
5.6.5 CHIR-AB1 110 genes 145
5.7 Avian antibody responses 110 7.10 Avian major histocompatibility
5.8 The chicken egg as a source of complexes 146
antibodies 112 7.11 Immunity, disease resistance, and the
5.8.1 Avian antibodies as tools for major histocompatibility complex in
research 112 wild birds 148
References 113 7.12 Sexual selection and the major
histocompatibility complex in wild
6. Avian T cells: Antigen Recognition birds 149
and Lineages 121 7.13 Origin and evolution of the immune
system 150
Adrian L. Smith and Thomas W. Göbel Acknowledgments 151
6.1 Introduction 121 References 151
6.2 T cell receptor structure and lineages 121
6.2.1 Somatic DNA recombination 121 8. Introduction to the avian innate
6.2.2 Organization of the T cell
immune system; properties,
receptor clusters 123
6.3 CD3 signaling complex 125 effects, and integration with other
6.3.1 Mammalian CD3 125 parts of the immune system 163
6.3.2 Chicken CD3γ/δ and CD3ε 126 Thomas W. Göbel and Adrian L. Smith
6.3.3 ζζ homodimer 126
6.3.4 T cell receptor complex—structural
8.1 Macrophages and dendritic cells 167
models 126
6.3.5 T cell receptor signal transduction 127 Kate Sutton, Adam Balic, Bernd Kaspers and
6.4 CD4 and CD8 127 Lonneke Vervelde
6.5 Costimulatory molecules 128 8.1.1 Introduction 167
6.6 T cell lineages 129 8.1.1.1 Antigen presentation 167
6.7 Methods to study T cell function 130 8.1.1.2 Dendritic cells 168
6.8 Perspectives 130 8.1.1.3 Macrophages 169
References 131 8.1.1.4 Development of myeloid
cells 171
7. The avian major histocompatibility 8.1.1.5 Sources of avian macrophages
and dendritic cells 172
complex 135
8.1.1.6 Avian myeloid cell lines 175
Jim Kaufman 8.1.1.7 Cell surface markers for
7.1 Introduction 135 avian myeloid cells 175
7.2 The biology of the major 8.1.1.8 Characterization of
histocompatibility complex 135 macrophages and DC in
7.3 The major histocompatibility complex: tissue sections 178
a genomic region or a biological unit? 136 8.1.1.9 Functional properties of
7.4 The chicken major histocompatibility chicken macrophages 178
complex and the major histocompatibility 8.1.1.10 Macrophage migration 178
complex syntenic region 137 8.1.1.11 Phagocytosis 179
7.5 Classical and nonclassical major 8.1.1.12 Respiratory burst activity 180
histocompatibility complex molecules 139 8.1.1.13 Nitric oxide production: a
7.6 Chicken classical major readout system for avian
histocompatibility complex molecules 140 macrophage activation 180
x Contents

8.1.1.14 Cytokine response of avian 8.5.1 Soluble components 217


macrophages 182 8.5.1.1 Host defense peptides 217
8.1.2 Functional properties of chicken 8.5.1.2 Collagenous lectins 219
antigen-presenting cells 183 8.5.1.3 Surfactant protein A and cLL 219
8.1.2.1 Maturation from antigen 8.5.1.4 Mannose-binding lectin 220
sampling to antigen 8.5.1.5 Collectin 10, -11, and -12 220
presenting 183 8.5.1.6 Complement 221
8.1.2.2 Migration 184 8.5.1.7 Components of the classical
8.1.2.3 Other nonmyeloid pathway 222
antigen-presenting cells 185 8.5.1.8 Components of the lectin
8.1.3 Concluding remarks 185 pathway 222
References 186 8.5.1.9 Components of the
alternative pathway 222
8.2 Avian granulocytes 197 8.5.1.10 Downstream components of
complement 223
Michael H. Kogut
8.5.2 The acute-phase response 223
8.2.1 Functional activities of heterophils 197
8.5.2.1 C-reactive protein 223
8.2.2 Receptors 198
8.5.2.2 Serum amyloid A 224
8.2.3 Other innate immune receptors 199
8.5.2.3 α1-acid glycoprotein 224
8.2.4 Genetic effects on heterophil
8.5.2.4 (Ovo)transferrin 224
genotype and phenotype 200
8.5.2.5 PIT54 225
8.2.5 Heterophil isolation 201
8.5.2.6 Hemopexin 225
References 201
8.5.2.7 Ceruloplasmin 225
Further reading 203
8.5.2.8 Fibrinogen 225
8.5.2.9 Other potential chicken APPs 225
8.3 Thrombocyte functions in the References 226
avian immune system 205
Jake Astill, R. Darren Wood and Shayan Sharif 8.6 Pattern recognition receptors 231
8.3.1 Introduction 205 Adrian L. Smith and Steven R. Fiddaman
8.3.2 Avian thrombocyte structure 205 8.6.1 Introduction 231
8.3.2.1 Physical characteristics 205 8.6.2 Tissue fluid and secreted pattern
8.3.2.2 Surface protein expression 206 recognition receptors 232
8.3.3 Avian thrombocytes and immune 8.6.2.1 C-reactive protein 232
responses 208 8.6.2.2 Collectins 232
8.3.3.1 Innate responses 208 8.6.2.3 Mannose-binding lectin 232
8.3.3.2 Adaptive immune responses 209 8.6.2.4 Ficolins 233
8.3.4 Infection of thrombocytes 210 8.6.2.5 Surfactants: surfactant
8.3.5 Conclusion 210 protein A and surfactant
References 210 protein D 233
8.6.2.6 Other collectins 233
8.4 Natural killer cells 213 8.6.2.7 Chicken mannose (or mannan)-
Thomas W. Göbel binding lectin-associated serine
8.4.1 Potential natural killer cell receptor protease proteins, linking soluble
families 213 pattern recognition receptor to
8.4.2 Phenotype of chicken natural killer complement activation 234
cells 214 8.6.3 Cell-associated pattern recognition
8.4.3 Natural killer cell function 214 receptors 234
References 215 8.6.3.1 Avian Toll-like receptors 234
8.6.3.2 TLR1/6/10-related molecules 235
8.6.3.3 TLR2 235
8.5 Soluble components and
8.6.3.4 TLR3 235
acute-phase proteins 217
8.6.3.5 TLR4 236
Edwin J.A. Veldhuizen and Tina Sørensen Dalgaard 8.6.3.6 TLR5 236
Contents xi

8.6.3.7 TLR7 and TLR8 236 9.6.4 Cytokines and factors in other
8.6.3.8 The absence of TLR9 237 birds 261
8.6.3.9 Avian Toll-like receptor without 9.7 Chemokines 263
mammalian orthologues: 9.7.1 XC and CX3C chemokines 264
chTLR15 and chTLR21 237 9.7.2 CC Chemokines 264
8.6.3.10 Toll-like receptor signaling 9.7.3 CXC chemokines 265
pathways in chickens 238 9.8 Cytokine and chemokine receptors 265
8.6.3.11 Genetic diversity and 9.8.1 Type I receptors 266
evidence of selection in 9.8.2 Type II receptors 266
avian Toll-like receptors 238 9.8.3 Transforming growth factor-β
8.6.3.12 Other transmembrane family receptors 267
pattern recognition receptor 239 9.8.4 Tumor necrosis factor
8.6.4 Cytosolic pattern recognition superfamily receptors 267
receptor 240 9.8.5 Chemokine receptors 267
8.6.4.1 Nucleotide-binding 9.8.6 Interleukin-1 family receptors 267
oligomerization domain-like 9.9 The importance of regulation of
receptors 240 cytokine responses 267
8.6.4.2 Retinoic acid-inducible 9.10 Therapeutic potential of chicken
gene-like receptors 240 cytokines 268
8.6.5 Closing comments: general 9.10.1 Alternatives to antibiotic
considerations in pattern recognition 241 growth promoters 268
Acknowledgments 241 9.10.2 Potential use of cytokines as
References 242 vaccine adjuvants 269
9.11 Conclusion 270
9. Avian cytokines and their References 270
receptors 249
Andrew G.D. Bean and John W. Lowenthal
9.1 Introduction 249
10. Immunogenetics and the mapping
9.2 Avian cytokine and chemokine
families 250
of immunological functions 277
9.3 The interleukins 250 Susan J. Lamont, Jack C.M. Dekkers,
9.3.1 The interleukin-1 family 250 Anna Wolc and Huaijun Zhou
9.3.2 T-cell proliferative interleukins 251 10.1 Introduction 277
9.3.3 T-helper interleukins 252 10.2 Genetics and immunological traits
9.3.4 Th1 interleukins 254 in the chicken 277
9.3.5 Th2 interleukins 254 10.3 Key gene loci for immunological traits 279
9.3.6 Th1 Th2 paradigm 255 10.4 Detecting quantitative trait loci 280
9.3.7 Other Th subsets 255 10.4.1 Linkage disequilibrium 281
9.4 Other interleukins 257 10.4.2 Experimental designs to detect
9.4.1 The interleukin-10 family 257 quantitative trait loci 281
9.4.2 The interleukin-6 family 258 10.5 Statistical procedures for quantitative
9.4.3 Other interleukins 259 trait loci detection 283
9.5 The interferons 259 10.6 Strategies to use molecular data in
9.5.1 Type I interferon 260 genetic selection 285
9.5.2 Type II interferon 260 10.6.1 Marker-assisted selection 285
9.5.3 Type III interferon 260 10.6.2 Whole-genome prediction 285
9.6 Other factors 260 10.7 Systems biology 286
9.6.1 The transforming growth 10.8 Transgenic animals 288
factor-β family 260 10.9 Future directions for systems
9.6.2 The tumor necrosis factor biology in avian immunology 289
superfamily 261 Acknowledgments 290
9.6.3 Colony-stimulating factors 261 References 290
xii Contents

11. The mucosal immune system 299 11.2.7 The immune system in the
parabronchi 333
Bernd Kaspers and Karel A. Schat 11.2.8 The phagocytic system of the
References 300 respiratory tract 334
11.2.9 Handling of particles in the
11.1 The avian enteric immune respiratory tract 335
system in health and disease 303 11.2.10 The secretory IgA system in the
Adrian L. Smith, Claire Powers and Richard Beal respiratory tract 335
11.1.1 General considerations 303 11.2.11 Gene expression analysis as a tool
11.1.2 Gut structure and immune to investigate host pathogen
compartments 304 interaction 336
11.1.2.1 Chicken gut-associated References 337
lymphoid tissue structures 305
11.1.2.2 Cellular composition of the 11.3 The avian reproductive immune
avian gut-associated system 343
lymphoid tissues 306 Paul Wigley, Paul Barrow and Karel A. Schat
11.1.2.3 The enterocyte as part of 11.3.1 Introduction 343
an integrated gut immune 11.3.2 The structure and function of the
system 306 avian reproductive tract 343
11.1.3 Development of the enteric 11.3.3 Structure and development of the
immune system 307 reproductive tract-associated
11.1.3.1 Development of immune immune system in the chicken 344
responses to model antigens 309 11.3.3.1 Organization of
11.1.3.2 Immunity to enteric lymphocytes in the
pathogens 309 reproductive tract 344
11.1.3.3 Development of immunity 11.3.3.2 Distribution of macrophages
to enteric pathogens 309 and other cells 344
11.1.3.4 Maternal antibody and 11.3.4 Local and systemic changes to the
protection of the young immune system at the onset of
chick 310 sexual maturity in hens 344
11.1.4 Viral infections of the gut 310 11.3.5 The innate immune system and the
11.1.5 Bacterial infections of the gut 311 reproductive tract 346
11.1.5.1 Salmonella 311 11.3.6 The reproductive tract immune
11.1.5.2 Campylobacter 313 system in infection 346
11.1.5.3 Necrotic enteritis 314 11.3.6.1 Bacterial infections of the
11.1.6 Parasitic infections of the gut 314 reproductive tract 346
11.1.6.1 Eimeria spp 315 11.3.6.2 The immune response to
11.1.6.2 Other parasitic infections 316 Salmonella infection of the
11.1.7 Concluding remarks 317 reproductive tract 346
Acknowledgments 317 11.3.6.3 Responses to vaccination
References 317 in the reproductive tract 348
11.3.6.4 The chicken as a model-
11.2 The avian respiratory immune understanding immunity in
system 327 ovarian cancer 349
11.3.6.5 What do we need to
Sonja Härtle, Lonneke Vervelde and know—directions for
Bernd Kaspers future research? 349
11.2.1 Introduction 327 11.3.6.6 What are the functions
11.2.2 Anatomy of the respiratory tract 327 and phenotypes of the cells
11.2.3 The paraocular lymphoid tissue 329 in the reproductive tract? 349
11.2.4 Nasal-associated lymphoid tissue 330 11.3.6.7 How does the immune
11.2.5 The contribution of the trachea to tissue of the reproductive
respiratory tract immune responses 331 tract integrate with the rest
11.2.6 The bronchus-associated lymphoid of the immune system? 349
tissue 331 References 349
Contents xiii

12. Impact of the gut microbiota on 13.5.3 The allantoic sac 379
the immune system 353 13.6 Concluding remarks 380
References 380
Michael H. Kogut
12.1 Introduction to the microbiota and 14. Avian immunosuppressive
avian immune system 353
diseases and immune evasion 387
12.2 Microbiota, metagenome, and
microbiome 354 Karel A. Schat and Michael A. Skinner
12.3 GI tract and immune system of poultry 354 14.1 Introduction 387
12.3.1 Intestinal barrier system 354 14.2 Immunosuppression 387
12.4 Influence of the microbiota in 14.2.1 Introduction 387
immunity 355 14.2.2 Stress-induced
12.4.1 Germ-free chickens 355 immunosuppression 388
12.4.2 Antibiotic-treated chickens 356 14.2.3 Mycotoxin-induced
12.4.3 Fecal microbial transplants 356 immunosuppression 389
12.4.4 Layer-type chickens versus 14.2.4 Coccidia-induced
broiler chickens 356 immunosuppression 390
12.5 Gut microbiota immune system 14.2.5 Virus-induced
communication 356 immunosuppression 390
12.5.1 Components of the microbiota 357 14.3 Mechanisms of immunosuppression 399
12.5.2 Microbial metabolites 357 14.3.1 Corticosteroids and
12.5.3 Microbial epigenetic stress-induced
modifications 357 immunosuppression 399
12.6 Gut microbiota: immune homeostasis 358 14.3.2 Apoptosis, necroptosis, and
12.7 Gut microbiota: immune dysfunction: pyroptosis 399
dysbiosis and inflammation 358 14.3.3 Virus-induced changes in the
12.8 Managing the microbiome for regulation of immune responses 400
immune modulation 359 14.4 Immunoevasion 401
References 359 14.4.1 Introduction 401
14.4.2 Immunoevasion by viral
13. Innate defenses of the avian egg 365 proteases 401
14.4.3 Immunoevasion mechanisms of
Sophie Réhault-Godbert, Maxwell Hincke, avian coronaviruses 402
Rodrigo Guabiraba, Nicolas Guyot and
14.4.4 Immunoevasion mechanisms
Joel Gautron
of the avian herpesviruses 402
13.1 Introduction 365
14.4.5 Immunoevasion mechanism
13.2 Egg basic structures and their role
of the avian poxviruses 402
in innate defense 365
14.4.6 Immunoevasion mechanism
13.2.1 Physicochemical barriers 366
of the avian orthomyxoviruses 404
13.2.2 Antimicrobial molecules 371
14.4.7 Immunoevasion mechanism
13.3 Modification of egg structures during
of the avian paramyxoviruses 405
embryonic development 373
14.4.8 Immunoevasion mechanism
13.4 Embryonic immunity 374
of the avian reoviruses 405
13.4.1 Toll-like receptors 374
14.4.9 Immunoevasion mechanism
13.4.2 Macrophages 374
of the avian birnaviruses 406
13.4.3 Heterophils 374
14.5 Conclusions 406
13.4.4 Dendritic cells 375
References 406
13.4.5 T lymphocytes 375
13.4.6 Natural Killer cells 376
13.4.7 Cytokines and chemokines 376 15. Factors modulating the avian
13.5 Extraembryonic structures and innate immune system 419
immunity 376 Tina Sørensen Dalgaard, Johanna M.J. Rebel,
13.5.1 Amniotic sac 377 Cristiano Bortoluzzi and Michael H. Kogut
13.5.2 Yolk sac 378 15.1 Endocrine regulation of immunity 419
xiv Contents

15.1.1 Stress hormones: epinephrine, 16.4.3 Immunological mechanisms 450


norepinephrine, dopamine, 16.4.4 Summary 452
and glucosteroids 419 Acknowledgments 452
15.1.2 Sex hormones 420 References 452
15.1.3 Metabolic hormones: thyroid
hormone, growth hormone, 17. Tumors of the avian immune
and leptin 422 system 457
15.1.4 Environmentally responsive
hormones: melatonin 422 Venugopal Nair
15.2 Physiological states 423 17.1 Introduction 457
15.2.1 Temperature and housing as 17.2 Tumors of the immune system 457
immune modulators 423 17.2.1 Marek’s disease 457
15.3 Dietary effects on immunity 424 17.2.2 Avian leukosis 459
15.3.1 Contribution of the 17.2.3 Reticuloendotheliosis 460
microbiome 425 17.3 Oncogenic mechanisms of tumor
15.3.2 Immunomodulatory nutrients viruses 460
and feed additives 425 17.3.1 Oncogenic mechanisms of
15.3.3 Immunometabolism 427 retroviruses 461
15.4 Assessment of immunocompetence 428 17.3.2 Oncogenic mechanisms of
15.4.1 Functional activity of the DNA tumor viruses 461
immune response 428 17.4 Immune responses to oncogenic
References 429 viruses 461
17.4.1 Immune responses to
16. Autoimmune diseases of poultry 437 leukosis/sarcoma viruses 462
17.4.2 Immune responses to
Gisela F. Erf reticuloendotheliosis virus 462
16.1 General characteristics of autoimmune 17.4.3 Immune responses to Marek’s
diseases 437 disease virus 462
16.2 Autoimmune vitiligo in Smyth-line 17.5 Antitumor responses 463
chickens 440 17.6 Conclusion 464
16.2.1 Introduction 440
References 464
16.2.2 The Smyth line chicken model
for autoimmune vitiligo 440
16.2.3 Characteristics of the
18. Practical aspects of poultry
Smyth-line chicken 441 vaccination 469
16.2.4 Pigmentation and normal J.J. (Sjaak) de Wit and Enrique Montiel
melanocyte function 442 18.1 Introduction 469
16.2.5 Target cell defects 442 18.2 Vaccine types 469
16.2.6 Immunological mechanisms 443 18.2.1 Live vaccines 469
16.2.7 Environmental factors 445 18.2.2 Inactivated vaccines 470
16.2.8 Summary 446 18.2.3 Poultry vaccine adjuvants 471
16.3 Spontaneous autoimmune 18.3 Vaccine application 471
(Hashimoto’s) thyroiditis in 18.3.1 Mass application 471
obese-strain chickens 446 18.3.2 Individual applications 472
16.3.1 Introduction 446 18.4 Factors influencing vaccine
16.3.2 Development and responses 474
characteristics of OS chickens 447 18.4.1 Status of the immune system
16.3.3 Immunological mechanisms 447 at the time of vaccination 474
16.3.4 Target cell/organ defects 448 18.4.2 Maternally derived antibodies 474
16.3.5 Summary 449 18.4.3 Vaccine storage, preparation,
16.4 Scleroderma in UCD 200/206 chickens 449 and administration 475
16.4.1 Introduction 449 18.4.4 Age at vaccination 476
16.4.2 Development and characteristics 18.4.5 Duration of immunity 476
of the UCD 200/206 lines 450 18.4.6 Interference between vaccines 476
Contents xv

18.4.7 Time intervals between 20. Evolutionary and ecological


vaccinations 477 immunology 519
18.5 Immunosuppression 477
18.5.1 Stress and immunosuppression 477 Michal Vinkler, James S. Adelman and
18.5.2 Mycotoxins 478 Daniel R. Ardia
18.5.3 Immunosuppression by 20.1 Introduction 519
vaccines 478 20.2 Assessing immune function in
18.5.4 Influence of immunosuppression free-living birds 520
on vaccination 478 20.2.1 Single-time-point assays 520
18.6 Quality control of response to 20.2.2 Multiple-time-point assays 522
vaccination 479 20.3 Development of the immune
18.6.1 Serology 479 system in free-living birds 524
18.6.2 PCR/culture 480 20.3.1 Ontogeny 524
Acknowledgments 480 20.3.2 Parental transmission of
References 480 antibodies 525
20.4 Factors causing variation in immune
19. Comparative immunology of responses 526
20.4.1 Age-related variation 526
agricultural birds 489
20.4.2 Social environment 527
Ursula Schultz and Katharine E. Magor 20.4.3 Condition, nutrition and
19.1 Introduction 489 individual quality 528
19.2 Innate immunity 490 20.4.4 Seasonality/annual cycles 530
19.2.1 Toll-like receptors 490 20.4.5 Parasite exposure 531
19.2.2 Retinoic acid induced gene-I 20.4.6 Other factors with
(R)-like receptors 491 immunomodulating effects 531
19.2.3 Antimicrobial peptides 492 20.5 Molecular variation and evolution in
19.3 Cytokines 492 immune genes 532
19.3.1 Interferons 492 20.5.1 The major histocompatibility
19.3.2 Interleukins 493 complex 532
19.3.3 Tumor necrosis factor family 498 20.5.2 Innate immune genes 535
19.3.4 Th2 cytokines 499 20.6 Immune function as an evolving life
19.4 Chemokines 499 history trait 536
19.4.1 CXC chemokines 499 20.6.1 Costs of mounting immune
19.4.2 CC chemokines 500 responses 536
19.5 CCR7 500 20.6.2 Parasite-mediated natural
selection and immune
19.6 Cell surface antigens 500 function 539
19.6.1 Anti-chicken monoclonal 20.6.3 Links with male secondary
antibodies cross-reacting with characters 542
turkey, quail, and duck 20.7 Priorities for future research 544
leukocytes 500 Acknowledgment 545
19.6.2 Evidence for T and B cell References 545
populations in ducks 503
19.6.3 Antigens expressed on duck 21. Advances in genetic engineering
lymphocyte subsets 503
of the avian genome 559
19.6.4 C-type lectin immune
receptors 506 Benjamin Schusser and Timothy Doran
19.6.5 Surface immunoglobulin 506 21.1 Methods to manipulate the avian
19.6.6 Major histocompatibility genome 559
complex 506 21.1.1 Viral vectors 559
19.7 Secreted antibodies 507 21.1.2 Transposons 560
19.8 Cell lines 508 21.1.3 Direct in vivo transfection 561
Acknowledgments 509 21.1.4 Sperm transfection-assisted
References 509 gene editing 561
xvi Contents

21.1.5 Primordial germ cells 562 21.3 Genetically modified quails 568
21.2 Genetically modified chickens 563 References 569
21.2.1 Chicken models for
immunological research 564
Appendix 1: Genetic stocks for immunological
21.2.2 Disease-resistant chickens 567
research 573
21.2.3 Genetically engineered
Abbreviations 583
chickens for basic research
and agriculture 567 Index 589
List of contributors

James S. Adelman Department of Biological Sciences, Steven R. Fiddaman Department of Zoology, University
University of Memphis, Memphis, TN, United States of Oxford, Oxford, United Kingdom
Daniel R. Ardia Department of Biology, Franklin and Joel Gautron INRAE, University of Tours, BOA,
Marshall College, Lancaster, PA, United States Nouzilly, France
Jake Astill Department of Pathobiology, Ontario Thomas W. Göbel Department for Veterinary Sciences,
Veterinary College, University of Guelph, Guelph, Veterinary Immunology Study Group, University of
ON, Canada; Artemis Technologies Inc., Guelph, ON, Munich, Munich, Germany
Canada
Rodrigo Guabiraba INRAE, Université de Tours, ISP,
Adam Balic Division Infection and Immunity, The Nouzilly, France
Roslin Institute and R(D)SVS, University of
Nicolas Guyot INRAE, University of Tours, BOA,
Edinburgh, Midlothian, United Kingdom
Nouzilly, France
Paul Barrow The School of Veterinary Medicine and
Sonja Härtle Department for Veterinary Sciences,
Science, University of Nottingham, Sutton Bonington,
Veterinary Immunology Study Group, University of
Loughborough, United Kingdom
Munich, Munich, Germany
Richard Beal Ryedale School, Nawton, United Kingdom
Maxwell Hincke Department of Innovation in Medical
Andrew G.D. Bean CSIRO, Health & Biosecurity at the Education, University of Ottawa, Ottawa, ON,
Australian Centre for Disease Preparedness, Geelong, Canada; Department of Cellular and Molecular
VIC, Australia Medicine, University of Ottawa, Ottawa, ON, Canada;
Cristiano Bortoluzzi Southern Plains Agricultural Loire Valley Institute for Advanced Studies, Orléans
Research Center, USDA-ARS, College Station, TX, and Tours, France, with BOA–INRAE Centre Val de
United States Loire, Nouzilly, France
Tina Sørensen Dalgaard Department of Animal Science, Thierry Jaffredo Sorbonne Université, IBPS, CNRS
Aarhus University, Tjele, Denmark UMR7622, Inserm U 1156, Developmental Biology
Fred Davison Institute for Animal Health, Newbury, Laboratory, Paris, France
United Kingdom Bernd Kaspers Department for Veterinary Sciences,
J.J. (Sjaak) de Wit Royal GD, Deventer, The Veterinary Immunology Study Group, University of
Netherlands; Utrecht University, Utrecht, The Munich, Munich, Germany
Netherlands Jim Kaufman School of Biological Sciences, Institute for
Jack C.M. Dekkers Department of Animal Science, Iowa Immunology and Infection Research, Ashworth
State University, Ames, IA, United States Laboratories, University of Edinburgh, Edinburgh,
Timothy Doran Australian Animal Health Laboratory, United Kingdom; Department of Pathology, University
CSIRO Health and Biosecurity, Geelong, VIC, of Cambridge, Cambridge, United Kingdom
Australia Michael H. Kogut Southern Plains Agricultural Research
Dominique Dunon Sorbonne Université, IBPS, CNRS Center, USDA-ARS, College Station, TX, United
UMR7622, Inserm U 1156, Developmental Biology States
Laboratory, Paris, France Susan J. Lamont Department of Animal Science, Iowa
Gisela F. Erf University of Arkansas, Division of State University, Ames, IA, United States
Agriculture, Department of Poultry Science, John W. Lowenthal CSIRO, Infectious Diseases
Fayetteville, AR, United States Program, Geelong, VIC, Australia

xvii
xviii List of contributors

Katharine E. Magor Department of Biological Sciences, Adrian L. Smith Department of Zoology, Jenner
University of Alberta, Edmonton, AB, Canada Institute, Oxford, United Kingdom; University of
Enrique Montiel Anitox Corporation, Lawrenceville, GS, Oxford, Oxford, United Kingdom
United States Kate Sutton Division Infection and Immunity, The
Nándor Nagy Department of Anatomy, Histology and Roslin Institute and R(D)SVS, University of
Embryology, Faculty of Medicine, Semmelweis Edinburgh, Midlothian, United Kingdom
University, Budapest, Hungary Edwin J.A. Veldhuizen Department of Biomolecular
Venugopal Nair Viral Oncogenesis Group, Pirbright Health Sciences, Division Infectious Diseases &
Institute, Woking, United Kingdom Immunology, Faculty of Veterinary Medicine, Utrecht
University, Utrecht, The Netherlands
Imre Oláh Department of Anatomy, Histology and
Embryology, Faculty of Medicine, Semmelweis Lonneke Vervelde Division Infection and Immunity, The
University, Budapest, Hungary Roslin Institute and R(D)SVS, University of
Edinburgh, Midlothian, United Kingdom
Claire Powers University of Oxford, Oxford, United
Kingdom Michal Vinkler Department of Zoology, Faculty of
Science, Charles University, Prague, Czech
Michael J.H. Ratcliffe Department of Immunology,
Republic
University of Toronto, Toronto, ON, Canada
Paul Wigley Department of Infection and Microbiome,
Johanna M.J. Rebel Department of Animal Health and
Institute of Infection Veterinary and Ecological
Welfare, Wageningen University & Research,
Sciences, University of Liverpool, Leahurst, Neston,
Wageningen, The Netherlands
United Kingdom
Sophie Réhault-Godbert INRAE, University of Tours,
Anna Wolc Department of Animal Science, Iowa State
BOA, Nouzilly, France
University, Ames, IA, United States; Hy-Line
Catherine Robin Hubrecht Institute, Royal Netherlands International, Dallas Center, IA, United States
Academy of Arts and Sciences (KNAW), University
R. Darren Wood Department of Pathobiology, Ontario
Medical Center Utrecht, Utrecht, The Netherlands;
Veterinary College, University of Guelph, Guelph,
Regenerative Medicine Center, University Medical
ON, Canada
Center Utrecht, Utrecht, The Netherlands
Laurent Yvernogeau Hubrecht Institute, Royal
Karel A. Schat Department of Microbiology and
Netherlands Academy of Arts and Sciences (KNAW),
Immunology, College of Veterinary Medicine, Cornell
University Medical Center Utrecht, Utrecht, The
University, Ithaca, NY, United States
Netherlands; Sorbonne Université, IBPS, CNRS
Ursula Schultz CellGenix GmbH, Freiburg, Germany UMR7622, Inserm U 1156, Developmental Biology
Benjamin Schusser Reproductive Biotechnology, TUM Laboratory, Paris, France
School of Life Sciences, Freising, Germany Huaijun Zhou Department of Animal Science,
Shayan Sharif Department of Pathobiology, Ontario University of California-Davis, Davis, CA, United
Veterinary College, University of Guelph, Guelph, States
ON, Canada
Michael A. Skinner Section of Virology, Faculty of
Medicine, Imperial College London, London, United
Kingdom
Foreword

The fascinating world of avian immunology might be best lymph node development, which opens an array of
surmised by the quote from Dr. Jim Kauffman, who stated antigen-processing questions for immune recognition and
with obvious eloquence “chickens are not mice with stimulation. Birds also lack several Toll-like receptors
feathers.” The point being that while many immunologi- found in other vertebras, yet share many that recognize
cal attributes are shared between the species, many are similar molecular motifs. These are but two examples to
also different and that we as immunologists must work to highlight the differences, but there are many others.
discover them. Perhaps Winston Churchill, in 1939, was also speaking of
In the last two decades, there has been a great increase avian immunology when he said, “it is a riddle, wrapped
in our knowledge of avian immunology. Molecular immu- in a mystery, inside an enigma.”
nologists, utilizing new technologies, have expanded our It is impossible to discuss the context of this book
ability to determine and decipher what is encoded in the without acknowledging the current global COVID-19
avian genome for immunological defense. In addition, pandemic caused by the SARS-CoV-2 coronavirus. This
strategic funding capabilities provided primarily, but not relatively novel virus, without existing population immu-
exclusively, by the United States and EU have resulted in nity, has spread rapidly across transcontinental bound-
the development of a plethora of monoclonal antibodies aries. These events are reminiscent of reports in the early
against avian immunological proteins (e.g., cytokines and 1930s when chicks arrived at the veterinary laboratories
cell markers) through “toolbox” grants. These two driving of the agricultural college in Fargo, North Dakota. A new
forces have opened doors and windows in a field that has disease was raging across poultry farms in Minnesota and
historically struggled to provide reagents for intricate North Dakota resulting in sick birds that were undergoing
analysis. respiratory distress with high mortality. As researchers
This book arrives at a key time in our need for knowl- investigated the disease, it was eventually identified as a
edge to handle the ever-changing pressures for sustainable viral agent and termed infectious bronchitis virus (IBV)
solutions to poultry production issues on a global scale. based on disease characteristics. However, it was not until
The overall health of birds, in particular the immune sys- the advent of electron microscopy in the 1960s that
tem, is critical to feed an ever-growing human population. pathologists were able to get a visual look at the virus and
As an example, the loss of antibiotic treatment for poultry in 1964 identified “lollipop-shaped” protrusions at the end
flocks in most countries has created a need for immuno- of the spikes of the virus that resembled a halo of gas sur-
logical intervention strategies to enhance control against rounding the sun. The relevance of these observations is
bacterial pathogens. In addition to the applied aspect of notable in that approximately a year after these publica-
avian immunology, a comparative analysis, including tions, similar morphological characteristics were identi-
genomics, function, and evolution, is important to deter- fied in virus samples obtained from a boy undergoing
mine how and why the avian immune system works as it flu-like symptoms. Scientists utilized these morphological
does. similarities found in both avian and human samples to
The avian genome in general is regarded as condensed identify a new viral family, coronavirus.
in nature to mammalian species in terms of immune gene Parallels are also found in the immunological arena.
repertoire. Immunologically speaking, this translates in To combat the SAR-CoV-2 virus, in 2020, vaccines have
that birds have to do the same (protect the animal) with been developed and approved for use in humans. Of note
less. Despite this, the immune responses between birds is that two of the most used at this time in the United
and mammals display similar defining characteristics. States and EU are nucleic acid based on the messenger
Both species employ an innate and adaptive immune RNA sequence of the SARS spike (S) gene. The resulting
response to a variety of microbiological incursions from protein is the main coronavirus immunogen and contains
bacteria, viruses, protozoa, and parasites. However, differ- epitopes for inducing neutralizing antibodies. The rele-
ences also stand out. For example, chickens lack draining vance to avian immunology is that over 20 years ago we

xix
xx Foreword

and others reported on the development and use of DNA This book brings together highly distinguished and
vaccines expressing the S1 from IBV in chickens, which internationally representative authors who are specialists
provided protective efficacy and induced both antibodies in their respective fields of avian immunology. This book
and T-cell responses. will be valued by bench scientists, graduate students,
The future of avian immunology is a bright one. poultry veterinarians, commercial industry researchers,
Knowledge gleaned from the release of the avian genome, and avian ecologists as an “immunological-bible” for a
including conservation of synteny, has already resulted in better understanding of avian immunology.
increased scrutiny of what is and is not available for
Darrell R. Kapczynski
defense. Functional studies, such as epitope binding of
U.S. Department of Agriculture, Agricultural Research
avian MHCs, are shedding new light and possibilities for
Service, U.S. National Poultry Research Center, Athens,
enhanced immune responses. Research in disease resis-
GA, United States
tance breeding is being accelerated by the development of
transgenic technologies, including CRISPR, that allow for
manipulation and testing in a more timelier manner. As
these resources are brought to bear, past barriers in the
field of avian immunology will fall.
Acknowledgments

Gathering this wealth of information would not have been The editors also thank Dr. Sonja Härtle for her edito-
possible without the commitment, dedication, and gener- rial work on numerous chapters and Dr. Ton Schat for the
ous participation of the large number of contributors to pictures of birds on the back cover.
the third edition of this book. The editors are indebted to The editors would also like to thank Susan Ikeda for
them for the considerable amount of work, their enthusi- her efforts to get this project accomplished despite the
asm and willingness to set aside other priorities to con- problems caused by the ongoing pandemic.
tribute to this volume. The editors would like to thank Dr.
Fred Davison for his contribution as the lead editor for
the first edition of Avian Immunology, setting the stage
for the subsequent editions.

xxi
Chapter 1

The importance of the avian immune


system and its unique features
Fred Davison
Institute for Animal Health, Newbury, United Kingdom

1.1 Introduction the 1950s and 1960s, when lymphocyte function became
an active subject for research [1]. The immunological sig-
The avian immune system provides an invaluable model nificance of lymphocytes emanated from some seminal
for studies on basic immunology. Birds and mammals studies carried out by Gowans, Chase and Mitchison,
evolved from a common reptilian ancestor more than 250 Simonsen, and their contemporaries [1,2]. In elegant
million years ago and have inherited many common immu- experiments with laboratory mammals and using cell trans-
nological systems. They also have developed a number of fers, these workers demonstrated that lymphocytes are
very different, and in some cases remarkable, strategies. essential for generating immune responses and retaining
Due to their economic importance, and the ready availabil- memory of previous exposure to an antigen. However, evi-
ity of inbred lines, most avian immunology research has dence that lymphocytes play such a key role in protection
involved the domestic chicken, Gallus gallus domesticus. against infection and in tumor rejection had, in fact, been
A remarkable consequence of this research has been the discovered almost 40 years earlier [3 6], though little
seminal contributions it has made to understand the funda- attention was paid to it [7]. This was almost certainly
mental immunological concepts, especially the complete because at the time they were made the observations could
separation of developing bursa- (B-) and thymus- (T-) not properly be explained and, possibly, because the exper-
dependent lymphocyte lineages. Some of these observa- imental animal involved was the chicken.
tions have been made by chance, while others have Between 1912 and 1921, James Murphy, an experi-
resulted from painstaking work which took advantage of mental pathologist working at the Rockefeller Institute for
special avian features, such as ease of access to, and pre- Medical Research in New York, performed a series of
cise timing of, all the stages in embryonic development. remarkable experiments using chickens and their embryos
Some of the avian findings were described before they to study the growth and rejection of tumor grafts. His
were recognized as important and subsequently explained experiments appeared to prove beyond question that the
in mainstream immunology. The story of avian immunol- lymphocyte is the active component in tissue graft rejec-
ogy is a fascinating one and by no means complete, as tion, in protection against infection and, by implication, in
there is still the need for explanations of a number of innate and acquired immune responses [7]. Murphy [4]
unique features and different strategies adopted by birds. In observed that fragments of rat tumors would not grow in
this chapter, some of the “firsts,” rightly attributed to avian the adult chicken, just as they did not grow in other spe-
immunology, are described and the importance of further cies (xenogenic rejection). However, they could be grown
studies in avian immunology is highlighted. on the chorioallantoic membrane (CAM) of developing
chick embryos, although only up to about 18 days incuba-
1.2 The contribution from avian tion. In older embryos tumor grafts were rejected, just as
they were if grafted onto the newly hatched chick or an
lymphocytes adult bird. Interestingly, Murphy [3] observed that the
The advent of modern cellular immunology, and the funda- grafts which grew on embryos could be transferred to
mental role that lymphocytes play, is generally credited to fresh embryos without any evidence of them being

Avian Immunology. DOI: https://doi.org/10.1016/B978-0-12-818708-1.00010-5


© 2022 Elsevier Ltd. All rights reserved. 1
2 Avian Immunology

altered. They also retained their tumorigenic capacity if understood how such lymphocytes could be the same as
regrafted onto a rat. Murphy [3] commented that these those cells involved in cell-mediated functions. The bursa
cellular changes occurring when living tissue is grafted of Fabricius, an obscure sac-like structure attached to the
onto an unsuitable host are the same, regardless of the proctodeal region of the bird’s cloaca (Fig. 1.1), played a
type of host resistance, be it natural resistance because of crucial role in unraveling this problem.
species differences (allogeneic or xenogeneic rejection) or The cloacal bursa, takes its name from Hieronymus
acquired immunity due to recovery from a tumor Fabricius of Aquapendente (1537 1619), is also known
implanted earlier. The histological picture consisted of as Girolamo Fabrizi d’ Acquapendente (Fig. 1.2). He was
edema surrounded by fibroplasia in the host tissue, the a professor of surgery at the University of Padua, Italy,
budding out of blood vessels, and infiltration of surround- from 1565 to 1613 [9] and by all accounts a brilliant anat-
ing host tissues with small lymphocytic cells. The omist, embryologist, and teacher. For his pioneering
inevitable consequence was that cells in the graft died work, he was later credited in Italian medical science as
fairly quickly leaving only a scar. These were quite pro- the “Father of Embryology.” Fabricius not only carried
found though, at the time, unappreciated observations. out dissections on human cadavers but also extended his
Murphy [4] also performed a series of elegant experi- anatomical studies to other species, providing the most
ments using adult chicken tissues cografted onto the beautiful, detailed drawings of his work. From his obser-
CAM with fragments of rat tumors. He observed that vations of avian anatomy, he surmised that the cloacal
chicken tissues containing an abundant supply of lympho- bursa, a hollow structure connected by a duct to the proc-
cytes, such as the spleen or bone marrow, caused tumor todeal region of the cloaca, most likely acts as a recepta-
grafts to be rejected, whereas these were not rejected if cle for storing donated semen.
the cografted tissue lacked a rich supply of lymphocytes.
“Since the sac is pervious, so that there is an open passage
Later on, Murphy [5] showed that after grafting fragments
from the anus to the uterus itself and another from the
of adult chicken spleen onto the CAM of a 7-day embryo,
uterus to the sac, that is, above and below, and since it is
the embryo’s own spleen became grossly enlarged
closed at the other end, I think it is the place into which the
(splenomegaly). This is the first published record of a
cock introduces and delivers semen so that it may be stored
graft versus host response (GvHR). Much later, it was
there” [4].
explained by Simonsen [8] as the immunologically com-
petent lymphocytes of the adult responding to mismatched This is not the case, however, but the role of the
major histocompatibility complex (MHC) molecules cloacal bursa continued to puzzle researchers over the fol-
expressed by the embryonic cells. The embryonic cells lowing 350 years. Some surmised that since the bursa
were recognized as foreign causing the adult cells to repli- of Fabricius regresses with sexual maturity, its size
cate and destroy the embryonic host’s lymphocytes. Graft having an inverse relationship with the size testes and
versus host disease, in which allogeneic bone marrow the adrenals, it must be some sort of endocrine or lym-
transplants recognize the tissues of the graft recipient and phoid gland associated with growth and sexual develop-
cause severe inflammatory disease, is a serious problem ment [10].
for immunosuppressed recipients receiving bone marrow Over the years many investigated the function(s) of the
transplants. The phenomenon became a major concern bursa including one young researcher, Bruce Glick, work-
with the introduction of human bone marrow transplanta- ing at the Poultry Science Department at Ohio State
tion. Nonetheless, the phenomenon was first described University, United States. Glick surgically removed the
using chick embryos as long ago as 1916. bursa from young chicks to investigate the effect on
growth. By chance, after one experiment was concluded, a
colleague, Timothy Chang, asked if he could use some of
1.3 Contribution of the bursa of Fabricius the birds for a class demonstration on antibody production.
Without doubt, the most significant contribution that stud- A group of the chickens was injected with Salmonella
ies on the avian system have made to development of spp. “O” antigen but 1 week later, when the class carried
mainstream immunology was delineating the two major out tests with blood and antigen, there was no evidence of
arms of the adaptive immune system. As already pointed agglutination. Chang, somewhat perplexed, reported the
out, in the 1960 the significance of lymphocytes was just failure to Glick who was able to identify the nonresponder
becoming appreciated, and it was generally accepted that chickens as those that had been bursectomized. At the time
there are two types of adaptive immune responses: they did not seem to fully appreciate the singular impor-
humoral responses involving antibodies and cellular tance of this finding [10] but were able to confirm their ini-
responses mediated by macrophages and lymphocytes. tial observations in further experiments and wrote up an
Since antibodies are produced by plasma cells, which article entitled: “The role of the bursa of Fabricius in anti-
themselves are derived from lymphocytes, it was not body production.” This was submitted to Science but
The importance of the avian immune system and its unique features Chapter | 1 3

FIGURE 1.1 A dissection showing the


positions of the major lymphoid tissues in
the chick. Insets provide details of: (A) the
seven thymic lobes lying alongside the left
jugular vein with the crop lying beneath;
(B) the spleen can be seen below the pro-
ventriculus, the dark-colored gall bladder
lies alongside; (C) cecal tonsils are found
at the proximal ends of the two blind cae-
ca, close to the ileo-cecal junction; (D) the
ovoid bursa of Fabricius is situated within
the pelvic girdle immediately dorsal to the
cloaca. From: Dr. Sven Reese, Institute for
Anatomy, Histology and Embryology, Faculty
of Veterinary Medicine, LMU Munich.

rejected on the grounds that further elucidation of the published their seminal paper on the delineation of the bur-
mechanisms was necessary before the article could be sal (B) and thymic (T) lymphoid systems in the chicken.
accepted for publication [10]. The article was subsequently These workers proposed that because of the similarities in
submitted to Poultry Science [11], where, for a time, it the lymphoid tissues and immune systems of birds and
failed to draw much attention. Several years passed before mammals, a mammalian equivalent for the bursa of
the significance of the work was properly appreciated and Fabricius must exist and provide a source of B-dependent
mainstream immunologists took an interest in the chicken’s lymphocytes to make antibodies. Later this bursa equiva-
immune system. It was eventually concluded that the avian lent was identified as bone marrow. The division of the
bursa must be essential for antibody-mediated immunity, adaptive immune system into B- and T-dependent compart-
whereas the thymus, which also undergoes involution dur- ments has remained a central tenet of immunological think-
ing sexual development, is necessary for cell-mediated ing ever since. The term B-lymphocyte is derived from
immunity [12 14]. Almost a decade after Glick and “bursa-derived lymphocyte” in honor of that peculiar avian
Chang’s initial observations [11], Cooper et al. [15] lymphoid structure which provided the original evidence.
4 Avian Immunology

together; diversity introduced at the joints between the


different gene segments by the variable addition and sub-
traction of nucleotides and, finally, (3) point mutations
introduced into the rearranged sequence diversifying these
region further (somatic hypermutation), allow for a vast
(B1011) antibody repertoire to be generated [16].
In contrast, the cluster of genes encoding the chicken
Ig light chain has only a single copy of the functional VL
and JL genes [18]. Hence, diversity due to VLJL joining is
very limited and the effects of VJ rearrangement are mini-
mal (a detailed description can be found in Chapter 4).
Likewise, with the Ig heavy chain locus, the presence of
single functional VH and JH genes means that little diver-
sity can be generated through VHDJH rearrangement.
However, clusters of pseudogenes, upstream of the heavy
and light chain Ig loci have a critical role in the genera-
tion of chicken antibody diversity. By a process known as
somatic gene conversion, VL and VH sequences are
replaced with pseudogene sequences. An enormous
amount of diversity is generated by substantial diversity
in the hypervariable regions of the donor V pseudogenes
FIGURE 1.2 Hieronymous Fabricius of Aquapendente, Professor of and somatic gene conversion events accumulate within
Surgery at the University of Padua from 1565 1613. Fabricius was the
first to describe the bursa, which is now known to be a primary lym-
single functional VL or VH genes. Perhaps, chickens rep-
phoid organ, attached by a duct to the proctodeum in birds and essential resent the extreme situation with only one functional VL
for the development of bursal-derived (B) lymphocytes and the avian gene, while other species such as the duck have up to four
antibody repertoire. functional VL genes, although they still use gene conver-
sion to introduce variability. It seems that, uniquely, birds
rely on somatic gene conversion for generating their anti-
1.3.1 Gene conversion and the bursa
body repertoire, which is the equal of that in immuno-
Antibodies recognize specific conformational molecular competent mammals. Interestingly, it has recently been
shapes on their targets through the immunoglobulin (Ig) shown that if gene conversion is blocked in chicken
molecule variable region. Antigenic shapes are legion so B cells then somatic hypermutation occurs instead [19].
an immunocompetent individual must be capable of gen- It has also been observed that gene conversion is not just
erating an antibody repertoire with a huge number of Ig limited to birds, it also occurs in rabbits [20] pigs, and
specificities [16]. Different B cells produce Ig molecules other mammalian species, though none appear to rely on
of different specificities, but each B cell is capable of pro- it as the exclusive means for generating the antibody
ducing only one Ig specificity. In man and mouse, the repertoire.
antibody repertoire of B cells is generated by a process The striking fact about avian somatic gene conversion
known as Ig gene rearrangement, which is ongoing is that it only occurs in the bursa of Fabricius (see
throughout life. In the case of the Ig light chain, genes Chapter 4). For instance, if the bursa is destroyed early in
that encode the variable region (VL gene segment), a join- development (60 hours), then those chicks that hatch pro-
ing region (JL gene segment), and the constant region (CL duce only nonspecific IgM and are unable to mount spe-
gene segment) are spatially separated by noncoding seg- cific antibody responses. In other words, they do not have
ments in germline DNA. Noncoding segments are spliced an antibody repertoire and are incapable of eliciting typi-
to allow the joining of VLJL regions in genomic DNA, cal responses or isotype switching to produce IgG or IgA.
while excision of the noncoding section in the RNA tran- However, if the bursa is removed much later during
script allows the VLJLCL regions to combine permitting embryonic development, but before 18 days when the B
translation of a functional Ig light chain molecule [17]. In lymphocytes have begun to migrate from the bursa into
the case of the Ig heavy chain, a further diversity (D) the peripheral lymphoid tissues, then the hatched chicks
gene is involved, making a VHDJH rearrangement, other- lack circulating Ig and are incapable of eliciting specific
wise the process is similar. Since multiple copies of V, J, antibody responses. We know that prebursal stem cells
and D genes exist in the mammalian Ig locus (1) random enter the bursa between 8 and 14 days incubation and
recombination of the different gene segments, (2) differ- have already undergone gene Ig rearrangement, probably
ent combinations of heavy and light chains paired in the embryonic spleen and bone marrow, for they
The importance of the avian immune system and its unique features Chapter | 1 5

express IgM on the surface (see Chapter 4). Within the evolved a number of methods for recognizing and
bursa, they undergo rapid rounds of cell division and only destroying cells with intracellular pathogens. These
within this unique environment gene conversion occurs. mechanisms allow the host’s effector lymphocytes to rec-
In the absence of the bursa, an antibody repertoire cannot ognize infected or neoplastic cells through the expression
be generated, and a major arm of the immune system of proteins that have been proteolyzed within the cell and
becomes nonfunctional. The chicken antibody repertoire are expressed on the cell surface as peptide fragments. In
is generated during the late embryonic stage and for a mammals, the molecules that present peptides on the sur-
short period after hatching. As the chick ages, its B cells face of cells are encoded by a highly polymorphic genetic
undergo additional rounds of somatic gene conversion region known as the MHC.
and the antibody repertoire becomes expanded until a The MHC region was originally recognized through
mature repertoire is achieved around 5 7 weeks when the its effects on tissue graft rejection and, of course, GvHR.
bursa is fully mature. Thereafter, the bursa begins to Two major types or classes of MHC molecules are
regress as sexual maturity approaches and the adult proba- encoded by genes in the mammalian MHC region and
bly relies on postbursal stem cells in the bone marrow as expressed on the surface of cells, class I and II MHC
the source of B cells. molecules (more fully described in Chapter 7). Although
Of course, generating the antibody repertoire in a burst both types of molecules are heterodimers: the class I
of activity in the young animal has its risks. Any pathogen MHC molecule consists of an alpha-chain encoded by the
that targets and destroys bursal cells will have a devastat- MHC together with an invariant β2-microglobulin mole-
ing effect on antibody-dependent immune responses. One cule from a gene outside of the MHC locus; the class II
such virus is the small RNA virus that causes infectious MHC molecule consists of two peptide chains (α and β)
bursal disease. Infection of the neonate chick with infec- whose genes are found in the MHC region. Unlike the
tious bursal disease virus (IBDV) may cause no clinical MHC class I heterodimer that is present on most types of
disease but destroys bursal B cells leaving the chick inca- cells, expression of class II MHC molecules is restricted
pable of mounting antibody responses to other pathogens, to antigen-presenting cells such as macrophages, den-
although paradoxically there is a good response to IBDV dritic, and B cells. During synthesis inside the cell, both
itself (see Chapter 14). The insidious nature of IBDV classes of MHC molecules trap peptide fragments in a
leaves the chick vulnerable to opportunistic infections and cleft on, what is to become, the outer surface of the extra-
unprotected by subsequent vaccinations. So, relying on cellular domain of the MHC heterodimer. On reaching the
the generation of the antibody repertoire in a single loca- cell surface, these peptides are displayed as peptide:MHC
tion and over a relatively short time span is not without complexes to signal T cells through their T-cell receptors.
danger and, perhaps, represents one of the more “risky” Since even the smallest virus produces a number of pro-
strategies birds have adopted. teins, and large pathogens can produce hundreds, there
needs to be a large repertoire of T-cell receptors
expressed by different T cells to recognize the multiplic-
1.4 The contribution of the chicken MHC ity of peptide fragments derived, not from the host’s own
Pathogens are diverse, cunning, and occupy different cells, but made by pathogens or neoplastic cells. This T-
niches within the body of the host. Apart from pathogens cell repertoire is developed within the thymus (described
that are found outside of cells, such as Clostridium, in Chapter 6), where developing T cells with receptors
Escherichia coli, and Bordatella, there are intracellular that recognize self-peptides associated with MHC mole-
pathogens that can be found in the cytoplasm or cellular cules are eliminated, or inactivated, before they mature to
vesicles. In addition, retroviruses and herpesviruses inte- prevent self-recognition and auto-immunity. Mature T
grate into the host’s own genome. To match this diversity, cells capable of recognizing “foreign” peptides are
and the different locations where pathogens are found, released from the thymus into the periphery and become
higher vertebrates have evolved a number of different activated if they recognize peptide fragments expressed
innate and adaptive immunological mechanisms to on MHC molecules. Interestingly, the T-cell repertoire in
improve the chances of survival. Antibodies recognize the chicken is developed in the thymus in a similar way
conformational epitopes on the pathogen’s molecules but to that of mammals. There is no evidence for a somatic
need to come into direct physical contact to neutralize a gene conversion mechanism such as occurs in the avian
pathogen, as well as recruit cells and other molecules that bursa.
bring about disposal. Ig molecules are large and cannot In mammals, the MHC is a large and complex region
easily enter a viable cell, so an intracellular pathogen can- that contains much redundancy [21]. In the human, it con-
not be recognized by an Ig molecule unless the patho- sists of about 4 million base pairs encoding at least 280
gen’s molecules are expressed on the surface of that genes. Separate regions contain several MHC class I and
infected cells. However, the cellular immune system has class II genes (there are a vast number of alleles) that are
6 Avian Immunology

highly expressed on cells. These regions are separated by heavily on vaccines to protect against a wide range of dif-
the third region that encodes immune response genes ferent infectious agents. Vaccinations are frequent and
(class III). Humans express two or three class I molecules begin from the day of hatching or even before. Chickens
and three or four class II molecules that are highly poly- are immunized with live-attenuated vaccines and killed
morphic. The high polymorphism is probably driven by vaccines delivered by various routes (injection, aerosol
the ever-changing variations in pathogens [22,23], spray, drinking water, etc.) in mass-vaccination programs
although different haplotypes appear to confer approxi- that dwarf such programs in human medicine.
mately the same degree of protection against most of the Every biology student knows that Edward Jenner is
infectious pathogens. In Chapter 7, Kaufman points out the founding father of vaccination. Jenner discovered that
that the associations between the human MHC and infec- cowpox pustules, obtained from an infected milkmaid,
tious disease are, actually, very slight. protected an 8-year-old boy, James Phipps, against the
The chicken MHC is known as the B locus since it related smallpox virus. However, further developments in
was first identified as a serological blood group locus [24] vaccination, and indeed the term vaccination, only came
encoding the polymorphic, and highly immunogenic BG into use about a century later, following studies by one of
antigen. This BG antigen is highly expressed on blood the greatest 19th-century scientists, Louis Pasteur. Here
cells and has no known mammalian equivalent. Later, it again the chicken had a privileged role and serendipity
was shown that that the B locus must constitute the avian played its part.
MHC, because of its strong association with cell- In 1878 Pasteur was investigating chicken cholera, a
mediated immune functions, such as graft rejection, disease with devastating effects, causing chickens to
mixed lymphocyte reactions, and GvHR. Remarkably, become anorexic, moribund, and usually leading to their
and in marked contrast to the large mammal MHC, the death. Pasteur investigated the causative agent, now
chicken B locus is minute, spanning only 92 kilobases known as the Pasteurella multocida, and succeeded in
and encoding 19 genes and making it approximately 20- growing the bacteria in culture. The story goes that
fold smaller than the human MHC [25]. Only two copies Pasteur’s research was interrupted by a holiday and a cul-
each of class I (BF) and class IIβ (B/Lβ) genes are found ture was left in a flask in the laboratory [29]. Upon
within the chicken B locus. The marked differences resuming his research, Pasteur inoculated chickens with
between the chicken MHC and its mammalian counterpart this stale culture. The chickens became sick but then
have led Jim Kaufman to argue that the chicken B locus recovered within a few days. We now know that the bac-
represents a minimal essential MHC that must have teria had become attenuated and no longer capable of
evolved after birds and mammals separated some 200 mil- causing mortality. Pasteur did not know this and decided
lion years ago. Another striking feature of the chicken to inject new chickens with a fresh bacterial culture but,
minimal MHC region is that not only does it affect a unfortunately (or fortunately!), his assistant found that
number of important cell-mediated immune functions but chickens at the local market were in short supply. A num-
also determines life or death in response to a number of ber of fresh birds were obtained but those that had recov-
pathogens [26 28]. In Chapter 7, Jim Kaufman develops ered from the inoculation with the stale culture needed to
the argument that chickens, with a minimal essential be reused. As expected, the new chickens all succumbed
MHC, appear to have adopted a completely different to the fresh pathogen and died but those that had recov-
strategy to that of mammals whose MHC is large and ered from the previous treatment with stale inoculum
complex. The close association between the chicken again recovered. Pasteur realized he had achieved with
MHC and disease resistance is fascinating and at first chicken cholera what Jenner had accomplished with
sight seems to be a suicidal strategy. However, many les- smallpox some 100 years earlier, only in this case he had
sons can be learned from studying avian immunology and attenuated (weakened) the pathogen by prolonged storage.
its parallel evolution. By investigating the minimal He called the attenuated culture a “vaccine” [30] in honor
chicken MHC, light should be thrown on the important of Edward Jenner and then began a, largely successful,
interactions between pathogens and the immune system search for similar vaccines against other infectious dis-
and the relevance of the different evolutionary strategies eases such as pig erysipelas, sheep anthrax, and rabies.
elucidated. The serendipitous discovery of attenuation was another
novel finding made using the chicken. Since then, the
development of vaccines has had far-reaching implica-
1.5 Contributions to vaccinology tions for the health and welfare of both humans and
In any review of the “firsts” credited to avian immunol- domesticated animals. The search for better, more effec-
ogy, tribute needs to be paid to pioneering developments tive vaccines still goes on apace.
in the practical uses of immunology, in other words the Another first in vaccine development was in the con-
use of vaccination. The modern poultry industry relies trol of Marek’s disease (MD) a naturally occurring
The importance of the avian immune system and its unique features Chapter | 1 7

neoplastic disease of chickens. MD became the scourge pressure of vaccine use. The risk, however, is that these
of the poultry industry in the 1950s and 1960s, causing hotter vaccines could themselves be capable of causing
major problems for animal health and welfare and becom- bursal damage and immunosuppression in chicks that are
ing a huge financial burden. Before the introduction of poorly protected by maternal antibodies or have a suscep-
MD vaccines, morbidity and mortality in laying flocks tible genotype. Here again, we have an example of a strat-
ranged from 0% 60% or greater, with losses of 30% egy that is holding at present but may not be sustainable
being common [31]. The development of an MD vaccine long term. More aggressive vaccines or vaccination
represents the first example of widespread use of vaccina- regimes cannot be introduced without the risk that the
tion to protect against a natural form of cancer [32]. Over vaccines themselves could be harmful.
the years it has been remarkably effective [33], although These issues have been addressed in epidemiological
not without problems. studies on implications of the use of vaccines on the evo-
MD was first described as a neurological disease lution of pathogen virulence. Researchers [38] were
(polyneuritis) by Josef Marek [34]. The condition caused chiefly concerned with the use of different vaccines
paralysis of the wings and legs and was associated with developed against the malaria parasite and its implications
mononuclear infiltrations and enlargement of the major for human populations. Mathematical modeling was car-
nerves. Later it was observed [35,36] that in addition to ried out based on the premise that most vaccines are
lesions in the nerves and central nervous system, chickens imperfect and rarely provide full protection from disease.
also developed lymphoid tumors in several visceral tis- Using various models, these authors predicted that vac-
sues (visceral lymphomatosis) such as the ovary, liver, cines designed to reduce the growth and/or toxicity of
kidneys, adrenal, and muscles. With intensification of pathogens actually diminished selection pressure against
poultry production, the acute form of MD became a domi- more virulent pathogens. Consequently, subsequent patho-
nant feature. Although the introduction of MD vaccines in gen evolution may lead to higher levels of intrinsic viru-
the 1970s controlled the disease, in some countries pro- lence and hence more severe disease in unvaccinated
blems with vaccine breaks have continued to occur with individuals. Such evolution would tend to erode any
regularity and there is now good evidence that the causa- population-wide benefits, such that overall mortality
tive herpesvirus (MDV) has been able to evade vaccine- could increase with the level of vaccination coverage.
induced immune responses by evolving to greater viru- Interestingly, the authors found evidence of this phenome-
lence. Since the 1980s the response of the industry in non in the practical problems arising from MD vaccina-
some countries has been to introduce more aggressive tion. Current MD vaccines target viral replication and do
vaccine strategies, using “hotter” vaccines, such as not prevent infection with MDV, consistent with mathe-
CVI988, either alone or in combination with other MD matical modeling that predicts the evolution of pathogen
vaccines (bivalent or trivalent combinations). The most virulence. Yet again, evidence from work on the chicken
efficacious current MD vaccine CVI988 is derived from a has proved to be the pathfinder and pointed to important
serotype 1 MDV that is weakly oncogenic in genetically problems to take account of in vaccine design.
susceptible chickens and this has led some to raise the
important question [37]: where do we go if hypervirulent
1.5.1 Embryonic (in ovo) vaccination
MDV pathotypes evolve that can break through the pro-
tection of CVI988? The problems associated with challenge from virulent
MDV is not the only example of a poultry virus that MDV when chicks are moved into rearing quarters, the
has changed in response to the introduction of widespread vast numbers of chicks requiring vaccinations at the
use of vaccines. More virulent isolates of another lympho- hatchery, as well as the occasional failures caused by
tropic virus, IBDV, were isolated in the late 1980s. IBDV manual vaccination, has led to a search for new ways for
is a small double-stranded RNA virus that encodes only mass vaccination that can be done at an even earlier stage
five viral proteins. As already pointed out, IBDV targets than the day-old chick. Sharma and colleagues at the East
B lymphocytes in the bursa of young chicks causing no Lansing Regional Poultry Laboratory, United States dem-
clinical signs in neonates but causing clinical disease and onstrated that chick embryos could be successfully vacci-
some mortality in older chicks (see earlier). Chicks are nated against MDV at 17 18 days incubation [39,40].
protected by maternal antibodies derived via the yolk, but The automated INOVOJECT system, which allows the
it became clear in the late 1980s that the very virulent automated mass application of vaccines to large numbers
IBDV being isolated from outbreaks was capable of caus- of eggs (up to 50,000 eggs per hour, [41]), was developed
ing disease in the presence of high levels of maternal anti- and has been widely applied in the poultry industries of
bodies. The response of the industry has been to introduce some countries (for a fuller description see Chapter 18).
more aggressive (hotter) vaccines to protect against the In the United States, almost all broilers (over 7 billion per
more virulent IBDV strains that have evolved under the year) are vaccinated by this method. In ovo vaccination is
8 Avian Immunology

achieved by puncturing a small hole through the blunt [7] Silverstein AM. The lymphocyte in immunology: from James B.
end of the egg with an oblique pointed needle then pass- Murphy to James L. Gowans. Nat Immunol 2001;2:569571.
ing down a smaller needle to deliver a small amount (usu- [8] Simonsen M. The impact of the developing embryo and newborn
ally 50 μL) vaccine into the amniotic cavity. Since the animal of adult homologous cells. Acta Pathol Microbiol Scand
1957;40:480500.
amniotic fluid is imbibed prior to hatching the vaccine is
[9] Adelmann HB. The embryological treatises of Hieronymus
then taken up by the embryo. Interestingly, the reasons
Fabricius of Aquapendente. Ithaca, NY: Cornell University Press;
why in ovo vaccination, applied to such an immunologi- 1942.
cally immature individual, is so effective remains to be [10] Glick B. How it all began: the continuing story of the bursa
fully explained. Nevertheless, the finding that a higher of Fabricius. In: Toivanen A, Toivanen P, editors. Avian immu-
vertebrate can be protected against challenge early after nology: basis and practice, vol. 1. Boca Raton, FL: CRC Press;
hatching (birth) by vaccinating the embryo is quite revo- 1987. p. 17.
lutionary and its practical application in mass vaccination [11] Glick B, Chang TS, Jaap RG. The bursa of Fabricius and antibody
a great achievement. The immunological explanations production. Poult Sci 1956;35:224225.
will surely come in due course [12] Szenberg A, Warner NL. Dissociation of immunological respon-
siveness in fowls with a hormonally-arrested development of lym-
phoid tissue. Nature 1962;194:146.
[13] Warner NL, Szenberg A. Effect of neonatal thymectomy on the
1.6 Conclusion immune response in the chicken. Nature 1962;196:784785.
Compared to those of mouse and man the avian immune [14] Warner NL, Szenberg A, Burnett FM. The immunological role
of different lymphoid organs in the chicken. I. Dissociation
system may seem like a poor relation and yet it has pro-
of immunological responsiveness. Aust J Exp Biol Med 1962;
vided important insights into fundamental immunological
40:373388.
mechanisms and can claim a number of important “firsts”
[15] Cooper MD, Peterson RDA, Good RA. Delineation of the thymic
especially with regard to vaccinology. From the immuno- and bursal lymphoid systems in the chicken. Nature 1965;
logical viewpoint, the chicken is, perhaps, the best- 205:143146.
studied nonmammalian species. The publication of the [16] Murphy K, Weaver C. Janeway’s immunobiology: The immune
chicken genome [42] and recent developments in reverse system in health and disease. 9th (ed.) London: Garland Science;
genetic technology (see Chapter 21) provide the opportu- 2016.
nity for a “quantum shift” in the search for new chicken [17] Tonegawa S. Somatic generation of antibody diversity. Nature
genes and exciting possibilities for developing new tools 1983;302:575583.
and reagents to study immune responses and immunoge- [18] Reynard CA, Anquez V, Dahan A, Weill JC. A single rearrange-
ment event generates most of the chicken immunoglobulin light
netics. Interest in studying immune responses of other
chain diversity. Cell 1985;40:283291.
avian species is increasing, including species of wild
[19] Arakawa H, Hauschild J, Buerstedde JM. Requirement of
birds. Ecologists are now taking an interest in measuring activation-induced deaminase (AID) gene for immunoglobulin
immunocompetence and determining its importance as a gene conversion. Science 2002;209:13011306.
heritable trait for the survival, both of the individual and [20] Becker RS, Knight KL. Somatic diversification of the immuno-
the population. Avian immunology is a fascinating and globulin heavy chain VDJ genes: evidence for somatic gene con-
growing field and will surely provide new and exciting version in rabbits. Cell 1990;63:987997.
insights for mainstream immunology in the future. [21] Trowsdale J. “Both man and bird and beast”: comparative organi-
sation of the MHC genes. Immunogenetics 1995;41:117.
[22] Doherty P, Zinkernagel R. Enhanced immunological surveillance
in mice heterozygous at the H-2 gene complex. Nature 1975;
References 256:5052.
[1] Burnet M. Cellular immunology. Cambridge: Cambridge [23] Zinkernagel RM, Doherty P. MHC-restricted cytotoxic T cells:
University Press; 1971. studies on the biological role of polymorphic major transplantation
[2] Gowans J. The life history of lymphocytes. Br Med Bull antigens determining T cell restriction specificity, function and
1959;15:1053. responsiveness. Adv Immunol 1979;27:52177.
[3] Murphy JB. Studies in tissue specificity. II. The ultimate fate of [24] Briles WE, Mcgibbon WH, Irwin MR. On multiple alleles affect-
mammalian tissue implanted in the chick embryo. J Exp Med ing cellular antigens in the chicken. Genetics 1950;35:633640.
1914;19:181186. [25] Kaufman J, Milne S, Göbel TW, Walker BA, Jacob JP, Auffray
[4] Murphy JB. Studies in tissue specificity. III. Factors of resistance C, et al. The chicken B locus is a minimal essential major histo-
to heteroplastic tissue-grafting. J Exp Med 1914;19:513522. compatibility complex. Nature 1999;401:923925.
[5] Murphy JB. The effect of adult chicken organ graft on the chick [26] Kaufman J, Völk H, Wallny H-J. The “minimal essential MHC”
embryo. J Exp Med 1916;24:110. and an “unrecognized MHC”: two extremes in selection for poly-
[6] Murphy JB. The lymphocyte in resistance to tissue grafting, morphism. Immunol Rev 1995;143:6388.
malignant disease and tuberculous infection. Monogr Rockefeller [27] Kaufman J, Salomonsen J. The “minimal essential MHC”
Inst Med Res 1926;21:130. revisited: both peptide-binding and cell surface expression level
Another random document with
no related content on Scribd:
compliance. To SEND DONATIONS or determine the status of
compliance for any particular state visit www.gutenberg.org/donate.

While we cannot and do not solicit contributions from states where


we have not met the solicitation requirements, we know of no
prohibition against accepting unsolicited donations from donors in
such states who approach us with offers to donate.

International donations are gratefully accepted, but we cannot make


any statements concerning tax treatment of donations received from
outside the United States. U.S. laws alone swamp our small staff.

Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.

Section 5. General Information About Project


Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could be
freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose network of
volunteer support.

Project Gutenberg™ eBooks are often created from several printed


editions, all of which are confirmed as not protected by copyright in
the U.S. unless a copyright notice is included. Thus, we do not
necessarily keep eBooks in compliance with any particular paper
edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg Literary
Archive Foundation, how to help produce our new eBooks, and how
to subscribe to our email newsletter to hear about new eBooks.

You might also like