You are on page 1of 67

Allergy Essentials, 2nd Edition - eBook

PDF
Visit to download the full and correct content document:
https://ebooksecure.com/download/allergy-essentials-2nd-edition-ebook-pdf/
Any screen.
Any time.
Anywhere.
Activate the eBook version
of this title at no additional charge.

Elsevier eBooks for Practicing Clinicians gives you the power to browse and search
content, view enhanced images, highlight and take notes—both online and offline.

Unlock your eBook today.


1. Visit expertconsult.inkling.com/redeem
2. Scratch box below to reveal your code
3. Type code into “Enter Code” box
4. Click “Redeem”
5. Log in or Sign up
6. Go to “My Library”

It’s that easy!


Place Peel Off
Sticker Here

For technical assistance:


email expertconsult.help@elsevier.com
call 1-800-401-9962 (inside the US)
call +1-314-447-8300 (outside the US)
Use of the current edition of the electronic version of this book (eBook) is subject to the terms of the nontransferable, limited license granted on
expertconsult.inkling.com. Access to the eBook is limited to the first individual who redeems the PIN, located on the inside cover of this book,
at expertconsult.inkling.com and may not be transferred to another party by resale, lending, or other means.
2020_PC
ALLERGY
ESSENTIALS
This page intentionally left blank
ALLERGY
ESSENTIALS
SECOND EDITION
Robyn E. O’Hehir, MD, PhD, FAHMS Gurjit K. Khurana Hershey, MD, PhD
Professor of Allergy, Immunology, and Kindervelt Endowed Chair in Asthma
Respiratory Medicine Research
Monash University and Alfred Hospital Professor of Pediatrics
Melbourne Director, Division of Asthma Research
Australia Cincinnati Children’s Hospital Medical Center
Director, Medical Scientist Training Program
University of Cincinnati College of Medicine
Stephen T. Holgate, MD, FMedSci Cincinnati, Ohio
Clinical Professor of Immunopharmacology United States
Medical Faculty in Clinical and Experimental
Sciences Aziz Sheikh, MD, FMedSci
Southampton General Hospital Professor of Primary Care Research and
Southampton Development Director and Dean of Data
United Kingdom Usher Institute
University of Edinburgh
Edinburgh
United Kingdom
Elsevier
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

ALLERGY ESSENTIALS, SECOND EDITION ISBN: 978-0-323-80912-2

Copyright © 2022 by Elsevier, Inc. All rights reserved.

Previous edition copyrighted 2017.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechani-
cal, 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 permis-
sions 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. Because of rapid advances in
the medical sciences, in particular, independent verification of diagnoses and drug dosages should be made.
To the fullest extent of the law, no responsibility is assumed by Elsevier, authors, editors or contributors 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.

Library of Congress Control Number: 2021942482

Content Strategist: Robin Carter


Content Development Manager: Meghan Andress
Publishing Services Manager: Deepthi Unni
Project Manager: Janish Paul
Design Direction: Patrick Ferguson

Printed in the United States

Last digit is the print number: 9 8 7 6 5 4 3 2 1


CONTRIBUTORS

Cezmi A. Akdis, MD Jonathan Corren, MD Stephen T. Holgate, MD, FMedSci


Professor and Director, Swiss Institute of Associate Clinical Professor of Medicine, Professor,
Allergy and Asthma Research (SIAF), Medicine and Pediatrics, Southampton School of Medicine,
University of Zürich, Zürich, Switzerland; David Geffen School of Medicine at UCLA, Southampton,
Professor, Christine Kühne Center for Los Angeles, CA, United States Hampshire, United Kingdom
Allergy Research and Education
(CK-CARE), Adnan Custovic, MD, PhD, FMedSci Lukas Joerg, MD
Davos, Switzerland Professor of Paediatric Allergy, Department of Rheumatology,
National Heart and Lung Institute, Immunology and Allergology,
Mübeccel Akdis, MD, PhD Imperial College London, Inselspital;
Swiss Institute of Allergy and Asthma London, United Kingdom Bern University Hospital;
Research (SIAF), University of Bern,
University of Zürich, Zürich, Switzerland; Pascal Demoly, MD, PhD Bern, Switzerland
Christine Kühne Center for Allergy Research Département de Pneumologie et
and Education (CK-CARE), Addictologie,
Catherine Lemière, MD, MSc
Davos, Switzerland Hôpital Arnaud de Villeneuve,
Professor of Medicine,
University Hospital of Montpellier,
Department of Chest Medicine,
Fuad M. Baroody, MD, FACS Institut Desbrest d’Epidémiologie et de Santé
CIUSSS du Nord de l’île de Montréal-
Professor of Surgery, Publique UMR INSERM,
Hôpital du Sacré-Coeur de Montréal,
Section of Otolaryngology-Head and Neck Université de Montpellier,
Université de Montréal,
Surgery; Montpellier, France
Montreal, Canada
Professor of Pediatrics,
Director of Pediatric Otolaryngology; Luz Fonacier, MD
Director, Residency Program, Department of Medicine, Donald Y.M. Leung, MD, PhD
Otolaryngology-Head and Neck Surgery; NYU Langone Health, Department of Pediatrics,
The University of Chicago Medicine and The Mineola, NY, United States National Jewish Health,
Comer Children’s Hospital, Denver, CO, United States
Chicago, IL, United States David B.K. Golden, MD
Associate Professor of Medicine, Tesfaye B. Mersha, PhD
Mark Boguniewicz, MD Johns Hopkins University, Associate Professor,
Department of Pediatrics, Baltimore, MD, United States Pediatrics,
National Jewish Health, Cincinnati Children’s Hospital Medical
Denver, CO, United States Clive E.H. Grattan, MA, MD, FRCP Center,
St John’s Institute of Dermatology, Cincinnati, OH, United States
Simon G.A. Brown, MBBS, DA(UK), Guy’s Hospital,
PhD, FACEM London, Anna Nowak-Węgrzyn, MD, PhD
Centre for Clinical Research in Emergency Middlesex, United Kingdom Professor of Pediatrics,
Medicine, NYU Langone School of Medicine,
Harry Perkins Institute of Medical Research; Oliver V. Hausmann, MD New York, NY, United States
Royal Perth Hospital, ADR-AC GmbH,
University of Western Australia, Adverse Drug Reactions,
Perth, WA, Australia Analysis and Consulting, Robyn E. O'Hehir MD, PhD, FAHMS
Bern, Switzerland; Professor of Allergy, Immunology, and
A. Wesley Burks, MD Loewenpraxis und Klinik, Respiratory Medicine,
Curnen Distinguished Professor and Chair, St. Anna, Monash University and Alfred Hospital,
Pediatrics, Lucerne, Switzerland Melbourne, Australia
University of North Carolina,
Chapel Hill, NC, United States Gurjit K. Khurana Hershey, MD, PhD Clive Robinson, BSc, PhD
Kindervelt Endowed Chair in Asthma Professor of Respiratory Cell Science,
Anca-Mirela Chiriac, MD, PhD Research Institute for Infection & Immunity,
Département de Pneumologie et Professor of Pediatrics St George’s, University of London,
Addictologie, Director, Division of Asthma Research London, United Kingdom
Hôpital Arnaud de Villeneuve, Cincinnati Children’s Hospital Medical
University Hospital of Montpellier, Center Umit Sahiner
Institut Desbrest d’Epidémiologie et de Director, Medical Scientist Training Program Professor, Hacettepe University,
Santé Publique UMR INSERM, University of Cincinnati College of Medicine School of Medicine,
Université de Montpellier, Cincinnati, Ohio Department of Pediatric Allergy,
Montpellier, France United States Ankara, Turkey

v
vi Contributors

Sarbjit S. Saini Michael G. Sherenian, MD Hille Suojalehto, MD, PhD


Professor of Medicine, Assistant Professor, Adjunct Professor,
Johns Hopkins University School of Department of Pediatrics, Respiratory Medicine and Allergology
Medicine, Cincinnati Children’s Hospital Medical Center, Department,
Baltimore, MD, United States Cincinnati, OH, United States Finnish Institute of Occupational
Health,
Helen E. Smith, BMedSci BMBS, MSc DM Helsinki, Helsinki, Finland
Hugh A. Sampson, MD
Professor,
Professor of Pediatrics,
Lee Kong Chian School of Medicine,
Icahn School of Medicine at Paul J. Turner, FRACP, PhD, MRCPCH
Nanyang Technological University,
Mount Sinai, Section of Inflammation,
Novena, Singapore
New York, NY, United States Repair and Development,
Geoffrey A. Stewart, BSc, PhD National Heart & Lung Institute,
Aziz Sheikh, MD, FMedSci Emeritus Professor, Imperial College London,
Professor of Primary Care Research and School of Biomedical, Biomolecular and London, United Kingdom
Development Chemical Sciences,
Director and Dean of Data University of Western Australia;
Usher Institute Director,
University of Edinburgh, Institute for Respiratory Health,
Edinburgh, United Kingdom Perth, WA, Australia
C ONTENTS

1. Introduction to Mechanisms of Allergic Diseases, 1 9. Drug Allergy, 184


Umit Sahiner, Mübeccel Akdis, and Cezmi A. Akdis Oliver V. Hausmann and Lukas Joerg

2. Precision Medicine, 25 10. Ur ticaria, 202


Gurjit K. Khurana Hershey, Michael G. Sherenian, and Tesfaye Clive E.H. Grattan and Sarbjit S. Saini
B. Mersha
11. Atopic Dermatitis and Allergic Contact Dermatitis, 212
3. Epidemiology of Allergic Diseases, 40 Mark Boguniewicz, Luz Fonacier, and Donald Y.M. Leung
Adnan Custovic
12. Food Allergy and Gastrointestinal Syndromes, 240
4. Indoor and Outdoor Allergens and Pollutants, 56 Anna Nowak-We˛grzyn, A. Wesley Burks, and Hugh A. Sampson
Geoffrey A. Stewart and Clive Robinson
13. Anaphylaxis, 271
5. Principles of Allergy Diagnosis, 95 Paul J. Turner and Simon G.A. Brown
Anca-Mirela Chiriac and Pascal Demoly
14. Occupational Allergy, 283
6. Allergen-Specific Immunotherapy, 111 Catherine Lemière and Hille Suojalehto
Helen E. Smith, Aziz Sheikh, and Robyn E. O'Hehir
15. Insect Allergy, 294
7. Asthma, 123 David B.K. Golden
Stephen T. Holgate
Internet Resources, 306
8. Allergic Rhinitis and Conjunctivitis, 170
Jonathan Corren and Fuad M. Baroody Index, 308

vii
This page intentionally left blank
1
Introduction to Mechanisms
of Allergic Diseases
Umit Sahiner, Mübeccel Akdis, and
Cezmi A. Akdis

CHAPTER OUTLINE
Summary of Important Concepts, 1 Cytokines in Allergic Inflammation, 10
Introduction, 2 Interleukin-4 (IL-4), 10
Innate Immunity, 2 Interleukin-5 (IL-5), 11
Microbial Pattern Recognition by the Innate Immune Interleukin-9 (IL-9), 11
System, 2 Interleukin-13 (IL-13), 11
Pattern Recognition Receptors, 2 Interleukin-25 (IL-25), 11
Cellular Responses of Innate Immunity, 2 Interleukin-33 (IL-33), 11
Innate Instruction of Adaptive Immune Responses, 3 Interleukin-35 (IL-35), 11
Innate Immunity and Allergy, 4 Thymic Stromal Lymphopoietin (TSLP), 12
Adaptive Immunity, 4 Chemokines in Allergic Diseases, 12
Adaptive Immune Response in Allergic Disease, 4 Asthma, 12
Main Components of the Adaptive Immune System, 4 Atopic Dermatitis, 13
Features of the Adaptive Immune Response, 4 Biology of Immune Cells, 14
Mechanisms of Diseases Involving Adaptive T Lymphocytes, 14
Immunity, 5 B Lymphocytes, 15
Immunoglobulin Structure and Function, 5 Innate Lymphoid Cells, 15
B Lymphocytes and the Humoral Immune Response, 5 Dendritic Cells, 16
Immunoglobulin Structure and Gene Rearrangement, 6 Mast Cells, 16
Immunoglobulin Function, 6 Basophils, 17
Immunoglobulins and Human Disease, 6 Eosinophils, 17
Immune Tolerance, 6 Contribution of Structural Cells to Allergic
Introduction, 6 Inflammation, 18
Central and Peripheral Tolerance Mechanisms, 6 Airway Epithelial Cells, 18
Central Tolerance, 6 Airway Smooth Muscle Cells, 19
Peripheral Tolerance, 8 Neuronal Control of Airway Function, 19
Histamine Receptors in Peripheral Tolerance, 8 Cytokine Networks in Allergic Inflammation, 19
Immune Effector Cells and Molecules, 8 Microbiome and Immune System, 21
Treg Cells and Regulatory B cells, 8 Food Allergy as a Model for Allergic Diseases, 21
Transforming Growth Factor-β (TGF-β), 9 Resolution of Allergic Inflammation and Major
Interleukin-10 (IL-10), 9 Pathways, 21
Cytokines and Chemokines in Allergic Inflammation, 10 References, 22

• The innate immune system first responds to early infectious


SUMMARY OF IMPORTANT CONCEPTS and inflammatory signals, activating and instructing the adap-
• Allergic inflammation is a result of a complex interplay tive immune system for antigen-specific T and B lymphocyte
among structural tissue cells and inflammatory cells, includ- responses and the development of immunologic memory.
ing mast cells, basophils, lymphocytes, dendritic cells, eosin- • Allergen recognition and uptake, allergic sensitization, inflam-
ophils, and, sometimes, neutrophils. mation, and disease originate in the innate immune system.
• Cytokines are families of secreted proteins that mediate • Adaptive immune responses depend on activation of naive
immune and inflammatory reactions at local or distant sites. CD4+ T cells and differentiation into effector cells. CD4+ T
1
2 Allergy Essentials

helper type 2 (Th2) cells are critical mediators of allergic receptors (Table 1.1). Secreted PRRs typically have multiple
inflammation. effects in innate immunity and host defense, including direct
• Production of IgE antibody is regulated mainly by Th2 cells. microbial killing, serving as helper proteins for transmem-
Activated Th2 cells trigger IgE production in B cells through brane receptors, opsonization for phagocytosis, and chemoat-
a combination of signals, including secreted cytokine (inter- traction of innate and adaptive immune effector cells. AMPs
leukin [IL]-4 or IL-13) and cell surface (CD40L). are secreted PRRs that are microbicidal and rapidly acting.
• Better understanding of the pathophysiology of allergic When secreted onto skin and mucous membranes, they cre-
inflammation will enable us to identify novel therapeutic ate a microbicidal shield against microbial attachment and
targets in the treatment of chronic allergic inflammation. invasion.
Transmembrane PRRs are expressed on many innate immune
cell types, including macrophages, DCs, monocytes, and B lym-
INTRODUCTION phocytes (Fig. 1.1). These PRRs are exemplified by the Toll-like
The inflammatory process has several common characteristics receptors (TLRs) and their associated recognition, enhancing,
shared by various different allergic diseases, including asthma, and signal transduction proteins (Fig. 1.1). Innate immune
allergic rhinitis (AR) or rhinosinusitis, atopic dermatitis (AD) response at the epithelial cell–related and DC-related processes
(eczema), and food allergy. Allergic inflammation is charac- are controlled by the activation of the epithelial PRR by PAMPS
terized by IgE-dependent activation of mucosal mast cells and found in the microorganisms as well as the host-derived dam-
an infiltration of eosinophils that is orchestrated by increased age-associated molecular patterns (DAMPs). Airway epithelial
numbers of activated CD4+ T helper type 2 (Th2) lymphocytes. cells and DCs express a wide range of TLRs, NOD-like recep-
In addition to these cells, various types of inflammatory cells tors (NLRs), RIG-I-like receptors (RLRs), AIM2-like recep-
produce multiple inflammatory mediators, including lipids, tors (ALRs), C-type lectin receptors (CLRs), protease-activated
purines, cytokines, chemokines, and reactive oxygen species. receptors (PARs), and others.2,3
Both innate and adaptive immune mechanisms and involve-
ment of multiple cytokines and chemokines play roles. Cellular Responses of Innate Immunity
Microbial detection by PRRs activates the cells that express or
bind them. Those in frontline positions for detection are the
INNATE IMMUNITY first responders of the innate immune system, such as tissue
Innate immunity is an essential part of the immune system macrophages, fibrocytes, epithelial cells, and mast cells.
and is the first line of defense against microorganisms and Innate immune activation also leads to multifaceted anti-
foreign bodies such as allergens. It acts by the action of a lim- microbial responses by tissue infiltrating immune cells (e.g.,
ited number of receptors specific to microbial components. neutrophils, NK cells, DCs, monocytes). These responses are
As a result, both rapid immune response and activation of the potent antimicrobial effectors that usually are recruited by an
adaptive immune system occurs. Starting from body surfaces, innate immune intermediary to induce the full weight of their
epithelial cells, dendritic cells (DCs), natural killer (NK) cells, response, but they can respond directly to microbial stimuli
innate lymphoid cells (ILCs), macrophages, mast cells, eosino- through their own surface-expressed PRRs. On reaching the
phils, basophils, and neutrophils are main players for the innate infected site, neutrophils phagocytose invading microorganisms
immune response. Epithelial barrier and microbiome as well as that are opsonized by complement C3 fragments (e.g., C3b, iC3b)
physicochemical factors such as mucus, antimicrobial peptides and immunoglobulin G (IgG).4 Recruited and activated NK cells
(AMPs), ciliary movement, cough, and peristaltism all play a mediate antimicrobial activities by induction of apoptosis of cell
role in innate defense mechanisms.1 targets and cytokine secretion that promote innate immune func-
tions and contribute to adaptive immune responses.
DCs are transformed into active antigen-presenting cells
Microbial Pattern Recognition by the Innate (APCs) by stimulation of the TLR and they initiate and mediate
Immune System adaptive immune responses. Additionally, DCs together with
Microbial recognition by the innate immune system is mediated interferon (IFN)-γ can induce macrophage polarization, which
by germline-encoded receptors with genetically predetermined is important for phagocytosis.5 DCs in the blood can be divided
specificities for microbial constituents. Natural selection has into two groups as myeloid DCs (mDCs) and plasmacytoid
formed and refined the repertoire of innate immune receptors to DCs (pDCs). mDCs selectively express TLR2–6 and TLR2–8
recognize highly conserved molecular structures that distinguish and respond to bacterial and viral infections by producing large
large groups of microorganisms from the host. These microbe- amounts of interleukin (IL)-12. However, pDCs express TLR7
specific structures are called pathogen-associated molecular pat- and TLR9 associated with the endosome and produce type 1
terns (PAMPs), and the pattern recognition receptors (PRRs) of IFNs.6 The newly described cell type native lymphoid cells have
the innate immune system recognize these structures (Table 1.1). effector functions in homeostasis and inflammation. ILC1s
and ILC3s are essential for defense against infection by viruses,
Pattern Recognition Receptors intracellular bacteria, and parasites. However, ILC2s direct
PRRs of the innate immune system can be divided into two type 2 inflammation and mediate allergic inflammation, tissue
groups: secreted receptors and transmembrane signal-transducing repair, and anti-helminth innate immunity.5
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 3

TABLE 1.1 Innate Pattern Recognition Receptors in Humans


Pattern Recognition Receptors PAMP Structures Recognized Functions
Secreted
Antimicrobial peptides
α- and β-Defensins Microbial membranes (negatively charged) Opsonization, microbial cell lysis, immune cell chemoattractant
Cathelicidin (LL-37)
Dermcidin
RegIIIγ
Collectins
Mannose-binding lectin Microbial mannan Opsonization, complement activation, microbial cell lysis,
chemoattraction, phagocytosis
Surfactant proteins A and D Bacterial cell wall lipids; viral coat proteins Opsonization, killing, phagocytosis, proinflammatory and
antiinflammatory mediator release
Pentraxins
C-reactive protein Bacterial phospholipids (phosphorylcholine) Opsonization, complement activation, microbial cell lysis,
chemoattraction, phagocytosis

Secreted and membrane bound


CD14 Endotoxin TLR4 signaling
LPS binding protein Endotoxin TLR4 signaling
MD-2 Endotoxin TLR4 co-receptor

Membrane bound
Toll-like receptors Microbial PAMPs Immune cell activation
C-type lectin receptors
Mannose receptor (CD206) Microbial mannan Cell activation, phagocytosis, proinflammatory mediator release
DECTIN-1 β-1,3-Glucan Cell activation, phagocytosis, proinflammatory mediator release
DECTIN-2 Fungal mannose Cell activation, phagocytosis, proinflammatory mediator release
DC-SIGN Microbial mannose, fucose Immunoregulation, IL-10 production
Siglecs Sialic acid containing glycans Cell inhibition, endocytosis

Cytosolic
NOD-like receptors
NOD-1 Peptidoglycans from gram-negative bacteria Cell activation
NOD-2 Bacterial muramyl dipeptides Cell activation
NLRP1 Anthrax lethal toxin PAMP recognition in inflammasome
NLRP3 (cryopyrin) Microbial RNA PAMP recognition in inflammasome
NLRC4 Bacterial flagellin PAMP recognition in inflammasome
RIG-I and MDA5 Viral double-stranded RNA Type 1 IFN responses
DC-SIGN, Dendritic cell–specific intracellular adhesion molecule 3 (ICAM-3)–grabbing non-integrin; DECTIN, dendritic cell–specific receptor;
IFN, interferon; IL, interleukin; LPS, lipopolysaccharide; MD-2, myeloid differentiation factor 2 (also called lymphocyte antigen 96 [LY98]); MDA5,
melanoma differentiation-associated 5 (also called interferon induced with helicase domain 1 [IFIH1]); NLR, NOD-like receptor; NOD, nucleotide-
binding oligomerization domain protein; PAMP, pathogen-associated molecular pattern; RegIIIγ, regenerating islet-derived 3 γ (REG3G); RIG-I,
retinoic acid-inducible 1 (also called DDX58); Siglecs, sialic acid–binding immunoglobulin-like lectins; TLR, Toll-like receptor.

Innate Instruction of Adaptive Immune Responses Microbial pattern recognition by innate immune cells controls
The immediate and infiltrative responses of innate immunity acti- the activation of adaptive immune responses by directing micro-
vate and instruct the adaptive immune system for antigen-specific bial antigens linked to TLRs and other PRRs through the cellular
T and B lymphocyte responses and the development of immuno- processes leading to antigen presentation and the expression of
logic memory. Because the adaptive immune system essentially costimulatory molecules (e.g., CD80 with CD86). This two-step
has a limitless antigen receptor repertoire, instruction is necessary activation of the immune system, an innate immune response
to guide adaptive antimicrobial immune responses toward patho- first and then an adaptive immune response, prevents unneces-
gens and not self-antigens or harmless environmental antigens. sary inflammatory responses and is highly effective.7
4 Allergy Essentials

Macrophages Neutrophils
APC

α3
HLA class I

Toll-like Antimicrobial peptides CD8


receptors Collectins C-type lectin
receptors α2
β1

α1
Epithelial NOD-like
cells receptors TCR

T cell

C-reactive protein Mast cells Fig. 1.2 Interaction of a human leukocyte antigen (HLA) class I
molecule on an antigen-presenting cell (APC) with a CD8+ T cell.
Dendritic cells The antigen receptor (i.e., T cell receptor [TCR]) complex (purple)
Fig. 1.1 Main categories of pattern recognition receptors and the recognizes a combination of an antigen peptide (red) and an HLA
innate immune cell types that express them. NOD, Nucleotide- molecule (brown and pink). The CD8 molecule (aqua blue) in the T
binding oligomerization domain protein. (Adapted from Liu AH. cells interacts with the α3 domain of the HLA molecule. HLA class
Innate microbial sensors and their relevance to allergy. J Allergy II molecules present antigen peptides to CD4+ T cells in a similar
Clin Immunol. 2008;122:846-858.) manner, interacting with the TCR and the CD4 molecules.

Innate Immunity and Allergy


The innate immune system of the airways, gastrointestinal tract, uniquely reacting lymphocytes first explained the B cell origin
and skin is continuously exposed to potential allergens. As of antibody diversity and applies to cellular (T cell) immune
with microbial antigens, allergens can engage innate PRRs, are responses.
processed through innate immune cells, and can lead to patho-
logic allergic/inflammatory immune responses. Although the Main Components of the Adaptive Immune System
circumstances leading to allergic immunity in humans are not All cells of the immune system are derived from the pluripo-
clear, evidence suggests that allergic susceptibilities can origi- tent hematopoietic stem cell found in the bone marrow. This
nate in the innate immune system.8 pluripotential stem cell gives rise to lymphoid stem cells and
myeloid stem cells. The lymphoid progenitor cell differentiates
into three types of cell, T cell, B cell, and ILC and NK cell, and
ADAPTIVE IMMUNITY contributes to the development of subsets of DCs. The myeloid
stem cell gives rise to DCs, mast cells, basophils, neutrophils,
Adaptive Immune Response in Allergic Disease eosinophils, monocytes, and macrophages, as well as mega-
A remarkable property of the adaptive immune system is its karyocytes and erythrocytes. Differentiation of these commit-
memory. Immunologic memory is made possible by the clonal ted stem cells depends on an array of cytokine and cell–cell
expansion of T and B lymphocytes in response to antigen interactions.
(including allergen) stimulation. From the time the human
immune system begins to differentiate in fetal life, lympho- Features of the Adaptive Immune Response
cytes possessing unique reactivity are created by the recom- APCs, which include DCs, monocytes, or macrophages, process
bination of genes encoding antigen receptors expressed on and present antigen within an antigen-binding cleft of major
the lymphocyte cell membrane. Through the expression of histocompatibility complex (MHC) molecules. These events
these receptors, T and B lymphocytes have the ability to bind start at the APC cell surface with the capture and endocytosis of
to and become activated by a specific antigen, which may be antigens, followed by a complex sequence of enzymatic activi-
natural or artificial. Interaction with antigen activates the lym- ties leading to the association of antigenic peptides with MHC
phocytes and generates long-lived, antigen-specific memory molecules and expression back to the cell surface. CD4+ T cells
T and B cell clones. When the same antigen enters the body, recognize antigenic peptides when presented in the context of
there is immediate recognition by these memory cells. Cellular a class II MHC molecule (Fig. 1.2) together with the appropri-
and humoral responses to the antigen are produced more rap- ate costimulatory signals and become activated in response to
idly than in the first encounter, and more memory cells are monocyte-derived IL-1 and other cytokines, including autocrine
generated. This process of expansion of clonal populations of stimulation by IL-2.
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 5

Subsets of Th cells dictate the cytokine production involved IMMUNOGLOBULIN STRUCTURE AND
in three types of immune responses. Th1 response, induced by
FUNCTION
IL-12 and IFN-γ, is responsible for T cell–mediated cytotoxic-
ity. Th2 response, induced by IL-4, IL-5, and IL-13, is respon- B Lymphocytes and the Humoral Immune
sible for the development of IgE- and eosinophil-mediated Response
allergic disease. Th17 response leads to a characteristic neutro-
Engagement of the B cell receptor (BCR) by antigen initiates
philic inflammation and is pathogenic in some experimental
receptor aggregation at the cell surface followed by recruitment
models of autoimmunity. Transforming growth factor-β (TGF-
to lipid rafts. Lipid rafts are specialized membrane microdo-
β), IL-23, and IL-6 are essential cytokines for developing the
mains that facilitate assembly and activation of downstream
Th17 response, which is mediated by IL-17A, IL-17F, IL-21, and
signaling molecules.10 This step places the complex in proxim-
IL-22.
ity to the LYN tyrosine kinase, which phosphorylates tyrosine
The defensive capacity of the immune system needs a mecha-
residues in the Igα/Igβ ITAM motifs and triggers recruitment of
nism to counterbalance this proinflammatory response and to
spleen tyrosine kinase (SYK) and Bruton tyrosine kinase (BTK).
minimize unnecessary tissue damage. Several processes ensure
Activated SYK phosphorylates and recruits the B cell linker
that the different immune effector cells are not activated against
(BLNK) protein, which provides binding sites for phospholi-
host tissues and innocuous substances and that they can down-
pase Cγ2 (PLCγ2), BTK, and VAV proteins, which are guanine
regulate a response after the threat is resolved. All of these pro-
nucleotide exchange factors. PLCγ2 generates the second mes-
cesses underlie immune tolerance, which is classified as central
sengers inositol triphosphate and diacylglycerol, which are nec-
when occurring in primary lymphoid organs, or as peripheral
essary for calcium release from intracellular stores and protein
when occurring in other tissues. Together with central and
kinase C activation. BCR signal transduction also leads to acti-
peripheral tolerance processes, a subset of T cells characterized
vation of the mitogen-activated protein kinase (MAPK) path-
by high levels of CD25 expression (IL-2R α chain) have been
way. B cell activation is further aided by a co-receptor complex
identified as regulatory T (Treg) cells because they were found
that amplifies signals delivered by the BCR. The members of this
to suppress the function of other T cells when present in the
complex include CD19, the complement receptor type 2 (CR2
same site (Fig. 1.3).9
or CD21), and CD81. The CR2 enables the complement path-
Mechanisms of Diseases Involving Adaptive way to synergize with BCR signal transduction, which enhances
B cell activation. Collectively, these signaling events lead to the
Immunity activation of the transcription factors known as nuclear fac-
Distinct mechanisms of immune-mediated diseases are tor of activated T cells (NFAT), nuclear factor-κB (NF-κB),
IgE-mediated hypersensitivity, antibody-mediated cytotox- and activator protein 1 (AP-1). Activation of the BCR on naive
icity, immune complex reaction, delayed hypersensitivity and memory B cells results in their activation and migration
response, antibody-mediated activation or inactivation of to the draining lymph node or other lymphatic tissue. B cells
biologic function, cell-mediated cytotoxicity, and granulo- can respond to three types of antigens, and the type of antigenic
matous reaction. exposure dictates the quality of the ensuing response.

Regulatory T

Dendritic cell cell


IL-10
TGF-β

CD4+ CD25+ T cell


Low-dose antigen CD4+ CD25+ FOXP3+
Chronic antigen exposure
Allergen immunotherapy
IL-10

Th1 or Th2 cell


Fig. 1.3 Regulatory T cells are generated by the interaction of antigen-presenting cells and T cells, mediated by the cytokines inter-
leukin-10 (IL-10) and transforming growth factor-β (TGF-β). These cytokines are secreted when the antigen is presented under certain
conditions, such as when administering allergen immunotherapy at very low concentration. Regulatory T cells secrete IL-10 and
inhibit effector T cells that share similar antigen specificity.
6 Allergy Essentials

Immunoglobulin Structure and Gene Rearrangement a number of mutations that give rise to hyper-IgM syndromes,
Immunoglobulins are composed of two identical heavy chains which result from the failure of B cells to undergo class-switch
and two identical light chains (Fig. 1.4A). Light chains lack recombination. These disorders highlight the critical role that
transmembrane domains and are anchored to heavy chains CD40–CD40L interaction plays in class-switch recombination,
by disulfide bonds. The two heavy chains are linked to each as revealed by the lack of IgG, IgA, and IgE antibodies in these
other by a distinct set of disulfide bonds. Each heavy chain patients.
or light chain has two major domains referred to as the con- There are also some other disorders characterized by abnor-
stant region (C) and the variable region (V), with each domain malities in immunoglobulins. IgG4-related disease is a chronic
responsible for a specialized function. They are denoted as CL inflammatory condition characterized by tissue infiltration by
and VL for the light chains and as CH and VH for the heavy lymphocytes and IgG4-secreting plasma cells, varying degrees
chains. of fibrosis (scarring), and generally rapid response to oral ste-
Heavy-chain variable regions are encoded by one V gene, roids. IgG4 serum levels increased in the acute period in two-
which encodes most V-region amino acids, as well as 1 of 23 thirds of the patients. IgA nephropathy, also known as Berger
diversity (D) and 1 of 6 joining (J) gene segments that are disease, is a kidney disease that occurs when IgA accumulates
located 3′ of the V gene cluster. In contrast, light chain variable in the kidneys and causes inflammation that damages kidney
regions are encoded by only two types of genes: V genes and J tissues. Hyperimmunoglobulinemia E syndromes (HIESs) are
genes. Whereas the Jκ genes are organized in a cluster 3′ to the a heterogeneous group of immune disorders characterized by
Vκ gene cluster, Jλ genes are interspersed with λ constant-region recurrent “cold” staphylococcal infections (due to inadequate
genes. accumulation of neutrophils), unusual eczema-like skin rash,
Immunoglobulin diversity has four sources: multiple V(D) pneumatoceles, and severe lung infections resulting in very high
J genes in the germline, random assortment of heavy chains serum IgE levels.
and light chains, junctional nucleotide variability introduced
during pre-B cell immunoglobulin gene rearrangement, and
somatic hypermutation of immunoglobulin variable regions IMMUNE TOLERANCE
after encounters with antigens.
Introduction
Immunoglobulin Function The physiopathology of immune tolerance–related diseases,
The five classes of antibody molecules are designated IgM, IgD, such as allergies, asthma, autoimmunity, organ transplantation,
IgG, IgA, and IgE. The IgG and IgA classes have more than one tumor, chronic infections, and abortions, is complex and is influ-
member. There are four IgG (γ) sub-classes, designated as IgG1, enced by factors, such as genetic susceptibility, environmental
IgG2, IgG3, and IgG4, and their constant regions exhibit 90% factors and route, dose, or time of the antigen exposure. Many
homology with each other. However, because each IgG sub- common biologic mechanisms prevent immune responsive-
class constant region is encoded by a separate constant-region ness to innocuous environmental allergens and to self-antigens.
gene, the IgG sub-classes are closely related isotypes that exhibit Although most autoreactive T cells undergo selection and clonal
a similar overall structure. The two sub-classes of IgA are simi- deletion in the thymus, a small fraction of cells escape into
larly related to each other. There are two types of light chains: κ the periphery. Additional immunologic control mechanisms
and λ. There are four λ sub-types but only one form of κ. The eliminate or inactivate potentially hazardous effector cells that
nine class and sub-classes of antibody molecules have signifi- emerge from the thymus and move into the periphery (Fig. 1.5).
cantly different expression levels, anatomic locations, and effec- Allergens enter the body through the respiratory and alimentary
tor functions (Table 1.2). The five antibody classes also display tract or injured skin, and the result usually is induction of toler-
characteristic structural features (Fig. 1.4B). ance in healthy individuals.11

Central and Peripheral Tolerance Mechanisms


The processes that constitute immune tolerance normally
IMMUNOGLOBULINS AND HUMAN DISEASE
ensure that immune effector cells are not activated against host
Human conditions of dysregulated immunoglobulin produc- tissues or innocuous agents. Immune tolerance is called central
tion include antibody deficiencies and overproduction of spe- when the response occurs in primary lymphoid organs, such as
cific antibodies. The most serious of the three major categories thymus or peripheral when it occurs in peripheral lymph nodes,
of antibody deficiencies result in reduced B cell numbers and Peyer’s patches, tonsils, or other tissues.
a severe decrease in all isotypes of serum immunoglobulin,
as in agammaglobulinemia. This type of immunodeficiency Central Tolerance
underscores the importance of tyrosine kinases in early B cell T cells experience the first step of tolerance during their matu-
BCR signal transduction. The second category includes selec- ration in the thymus. Prethymic T cells reach the subcapsular
tive deficiencies of IgA or IgG2 production and various genetic region of the thymus, where they proliferate. Maturing cells
mutations that result in hypogammaglobulinemia, such as defi- move deeper into the cortex and adhere to cortical epithelial
ciencies in transmembrane activator and calcium-modulating cells. The T cell receptors (TCRs) on thymocytes are exposed
cyclophilin ligand interactor (TACI). The third category includes to epithelial MHC molecules through these contacts. Negative
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 7

F(ab´)2 fragment IgG1 IgA1 IgM


N termini
Fab fragment VH VH VH
Fab VL VL VL
Cγ1 Cα1 Cµ1
CL
CL
Vh Hinge Cµ2 CL
Variable Cγ2

S-S
S-S

region Vl Cα2

S-S
S-S
Fc
Cγ3 Cµ3
Cα3
Ch1 Cµ4

S-S
S-S

Tailpieces

S-S
S-S

Constant region Cl C termini Tailpieces


S-S S-S Light chain
Papain cleavage S-S Hinge IgE IgD Secretory IgA
S-S
Pepsin cleavage VH VH VH
Ch2 VL Cδ1 VL Cα1 VL
Cε1
S-S

S-S

CL
CHO CHO CL CL
Fc fragment Cε2 Hinge Cα2 Cα2
Heavy chain − +
Cε3 + − Cα3
S-S

S-S

pFc´ Cδ2
Ch3
Cε4 J chain
Cδ3
A COOH COOH Tailpieces

VH

Cµ1
Secretory
Cµ2
component

Cµ3
Cµ4

J
chain

B
Fig. 1.4 Basic structure of immunoglobulin molecules. (A) In the monomeric structure of immunoglobulin molecules, disulfide
bridges link the two heavy chains and the light chains with heavy chains. Enzymatic digestion with papain cleaves the immuno-
globulin molecule into three fragments: two Fab fragments, each of which can bind a single antigen epitope, and the Fc fragment,
which can bind to Fc receptors. Alternatively, pepsin digestion of immunoglobulins results in a single F(ab′)2 fragment, which remains
capable of cross-linking and precipitating multivalent antigen. The Fc portion usually is digested into several smaller peptides by pep-
sin (pFc′). (B) Schematic structures of the five classes of antibodies. IgG1 and IgA1 are shown as examples of the basic structure of
the IgG and IgA classes of antibodies. The other IgG sub-classes differ primarily in the nature and length of the hinge, and the IgA2
hinge region is very short compared with IgA1. Although membrane IgM and IgA exist as monomers, secreted IgA can exist as
dimers, and secreted IgM as pentamers, when linked by an extra polypeptide called the J chain. Both multimeric forms of antibod-
ies can be transported across mucosal surfaces by binding to the polymeric immunoglobulin receptor. Dimeric IgA coupled to the
J chain and secretory component, a part of the polymeric immunoglobulin (Ig) receptor remaining after transport through epithelial
cells, is shown as an example of secretory Ig. (From Delves PJ, Martin SJ, Burton DR, Roitt IM. Roitt’s Essential Immunology. 12th
ed. Oxford: Wiley-Blackwell; 2011:56, 62.)

selection occurs by deletion of self-reactive T cells. Autoantigens thymus, and T cells escape from deletion and negative selection
are presented by medullary thymic epithelial cells, interdigitat- and enter the peripheral circulation. T cell infiltration of the tis-
ing cells, and macrophages at the corticomedullary junction. sues and autoantibody production results in tissue destruction.12
Cells expressing CD4 or CD8 subsequently exit to the periphery. Cells that have escaped negative selection in the thymus
The autoimmune polyendocrine syndrome is a good example are still subject to control in the periphery, because some self-
of central tolerance loss, which is caused by mutations in the reactive CD4+ T cells that are not deleted by negative selection
AIRE gene. In this disease, self-antigens are not displayed in the develop into central Treg cells. These central Treg cells circulate
8 Allergy Essentials

TABLE 1.2 Selected Biologic Properties of Human Immunoglobulin Isotypes


Characteristics IgG1 IgG2 IgG3 IgG4 IgM IgA1 IgA2 IgD IgE
Physical properties
Molecular weight (kDa) 146 146 165 146 970a 160 160 170 190
Serum half-life (days) 29 27 7 16 5 6 6 – 2

Anatomic distribution
Mean serum level (mg/mL) 5–12 2–6 0.5–1.0 0.2–1.0 0.5–1.5 0.5–2.0 0–0.2 0–0.4 0–0.002
Transport across placenta +++ + ++ ± − − − − −
Transport across epithelium − − − − + +++b +++b − −
Extravascular diffusion +++ +++ +++ +++ ± ++ c
++ c
+ +

Functional activity
Antigen neutralization ++ ++ ++ ++ ++ ++ ++ − −
Complement fixation ++ + ++ − +++ + + − −
ADCC + + + ± − − − − +
Immediate hypersensitivity − − − − − − − − +++
ADCC, Antibody-dependent cellular cytotoxicity; −, no effect; ±, no effect or negligible degree; +, small degree; ++, moderate degree; +++, large
degree.
a
Pentameric IgM plus J chain.
b
Dimer.
c
Monomer.

in the periphery as mature T cells and inhibit immune or inflam- of costimulatory receptors, they enter into a state of unrespon-
matory responses against self-antigens. siveness. This state has been called T cell anergy. In addition
to Treg cells, different subgroups of regulatory B cells (Breg)
Peripheral Tolerance play important roles in peripheral tolerance to allergens as
There are multiple mechanisms of peripheral immune tolerance well as immune tolerance in autoimmunity, tumor and chronic
(Fig. 1.5). These mechanisms prevent overactivation of immune infections.
system which cause intensive tissue inflammation. The funda-
mental strategy of immunotherapy for allergic diseases is to Histamine Receptors in Peripheral Tolerance
correct dysregulated immune responses by inducing peripheral One of the primary mediators released from mast cells is his-
allergen tolerance. tamine and this acts through histamine receptors. Histamine
During inflammation, apoptosis of immune effector cells receptor 2 (HR2) activation mediates early desensitization of
is induced by neighbor cells’ death-inducing ligands. Immune basophils. The initial decrease in basophil activity is also associ-
effector cells can undergo apoptosis by expressing death recep- ated with symptom scores in grass pollen immunotherapy. H2R
tors and ligands simultaneously. To keep tissue inflammation at suppresses allergen-associated FcεRI-mediated basophil activa-
low levels, effector T cells are directly tolerized by suppressive tion. HR2 mainly plays a role in immune tolerance mechanisms.
cytokines released by tissue cells. Treg cells suppress effector T Its expression increases in Th2 cells and both suppress allergen-
cells. DCs induce tolerization of host T cells. In asthma, spa- induced T cell responses and trigger the development of periph-
tial separation of T cells and tissue cells, such as the presence eral tolerance by increasing IL-10 production in beekeepers.13–15
of a basement membrane between the epithelium and immune Histamine acts through HR2 and induces IL-10 production by
cells, results in ignorance of effector mechanisms. Tissue cells DCs and Th2 cells; it increases the suppressive effect of TGF-β
in organs with immune privilege use many mechanisms to sup- on T cells and decreases the production of Th2 cytokines, IL-4,
press or delete highly activated effector cells that could other- and IL-13, which are central Th2-type cytokines.14,16
wise damage these tissues.
During an immune response, CD4+ T cells normally receive
signals activated through engagement of the TCR, which rec- Immune Effector Cells and Molecules
ognizes peptides of specific antigens presented on the surface Treg Cells and Regulatory B cells
of APCs by MHC class II molecules. Costimulatory recep- Although various types of cell contribute to establishing
tors, such as CD28, CD2, and inducible costimulator (ICOS) immune tolerance, CD4+FOXP3+ Treg cells play a central role
recognize ligands, such as B7 proteins, CD80, CD86, lympho- in immune control in the periphery. Additionally, in peanut
cyte function–associated antigen 3 (LFA-3), and ICOS ligand allergy, demethylation of FOXP3+ has been shown to be asso-
(ICOSL) expressed on the surface of APCs. These costimulatory ciated with tolerance development.17 Two broad categories of
receptors contribute to activation of the T cell. When T cells Treg cells have been described: naturally occurring Treg cells
receive stimulus only through the TCR without any engagement and antigen-induced Treg cells that secrete inhibitory cytokines,
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 9

FAS FASLG FAS such as IL-10 and TGF-β. In allergic disease, the balance
T T T between allergen-specific Treg cells and disease-promoting
FASLG
Th2 cells appears to determine whether an allergic or healthy
A Deletion or activation-induced cell death
immune response against allergen occurs. In healthy individu-
als, predominant Treg cells are specific for common environ-
mental allergens, indicating a state of natural tolerance.
IL-10–secreting allergen-specific Breg cells have been
TGF-β
T defined in bee venom–tolerant beekeepers, and patients
treated with venom immunotherapy (VIT). Breg cells are
B Direct suppression by tissue cells
CD73−CD25+CD71+ B cells, which are capable of suppressing
allergen-specific CD4+ T cells and produce allergen-specific
IgG4 antibodies after allergy immunotherapy (AIT). Moreover,
TGF-β Breg cells produce IL-35 and TGF-β. IL-10–producing NK reg-
Treg IL-10 Teff ulatory cells suppress allergen-stimulated T cell proliferation in
C Suppression by Treg cells
patients during AIT, and these cells may take place in tolerance
development as other regulatory cell types.14,18
T follicular helper cells (Tfh) are a newly defined cell type iden-
tified by CXCR5+ surface receptor and function in B cell matura-
TGF-β tion and immunoglobulin class switching. A subgroup of Treg,
DC IL-10 Teff
defined as CXCR5+ FoxP3+ Treg cells, are called follicular regula-
CD80-CTLA4 tory T (TFR) cells. They act in the germinal centers of the lymph
PD-L1 or PD-L2-PD1 nodes and suppress T and B cell responses. TFR cells produce
D Tolerization by dendritic cells
more IL-10 compared to TFH cells. There is plasticity between
TFH and TFR cells, and this suggests that TFR cells may play
essential roles in allergen-specific IgE production and suppres-
sion of Th2 responses during immune tolerance development.13,19
T
Transforming Growth Factor-β (TGF-β)
E Ignorance TGF-β is associated with the resolution of immune responses
and the induction of Treg cell populations (Table 1.3). However,
FASLG
Trail the effects of TGF-β in allergic disease are complex, with evi-
TWEAK dence of both disease inhibition and promotion. TGF-β can
inhibit human Th2 responses in vitro. In a murine model, over-
T Privileged tissue expression of TGF-β1 in OVA-specific CD4+ T cells abolished
TGF-β
airway hyperresponsiveness and airway inflammation induced
IL-10 by OVA-specific Th2 cells.
On the other hand, in a mouse model exhibiting properties
F Immune privilege
of chronic asthma, blockade of TGF-β significantly reduced
Fig. 1.5 Multiple mechanisms of immune tolerance. (A) Direct dele- peribronchiolar extracellular matrix (ECM) deposition, airway
tion of immune effector cell by expression of death-inducing ligands. smooth muscle (ASM) cell proliferation, and mucus production
(B) Direct tolerization of effector T cells by suppressive cytokines
in the lung without affecting established airway inflammation
released by tissue cells. (C) Suppression of effector T cells by regula-
or Th2 cytokine production. TGF-β1 may be involved in a nega-
tory T cells. (D) Tolerization of host T cells by tolerizing dendritic cells.
(E) Ignorance of effector mechanisms as a result of spatial separa- tive feedback mechanism to control airway inflammation and
tion of T cells and tissue cells, such as by basement membranes repair of asthmatic airways, inducing remodeling and fibrosis
between the epithelium and immune cells in asthma. (F) Immune to exaggerate disease development in humans.
privilege refers to certain sites in the body that can tolerate the
introduction of antigen without eliciting an inflammatory immune Interleukin-10 (IL-10)
response. These sites include the eyes, the placenta and fetus, and IL-10 plays a role in the control of allergy and asthma. IL-10
the testicles. Tissue cells in these organs use many mechanisms to inhibits many effector cells and disease processes, and its lev-
suppress or delete highly activated effector cells that can damage els are inversely correlated with disease incidence and severity.
these tissues. CTLA4, Cytotoxic T lymphocyte–associated protein IL-10 is synthesized by a wide range of cell types, including B
4; DC, dendritic cell; FAS, member of the tumor necrosis factor
cells, monocytes, DCs, NK cells, and T cells. It inhibits proin-
receptor superfamily, member 6; FASLG, FAS ligand; IL, interleukin;
flammatory cytokine production and Th1 and Th2 cell activa-
PD-L1, programmed death-ligand 1; PD-L2, programmed death-
ligand 2; PD-1, programmed cell death 1; T, T cell; Teff, effector T tion, which is likely attributable to the effects of IL-10 on APCs
cell; TGF-β, transforming growth factor β; Trail, tumor necrosis fac- and its direct effects on T cell function (Table 1.3).
tor (ligand) superfamily, member 10; TWEAK, tumor necrosis factor IL-10 levels inversely correlate with the incidence and sever-
(ligand) superfamily, member 12; Treg, regulatory T cell. ity of asthmatic disease in the lung. In addition, the levels of
10 Allergy Essentials

TABLE 1.3 Functions of Interleukin-10 and Transforming Growth Factor-β


Cell Type IL-10 TGF-β
Dendritic cells (DCs) Inhibits DC maturation, reducing MHC class II and costimulatory ligand Promotes Langerhans cell development
expression Inhibits dendritic cell maturation and antigen presentation
Inhibits proinflammatory cytokine secretion Downregulates FcεRI expression on Langerhans cells
Inhibits APC function for induction of T cell proliferation and cytokine
production (Th1 and Th2)
T cells Suppresses allergen-specific Th1 and Th2 cells Promotes T cell survival
Blocks B7/CD28 costimulatory pathway on T cells Inhibits proliferation, differentiation, and effector function,
including allergen-specific Th1 and Th2 cells
Promotes the Th17 lineage
B cells and Enhances survival Inhibits proliferation
immunoglobulin (Ig) E Promotes Ig production, including IgG4 Induces apoptosis of immature or naive B cells
Suppresses allergen-specific IgE Inhibits most Ig class switching
Switch factor for IgA
Suppresses allergen-specific IgE
CD25+ Tregs Indirect effect on the generation Upregulates FOXP3
Promotes generation in the periphery
Potential effects on homeostasis
IL-10–secreting Tregs Promotes induction of IL-10–secreting Tregs Can promote IL-10 synthesis
Monocytes and Inhibits proinflammatory cytokine production and antigen presentation Inhibits scavenger and effector functions, including
macrophages proinflammatory cytokine production and antigen
presentation
Promotes chemotaxis
Eosinophils Inhibits survival and cytokine production Chemoattractant
Mast cells Inhibits mast cell activation, including cytokine production Promotes chemotaxis
Variable effects on other functions
May inhibit expression of FcεR (receptor 1)
Neutrophils Inhibits chemokine and proinflammatory cytokine production Potent chemoattractant
APC, Antigen-presenting cell; FcεR, Fc fragment of IgE receptor; FOXP3, Forkhead box P3 protein; IL, interleukin; MHC, major histocompatibility
complex; TGF-β, transforming growth factor-β; Th, T helper cell subset; Treg, regulatory T cell.

IL-10 inversely correlate with skin-prick test reactivity to aller-


TABLE 1.4 Sources of Interleukins IL-4
gens. Beekeepers, who undergo multiple bee stings and are natu-
and IL-13
rally tolerant to bee venom allergen, have a high IL-10 response.
IL-10 and IL-10–producing Treg and Breg cells play essential Cell Source IL-4 IL-13
roles in immune tolerance to allergens. In addition, the roles of T helper lymphocytes
Treg and Breg cells and IL-10 have been shown in many autoim- Naive T cells No No
mune, organ transplantation, tumor tolerance conditions.20 T follicular helper (Tfh) cells Yes No
Cytotoxic T lymphocyte–associated antigen 4 (CTLA-4) and Th2 cells Yes Yes
programmed death 1 (PD-1) are negative regulators of T cell func- Natural killer (NK) T cells Yes Yes
tion. Inhibition of these targets leads to increased activation of the Basophils Yes Yes
immune system. While CTLA-4 is thought to regulate T cell pro-
Eosinophils Yes Yes
liferation early during an immune response, particularly in lymph
Mast cells Yes Yes
nodes, PD-1 is thought to suppress T cells later, especially in
peripheral tissues. In other words, CTLA-4 acts early on tolerance Type 2 innate lymphoid cells (ILC2) Yes Yes
induction and PD-1 acts late to maintain long-term tolerance.21

IL-4 induces immunoglobulin isotype switch from IgM to IgE.


CYTOKINES AND CHEMOKINES IN ALLERGIC IL-4 has important influences on T lymphocyte growth, differen-
INFLAMMATION tiation, and survival. As discussed later, IL-4 establishes the dif-
ferentiation of naive Th0 lymphocytes into the Th2 phenotype.
Cytokines in Allergic Inflammation Another important activity of IL-4 is its ability to induce
Interleukin-4 (IL-4) expression of vascular cell adhesion molecule-1 (VCAM-1)
In addition to T helper lymphocytes, IL-4 is derived from baso- on endothelial cells. This enhances adhesiveness of endothe-
phils, NK T cells, ILC2 mast cells, and eosinophils (Table 1.4). lium for T cells, eosinophils, basophils, and monocytes, but not
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 11

Interleukin-13 (IL-13)
IL-13 is homologous to IL-4 and shares many of its biologic
IL-4
IL-13 activities on mononuclear phagocytic cells, endothelial cells,
epithelial cells, and B cells. Thus IL-13 induces IgE isotype switch
IL-13 and VCAM-1 expression. Biologic activities of IL-4 and IL-13
IL-13 are additionally distinguished by their distinct cellular sources
IL-4Rα IL-13Rα1 IL-4Rα (Table 1.4). IL-13, acting through this hormonal mechanism,
γC IL-13Rα1
causes mucus hypersecretion and nonspecific airway hyperre-
IL-4Rα activity (AHR), and its expression results in the characteristic
Type 2 IL-4 receptor IL-13Rα2 airway metaplasia of asthma, with the replacement of epithelial
cells with goblet cells. The importance of IL-13 in presentations
Type 1 IL-4 receptor Decoy IL-13 receptor of asthma associated with a robust IL-13 signature is supported
Fig. 1.6 IL-4 and IL-13 receptors. Type 1 IL-4 receptors are het- by the efficacy of IL-13–targeting therapies in this endotype.
erodimers of IL-4Rα interacting with the shared γC chain and
bind only IL-4. Their unique expression on most T helper cells Interleukin-25 (IL-25)
and mast cells renders these cells only responsive to IL-4. Type
IL-25 is a member of the IL-17 family (IL-17E), but because of
2 receptors can bind both IL-4 and IL-13. They are more widely
its unique spectrum of activities, it has been given this distinct
expressed and consist of heterodimers of IL-4Rα and IL-13Rα1.
In addition, IL-13 can bind to the IL-13Rα2, which lacks a cyto- nomenclature. Binding of IL-25 occurs via a heterodimer com-
plasmic domain and thereby functions as a decoy receptor. IL, plex composed of IL-17RB and IL-17RA.22 It is mainly derived
Interleukin. from epithelial cells. The production of IL-25 by injured epi-
thelial cells is an important innate immune signal driving
Th2 immune deviation in the subsequent adaptive immune
neutrophils, as a characteristic of allergic reactions. IL-4 receptors response. IL-25 stimulates release of IL-4, IL-5, and IL-13 from
are present on mast cells, where they function to stimulate IgE Th2 lymphocytes but, of note, also drives IL-5 and IL-13 secre-
receptor expression, along with the expression of the enzyme leu- tion from type 2 innate lymphoid cells (ILC2).
kotriene C4 (LTC4) synthase. Functional IL-4 receptors are het-
erodimers consisting of the IL-4Rα chain interacting with either Interleukin-33 (IL-33)
the shared γ chain or the IL-13Rα1 chain (Fig. 1.6). This shared IL-33 is a member of the IL-1 superfamily (in which it is des-
use of the IL-4Rα chain by IL-4 and IL-13 and the activation by ignated IL-1F11) that signals through an IL-1 receptor–related
this chain of the signaling protein STAT6 serve to explain many of protein (originally termed ST2) and its co-receptor IL-1RAcP.23
the common biologic activities of these two cytokines. IL-33 is primarily expressed by bronchial epithelial cells,
with additional sources including fibroblasts and smooth
Interleukin-5 (IL-5) muscle cells and it is also inducible in lung and dermal fibro-
IL-5 is the most important eosinophilopoietin and also can blasts, keratinocytes, activated DCs, and macrophages. IL-33
induce basophil differentiation. In addition to stimulating receptors are expressed on T cells (specifically, Th2-like cells),
eosinophil production, IL-5 is chemotactic for eosinophils and macrophages, hematopoietic stem cells, eosinophils, basophils,
activates mature eosinophils, inducing secretion and enhancing mast cells, ILC2, and fibroblasts. As discussed, IL-33 enhances
their cytotoxicity. IL-5 promotes accumulation of eosinophils cytokine secretion by Th2 cells and, like IL-25, induces IL-5 and
through its ability to upregulate responses to chemokines and IL-13 secretion by ILC2.
αdβ2 integrins on eosinophils, thereby promoting their adher- It is possible to avoid food allergy development and to sup-
ence to VCAM-1–expressing endothelial cells. IL-5 prolongs press ongoing food allergy by blocking the IL-25, IL-33, and
eosinophil survival by blocking apoptosis. TSLP.24 Moreover, presence of IL-33 in the airways together
with an inhaled allergen which is tolerogenic previously causes
Interleukin-9 (IL-9) the breakdown of the tolerance.25
The primary source of IL-9 is the T helper lymphocyte population,
including Th2 cells, with additional amounts coming from mast Interleukin-35 (IL-35)
cells ILC2 and eosinophils. IL-9 contributes to mast cell–medi- IL-35 is an antiinflammatory cytokine included in the IL-12
ated allergic responses through its ability to stimulate production superfamily. IL-35 is predominantly secreted by Treg and Breg
of mast cell proteases, inflammatory cytokines, and chemokines. cells. It consists of two chains, IL-12α chain p35 and IL-27α
Additionally, IL-9 primes mast cells to respond to allergens by chain EBV-induced gene3 (Ebi3). IL-35 is involved in the
increasing their expression of FcεRIα. IL-9 synergizes with IL-4 development of tolerance and the production of regulatory
to enhance production of IgE and memory B cell differentiation. cells that express IL-35. Bregs secrete IL-35, which has an auto-
The same synergy leads to enhanced IL-5 production resulting in crine role, to further expand Breg cells to produce more IL-35
greater numbers and maturation of immature eosinophil precur- and IL-10.26,27 In addition to its biological function in immune
sors. IL-9 acts on airway epithelial cells by inducing T cell and cells, IL-35 is required for the maximum suppressive activity
eosinophil chemotactic factors, such as CCL11 (eotaxin), CCL2 of Treg cells. IL-35 mediates the differentiation of a new sub-
(MCP-1), CCL3 (MIP-1α), and CCL7 (MCP-3). set of inducible Treg cells known as iTR35.28 While IL-35 can
12 Allergy Essentials

inhibit the proliferation of Th1 and Th17 cells by blocking cell Chemokines in Allergic Diseases
division, it can also hinder Th2 development through GATA3 Asthma
and IL-4 suppression.29 In addition to these effects, IL-35 medi-
Asthma is a chronic inflammatory lung disease characterized
ates the transformation of Th2 cells into Treg cells, which can be
by airway inflammation, mucus hypersecretion, and bronchial
reversed in the presence of IFN-γ.30 Despite the limited number
hyperresponsiveness. The cellular inflammatory infiltrate in
of studies in humans, it is clear that IL-35 has essential roles in
asthma is composed of eosinophils, lymphocytes, mast cells,
the development of immune tolerance.31
and to a varying extent, basophils and neutrophils.
Thymic Stromal Lymphopoietin (TSLP) Airway exposure to proteases from common allergens, such
as mites and molds, disrupts airway epithelial integrity and
TSLP is another important contributor to Th2 immune devia-
induces epithelial TSLP production (Fig. 1.8). TSLP expands
tion.32 TSLP is expressed by epithelial cells of the skin, gut, and
the number of basophils, prolongs eosinophil survival, and
lung and primes resident DCs in such a way as to promote Th2
increases eosinophil production of CCL2, CXCL1, and CXCL8.
cytokine production by their subsequently engaged effector T
Two other epithelial cytokines, IL-25 and IL-33, also are pro-
cells. High levels of TSLP are found in the keratinocytes of patients
duced on allergen exposure or epithelial damage. IL-25 and
with AD and in the lungs of asthmatic patients. The TSLP recep-
IL-33 upregulate the production of TSLP by epithelial cells and
tor is a heterodimer composed of a unique TSLP-specific recep-
mast cells; induce mast cell release of IL-4, IL-5, IL-13, CCL1,
tor and the IL-7Rα chain (CD127). TSLP receptors are expressed
and CXCL8; promote eosinophil survival; and enhance eosino-
primarily by DCs, but their expression by mast cells Th2 cells and
phil production of CCL2 and CCL3. Activated basophils release
ILC2 also promotes secretion of Th2 signature cytokines.
IL-4, IL-13, granulocyte-macrophage colony-stimulating fac-
The role of IL-25, IL-33, and TSLP in promoting a Th2-
tor (GM-CSF), and CCL3 as well as histamine and leukotriene
associated milieu is summarized in Fig. 1.7. In this model,
C4 (LTC4), which causes vasodilation and increases vascular
injured epithelium has a central role in driving allergic inflam-
permeability. Activated eosinophils generate IL-3, IL-4, IL-5,
mation through its ability to produce these cytokines. TSLP acts
tumor necrosis factor-α (TNF-α), LTC4, platelet-activating fac-
primarily on DCs to drive them to induce a Th2-like process. In
tor (PAF), CCL3, CCL5, and CCL11. In addition to tryptase
addition, both IL-25 and IL-33 act directly on mast cells to drive
and chymase, activated mast cells are also a significant source of
their repertoire of Th2-associated cytokines. More important,
histamine, lipid mediators (LTB4, PGD2), cytokines (IL-3, IL-5,
IL-25, TSLP, and IL-33 act on ILC2 to increase their selective
IL-13, IL-6, IL-10, TNF-α, GM-CSF), and chemokines (CCL1,
production of IL-5 and IL-13. These actions on ILC2 and mast
CCL2, CCL3, CCL5, CCL17, CCL22, CXCL8).
cells can occur independent of ongoing allergen exposure, sug-
Activation and differentiation of naive T cells into Th2 cells
gesting a mechanism for allergen-independent perpetuation of
are marked by downregulation of L-selectin and CCR7 and
allergic inflammation.
appearance of CCR4, CCR8, CRTh2, and the BLT1 receptor

IL-4
IL-25 IL-5
ILC2
IL-9
IL-13
Epithelial
damage

Allergen IL-33 Th0 Th2 IL-4


IL-5
IL-9
IL-13
Microbes
TSLP Mast
cell

DC

Fig. 1.7 Epithelium-derived cytokines in Th2 differentiation and allergic inflammation. The interleukins IL-25 and IL-33 and thymic
stromal lymphopoietin (TSLP) are produced by injured epithelium and play critical roles in driving expression of Th2 cytokines. TSLP
acts on dendritic cells to direct them to promote the differentiation of naive T cells into Th2 cells. By contrast, IL-25 and IL-33 act
directly on the naive T cells to promote Th2 immune deviation. In addition, these three cytokines can generate a Th2 cytokine milieu
independent of the adaptive immune system. TSLP and IL-33 directly induce the full repertoire of Th2 cytokine secretion from mast
cells. Similarly, IL-25, TSLP, and IL-33 act on type 2 innate lymphoid cells (ILC2) to drive their more restricted secretion of IL-5 and IL-13.
DC, Dendritic cell; IL, interleukin; Th, T helper.
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 13

Lung Proteases
Mites LPS

Molds Epithelium

Epithelial damage Smooth


Draining lymph nodes DC muscle
Afferent
TSLP CCL11
CCL22 TNF
IL-25 CCL13
CCL17 IL-1β
IL-33 CCL5
CXCL10
CCL20
Naive DC Eosinophil Basophil
or
Teff Mast
Memory
cell Fibroblast
T cell Th2
CCL11
Th2 Th2 IL-4, IL-5, IL-13, IL-4, IL-5, IL-4,
Th2 Th2 CCL17, CCL22, CCL2, IL-13
Th2
Th2 Th2 CCL1, LTB4, CCL11
PGD2
HEV Th2 Th2 CCR4 CCR8 Amplification
↑IL-4, IL-5,
Th2 IL-13
CRTH2 BLT1
CXCR3
CCR6
Th1 Th17
Efferent

CCR7

Naive or Th2 Th2 Th2 Th2 Th2 Th2 Th2 Th2 Th1 Th2 Th2 Th2 Th2 Th2 Th17
Memory Th17 Th1 Blood
Th2 Th2 Th2 Th2 Th2 Th2 Th1 Th2 Th2 Th2
T cell
L-selectin

Chemoattractant receptors L-selectin IgE Antigen

Fig. 1.8 Chemokines and asthma. Asthma is characterized by the infiltration of lung tissue with T helper type 2 (Th2) cells producing
IL-4, IL-5, and IL-13. Allergen proteases disrupt airway epithelial integrity and induce thymic stromal lymphopoietin (TSLP), IL-25, and
IL-33, while epithelial toll-like receptor activation leads to IL-1β and tumor necrosis factor (TNF) production. These cytokines upregu-
late CC chemokine and Th2 cytokine production and release by smooth muscle cells, fibroblasts, mast cells, eosinophils, and baso-
phils. Activated antigen-presenting cells travel to the draining lymph nodes and promote the generation of Th2 cells, which enter the
lung and release more Th2 cytokines, thus amplifying the allergic response in the lung. CCL, C–C chemokine ligand; CCR, C–C che-
mokine receptor; DC, dendritic cell; HEV, high endothelial venules; IgE, immunoglobulin E; IL, interleukin; LPS, lipopolysaccharide.

for leukotriene B4 (LTB4). These receptors enable Th2 cells to inflammatory infiltrate in this disorder. These include CCR2
move down the concentration gradient in response to CCL17, and CCR3 ligands (CCL13, CCL11, and CCL26) for eosinophil
CCL22, CCL1, prostaglandin D2 (PGD2), and LTB4, mediators and mast cell recruitment, CCR4 and CCR8 ligands (CCL22
released by DCs and activated mast cells. IL-4 and IL-13 induce and CCL1) for Th2 cell recruitment, CCR10 ligand (CCL27) for
lung-residing macrophages, DCs, epithelial cells, and endothe- T cell entry into the epidermis, and CCL18.
lial cells to produce CCL11, CCL24, CCL26, CCL1, CCL17, and The pathophysiology of AD begins with intense pruritus
CCL22, thus amplifying the allergic inflammatory response by and the mechanical injury that results from chronic scratching
attracting more eosinophils and Th2 cells. (Fig. 1.9). Mechanical trauma can directly activate mast cells, which
release histamine, neuropeptides, proteases, kinins, and cytokines,
Atopic Dermatitis many of which further exacerbate pruritus. Furthermore, TSLP
AD is a pruritic chronic inflammatory disease of the skin in levels increase acutely in the skin after mechanical trauma. TSLP
which CD4+ memory T lymphocytes, DC subsets, eosinophils, induces DC activation and DC production of CCL17 and CCL22.
and mast cells infiltrate the perivascular, subepidermal, and The trafficking of memory T cells into the skin requires
intraepidermal areas. A number of chemokines are aberrantly cutaneous lymphocyte antigen (CLA), which interacts with
expressed in the skin of patients with AD and help recruit the E-selectin on inflamed endothelium, and initiates rolling. The
14 Allergy Essentials

Skin Itch

Scratch

Mechanical injury

Allergen
SEB Epidermis

↑CCL27 SEB
↑CCL8

SEB
Eosinophil
DC SEB TSLP
Skin draining lymph nodes
SEB Degranulation
Afferent ↑Survival
Macrophage
lymph CCL22 Mast ↑IL-4, IL-5
CCL17 cell CCL2
CCL17 CCL11
DC CCL8 Degranulation CCL2
Naive
or CCL22, CCL17,
Memory CCL1, IL-4, IL-5, IL-13
T cell Th2
Th2 Th2
Th2 Th2
Th2 Amplification
Th2 Th2 CCR8 CCR10
Th2 Th2 ↑↑IL-4, IL-5
HEV Th2 IL-13
CCR4 CCR4
CLA
Efferent
lymph
↑E-selectin
↑CCL17 ↑CCL17
CCR7 CLA
Th2 Th2 Th2 Th2 CCR4
Naive CCR4
Th2 Th2 Th2 Th2 Th2 Th2 Blood
or
Memory CCR8
Th2 Th2 Th2 Th2 Th2 Th2 CCR10
L-selectin T cell Skin-tropic Th2 cells

Chemoattractant receptors L-selectin IgE Antigen CLA

Fig. 1.9 Chemokines and atopic dermatitis. Atopic dermatitis begins with intense pruritus, chronic scratching, and mechanical injury
to the skin. Mechanical trauma leads to mast cell release of Th2 cytokines and CC chemokines and upregulates local TSLP produc-
tion, while loss of normal barrier function increases exposure to allergens and SEB. TSLP-activated dendritic cells travel to the
draining lymph nodes and promote Th2 cell differentiation. Th2 cells enter the skin and release Th2 cytokines, thus amplifying the
allergic response in the skin. CCL, C–C chemokine ligand; CCR, C–C chemokine receptor; CLA, cutaneous lymphocyte antigen; DC,
dendritic cell; IgE, immunoglobulin E; IL, interleukin; SEB, staphylococcal enterotoxin B; Th2, T helper type 2; TSLP, thymic stromal
lymphopoietin.

trafficking molecules most highly expressed by T cells isolated BIOLOGY OF IMMUNE CELLS
from healthy skin are CLA, CCR4, CCR6 (>80%–90%), and,
to a lesser extent, CCR8 (50%). Whereas the ligands for CCR6 T Lymphocytes
and CCR8 are upregulated in inflammation, skin endothelial Two classes of α/β T lymphocytes that bear the co-receptors CD4
cells and keratinocytes constitutively express CCL17 (one of the or CD8 are involved in adaptive immune responses. CD4+ T cells
ligands for CCR4) and CCL27 (only known ligand for CCR10), are traditionally called Th cells because they activate and direct
respectively. other immune cells. There are also populations of CD4+ Treg
Eczema lesions as the hallmark of AD and allergic contact cells that modulate immune responses. CD4+ T cells recognize
dermatitis lesions are induced by keratinocyte apoptosis, related antigen presented by class II MHC molecules on APCs, includ-
to IFN-γ, Fas-Fas–ligand interaction, TNF-α, TNF-related weak ing DCs, B cells, and macrophages. Exogenous protein antigens
inducer of apoptosis (TWEAK), and IL-32.33,34 are taken up by APCs and processed into peptides in endocytic
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 15

vesicles, which are presented on the cell surface bound to class CD4+ T cells. IL-6, IL-1β, TGF-β1, and in some situations, IL-23
II MHC molecules. The CD8+ cytotoxic T cells (CTLs) recog- promote Th17 development.
nize antigen presented on MHC class I molecules. Class I MHC In the secondary lymphoid tissue, a naive T cell differenti-
molecules are present on the surface of all nucleated cells. Their ates into an effector cell. Compared with naive T cells, effector
cytotoxic functions are carried out by release of preformed cells do not require costimulation to be activated, allowing these
effector molecules and by interactions of cell surface molecules. cells to respond to antigen with hair-trigger rapidity to produce
Antigen-activated CD4+ T cells have the potential to differen- high levels of cytokines and chemokines, which then direct the
tiate into effector cells, each with distinct functional properties immune response. Most activated effector CD4+ T cells die sub-
conferred by the pattern of cytokines they secrete (Fig. 1.10).35 sequent to an immune response through the process of activa-
Th1 cells are a subset of CD4+ T cells that secrete IFN-γ, whereas tion-induced cell death, but a subset of CD4+ T cells will persist
Th2 cells produce IL-4, IL-5, IL-9, IL-10, and IL-13. Th17 cells as memory cells for the life of the host. CD4+ memory T cells
produce IL-17A, IL-17F, and IL-22. Treg cells produce IL-10 persist in lymphoid organs as central memory cells and in non-
and TGF-β1, are naturally occurring and induced, suppress T lymphoid tissues as effector memory cells. The effector memory
cell differentiation and APC activation, and are not considered T cells respond rapidly to repeat exposures to antigen, whereas
effector cells. Th1 cells stimulate strong cell-mediated immune central memory T cells are slower to be mobilized.
responses, particularly against intracellular pathogens. Th2 cells
are elicited in immune responses that require a strong humoral B Lymphocytes
component and in antiparasitic responses. Th17 serve critical The humoral immune response is generated by B cells. Mature
host defense functions at mucosal surfaces. B cells express immunoglobulin on its cell surface, which con-
Cytokines are the primary factors that influence the CD4+ stitutes the antigen-specific BCR. BCR is a molecular complex
Th cell generation and are considered the third signal in CD4+ made up of antigen-binding or variable (V) regions. This region
T cell differentiation.20 IFN-γ and IL-12 stimulate the induction of the protein varies among immunoglobulins, allowing each
of Th1 cells. IL-4 drives Th2 cell generation by direct action on antibody to bind to any foreign structure that the individual
CD4+ T cells. IL-13 is involved in the induction of Th2 cells by may encounter. To generate this diverse immunoglobulin
an unknown mechanism, although not through direct effects on repertoire, during development in the bone marrow, B cells
undergo somatic deoxyribonucleic acid (DNA) recombination
of the variability (V), diversity (D), and joining (J) regions of
the immunoglobulin heavy and light chains. The invariant or
STAT4 activation constant region of the antibody is specialized for different effec-
DC1 IL-12 IL-12Rβ2 tor functions in the immune system after antibody is secreted.
(mature) Th1 GATA-3 There are five main constant-region forms: IgM, IgD, IgG, IgE,
CD80/CD86 MAF
CD28 and IgA. The BCR in the membrane-bound form recognizes
MHC + peptide
CD4 and binds antigen and transmits activation signals into the cell.
GATA-3lo Naive B cells recirculate through peripheral lymphoid tissues
T cell
MAFlo STAT6 activation
(naive) until it binds specific antigen through surface immunoglobu-
GATA-3
IL-4 Th2 MAF lin and is activated (i.e., signal 1). Most antibody responses,
including antibody responses to protein antigens, require
DC IL-10
antigen-specific T cell help. Antigen bound to surface immu-
noglobulin is internalized, processed, complexed with MHC
IL-6
TGF-β1 class II molecules, and displayed on the cell surface. Previously
RORγ t primed CD4+ T cells that recognize the peptide-MHC class II
Th17 complex on the B cell provide the second signal for activation.
The cytokines secreted by CD4+ Th cells during B cell activation
regulate which immunoglobulin heavy-chain constant regions
Fig. 1.10 Generation of helper T cell types 1, 2, and 17 (Th1, Th2, will be selected during class-switch recombination to best serve
and Th17) from a naive CD4+ T cell. A naive CD4+ T cell does not the functions of the specific immune response. Th2 responses to
secrete cytokines and has low expression levels of transcription allergens stimulate B cell activation and result in elevated levels
factors GATA-3 and MAF. Differentiation along the Th1, Th2, or of allergen-specific IgE.
Th17 pathway is triggered by stimulation by antigen presented to
the T cell receptor in the context of the major histocompatibility Innate Lymphoid Cells
complex (MHC) by the appropriate antigen-presenting cell (APC)
Populations of lymphoid cells that lack rearranged antigen
and a second signal imparted by ligation of costimulatory mol-
ecules CD80/CD86 and CD28. Dendritic cells (DCs) represent the receptors, which were called ILCs, have been recently identi-
key APCs for naive T cells. Those that produce interleukin-10 (IL- fied. These ILC populations can be divided into three groups,
10) favor Th2 differentiation, and those that produce interleukin-12 based on shared phenotypic and functional properties like T
(IL-12) stimulate Th1 differentiation. Th17 cells can be generated cells. Type 1 ILC (ILC1) constitutively express T-bet and are
in the presence of interleukin-6 (IL-6) and transforming growth able to produce IFN-γ upon activation. Type 2 ILC (ILC2) con-
factor-β1 (TGF-β1), presumably produced by DCs. stitutively express GATA-3 and in response to IL-25, IL-33, and
16 Allergy Essentials

TSLP stimulation produce IL-5 and IL-13. Type 3 ILC (ILC3) functional itBreg cells. These cells can contribute to immune
constitutively express ROR-γ and in response to IL-1β and IL-23 tolerance induction and suppression of T cell responses both by
produce IL-17, IL-22, and IFN-γ.36 a cell-to-cell contact through programmed cell death-ligand 1
ILC type 2 seems to be important in allergic responses. The and by secretion of IL-10.13
ILC2/ILC1 ratio is high in patients with perennial AR sensi-
tized to house dust mite; however, it turns to normal levels fol- Dendritic Cells
lowing a successful AIT. In the presence of retinoic acid, ILC2 DCs are the most important APCs found throughout the body
cells transformed into regulatory ILCs (ILCregs) which produce and are mainly recognized for their exceptional potential to gen-
IL-10. These cells can suppress Th2 cell and ILC2 activation. DCs erate a primary immune response and sensitization to allergens.
that have the capability of retinoic acid production also induce DCs determine the T cell polarization process that produces Th1
peripheral Treg cell differentiation. Putting these together, one cells (generating mainly IFN-γ), Th2 cells (generating mainly
may suggest that ILCregs may participate in tolerance induction IL-4, IL-5, and IL-13), Th17 cells (generating mainly IL-17),
in the mechanisms of AIT.13,19 ILC2s take place in many functions and Treg cells (generating mainly IL-10 and TGF-β). These cells
during the inflammatory process in asthma and AD (Fig. 1.11) are also recognized for their ability to produce ongoing effector
Another type of ILC, ILC type 3, may have essential roles in responses that are crucial in maintaining allergic inflammation.
immune tolerance induction. CD40L-expressing ILC3s locate In humans, circulating DCs can be broadly divided into two
in close contact with B cells in tonsils. Both cells work inter- groups: (1) mDCs and (2) pDCs. Both subsets express a differ-
dependently, as ILC3s induce IL-15 production in B cells and ent repertoire of TLRs and display a diverse cytokine signature
IL-15 which is a potent growth factor for ILC3s increases CD40L after microbial stimulation. mDCs selectively express TLR2–6
expression on ILC3s. CD40L+ ILC3s induce IL-10–secreting and TLR8 and respond to bacterial and viral infections by pro-
Breg cells through the CD40L and BAFF-receptor–dependent ducing large amounts of IL-12. In contrast, pDCs constitutively
pathway. ILC3-induced Breg cells are characterized by CD27– express the endosome-associated TLR7 and TLR9, and they are
IgD+IgM+CD24highCD38highCD1d+ immature transitional the main producers of type 1 IFNs in humans.6
(itBreg) phenotype. This interaction is important for the main-
tenance of immune tolerance against innocuous antigens and Mast Cells
is inadequate in allergic diseases. In tonsils, generation of func- Mast cells are present throughout connective tissues and muco-
tional allergen-specific Treg cells takes place. ILC3s, Breg cells, sal surfaces and are especially prominent at the interface with
and Treg cells localize side by side in the interfollicular regions the external environment, such as the skin, respiratory tract,
of palatine tonsils. CD40L+ ILC3s may be essential in the main- conjunctiva, and gastrointestinal tract. Mast cells contribute to
tenance of immune tolerance in tonsils through induction of the maintenance of tissue homeostasis, with important roles in

Allergen
FLG mutation(+)

IL-25
E Cadherin
DC ILC Type 2
TSLP Amphiregulin Lineage-
IL-7R+(CD127)
IL-33 c-KITInt(CD117)
Tissue CRTH2+
repair ST2+
IL-25R+
KLRG1
IgE Isotype switc IL-1R+
h ILC2
B cell
IL-9, IL13
IL-5
IL-4 PGD2

Treg
Eosinophil Mast cell

Basophil
Fig. 1.11 The role of ILC2 during the inflammatory process in asthma and atopic dermatitis. DC, Dendritic cell; IgE, immunoglobulin
E; IL, interleukin; ILC, innate lymphoid cell; PGD2, prostaglandin D2; Treg, regulatory T.
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 17

wound repair, revascularization, and protective responses to bac- less than 1% of blood leukocytes. Basophils play a critical role in
terial infection and envenomation. Their “misguided” activation allergic disease by infiltrating sites of allergic inflammation and
by allergens contributes to the development of allergic symptoms. releasing mediators and cytokines that perpetuate type I (imme-
The best-studied mechanism of mast cell activation, and the one diate) hypersensitivity reactions. Degranulation events resulting
considered most relevant to allergic disease, is activation mediated in the release of these mediators are preceded by the interaction
through the high-affinity IgE receptor FcεRI. IgE-dependent signal- of allergen with specific IgE molecules bound to the high-affinity
ing in vivo is initiated when multivalent allergen binds to allergen- IgE receptors on the surface of these cells. This IgE-dependent
specific IgE bound to the FcεRIα chain. IgE-dependent activation activation also leads to the production of immunomodulatory
of the mast cell induces granule swelling, crystal dissolution, and cytokines. In particular, basophils are a significant source of IL-4
granule fusion. This sequence is followed by exocytosis with release and IL-13, two Th2 cytokines, whose expression is characteristic
of mediators into the extracellular space—a process termed ana- of allergic lesions and which are now considered critical compo-
phylactic degranulation. In addition to the stored granule-derived nents in the pathogenesis of allergic disease.
mediators, newly formed metabolites of arachidonic acid also are
released from mast cells after IgE-dependent activation (Table 1.5). Eosinophils
Eosinophils are bone marrow–derived granulocytes that play an
Basophils important pathophysiologic role in a wide range of conditions,
Basophil granulocytes develop in the bone marrow and are including asthma and related allergic diseases and parasitic hel-
released into the circulation as mature end-stage cells representing minth infections. Eosinophils are unique among circulating
leukocytes in their prodigious capacity to produce a variety of
mediators, including granule proteins, cytokines, lipids, oxidative
TABLE 1.5 Classical Preformed and Newly products, and enzymes (Table 1.6). Eosinophils express receptors
Generated Human Mast Cell Autacoid recognizing the Fc portion of various immunoglobulins (FcR).
Mediators and Proteases With Examples of Beads coated with IgA or secretory IgA (sIgA) induce degranula-
Their Biologic Effects tion of eosinophils, and eosinophils from allergic individuals dis-
Mediator Activity play enhanced FcαR expression. However, most reports suggest
Histamine Bronchoconstriction; tissue edema; ↑vascular
that ligation of FcεRI does not result in measurable eosinophil
(stored) permeability; ↑ mucus secretion; ↑ fibroblast degranulation. Exposure of eosinophils ex vivo to various cyto-
proliferation; ↑ collagen synthesis; ↑ endothelial kines mimics in vivo primed eosinophils. IL-5 activates LTC4 and
cell proliferation, dendritic cell differentiation and O2− generation, phagocytosis, and helminthotoxic activity, as
activation well as Ig-induced degranulation. Both TSLP and IL-33 activate
Heparin (stored) Anticoagulant; mediator storage matrix; sequesters eosinophil effector functions, such as adhesion to matrix pro-
growth factors; fibroblast activation; endothelial cell teins, cytokine production, and degranulation.
migration
Tryptase (stored) Degrades respiratory allergens and cross-linked
IgE; generates C3a and bradykinin; degrades
neuropeptides; TGF-β activation; increases basal TABLE 1.6 Eosinophil Mediators
heart rate and ASM contractility; ↑ fibroblast
proliferation and collagen synthesis; epithelial ICAM- Granule proteins
1 expression and CXCL8 release; potentiation of mast Major basic protein (MBP)
cell histamine release; neutrophil recruitment MBP homolog (MBP2)
Eosinophil cationic protein (ECP)
Chymase (stored) ↑ mucus secretion; ECM degradation, type I procollagen
Eosinophil-derived neurotoxin (EDN)
processing; converts angiotensin I to angiotensin II;
Eosinophil peroxidase (EPX)
↓ T cell adhesion to airway smooth muscle; activates
Charcot–Leyden crystal (CLC) protein
IL-1β, degrades IL-4, releases membrane-bound SCF
Secretory phospholipase A2 (sPLA2)
PGD2 Bronchoconstriction; tissue edema; ↑ mucus secretion; Bactericidal/permeability-inducing protein (BPI)
(synthesized) dendritic cell activation; chemotaxis of eosinophils, Acid phosphatase
Th2 cells, and basophils via the CRTH2 (CD294) Arylsulfatase
receptor β-Glucuronidase
LTC4/LTD4 Bronchoconstriction; tissue edema; ↑ mucus secretion;
(synthesized) enhances IL-13–dependent airway smooth Lipid mediators
muscle proliferation; dendritic cell maturation and Leukotriene B4 (negligible)
recruitment; eosinophil IL-4 secretion; mast cell IL-5, Leukotriene C4
IL-8, and TNF-α secretion; tissue fibrosis 5-HETE
5,15- and 8,15-diHETE
ASM, Airway smooth muscle; CRTH2, chemoattractant receptor of 5-oxo-15-hydroxy-6,8,11,13-ETE
Th2 cells; ECM, extracellular matrix; ICAM-1, intercellular adhesion
Platelet-activating factor (PAF)
molecule 1; IgE, immunoglobulin E; IL, interleukin; LTC4, leukotriene
Prostaglandin E1 and E2
C4; LTD4, leukotriene D4; PGD2, prostaglandin D2; SCF, stem cell factor;
TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α. Thromboxane B2
(Continued)
18 Allergy Essentials

TABLE 1.6 Eosinophil Mediators —cont’d


Oxidative products
Superoxide radical anion (OH−)
Hydrogen peroxide (H2O2)
IL-6 TSLP INF-γ CCL1 IL-β BAFF
Hypohalous acids CCL3 GM-CSF CXCL9 CCL17 IL-6?? APRIL
CXCL8 IL-15 CXCL10 CCL22 TGF-β??
Enzymes IL-33 CXCL11 IL-33
Collagenase CCL20 IL-β
IL-4
Metalloproteinase-9 IL-11
Indoleamine 2,3-dioxygenase (IDO)
Neutrophil B cell
Th17 cell
Cytokinesa Th1 cell
IL-1α Dendritic cell
IL-2 Th2 cell
IL-3
Fig. 1.12 Interaction between airway epithelial cell–derived cyto-
IL-4
kines and inflammatory cells. APRIL, A proliferation-inducing ligand;
IL-5
BAFF, B cell–activating factor of the TNF family; CCL, C–C chemokine
IL-6
ligand; GM-CSF, granulocyte-macrophage colony-stimulating factor;
IL-9
IFN-γ, interferon-γ; IL, interleukin; TGF-β, transforming growth factor-β;
IL-10
Th, helper T cell subset; TSLP, thymic stromal lymphopoietin.
IL-11
IL-12
IL-13
IL-16 response to inflammation. Accordingly, these structural cells
Leukemia inhibitory factor (LIF) play crucial roles in the pathogenesis and symptoms of allergic
Interferon-γ (IFN-γ) disease and asthma in concert with immune cells.
Tumor necrosis factor-α (TNF-α)
GM-CSF Airway Epithelial Cells
APRIL
The epithelium constitutes the interface between the external
Chemokines environment and the internal milieu of the lung. It is the site
CXCL8 (IL-8) of first contact with inhaled particles, pollutants, respiratory
CCL2 (MCP-1) viruses, and airborne allergens. Consequently, the epithelium
CCL3 (MIP-1α) plays an important role as a physical and immune barrier. The
CCL5 (RANTES) epithelium senses PAMPs on inhaled foreign substances via
CCL7 (MCP-3) their PRRs and regulates airway homeostasis through the pro-
CCL11 (eotaxin) duction of a multitude of mediators, such as GM-CSF, TSLP,
CCL13 (ECP-4)
IL-25, and IL-33, which promote a Th2 bias in DC precursor
Growth factors (Fig. 1.12). In other words, epithelial cells bridge the innate and
Nerve growth factor (NGF) adaptive immune responses by translating environmental expo-
Platelet-derived growth factor (PDGF) sures into disease phenotypes.
Stem cell factor (SCF) Epithelial cell structure and function are abnormal in patients
Transforming growth factor (TGF-α, TGF-β) with asthma. At a gross level, the composition of the asthmatic
APRIL, A proliferation-inducing ligand; ETE, eicosatetraenoic acid; airway epithelium is different from that of the non-asthmatic
GM-CSF, granulocyte-macrophage colony-stimulating factor; HETE, population. For example, goblet cell hyperplasia and excessive
hydroxyeicosatetraenoic acid; IL, interleukin. mucus production are common features of asthma that contrib-
a
Physiologic significance of these cytokines needs to be confirmed.
ute significantly to morbidity and mortality. Moreover, epithe-
lial cells isolated from patients who have asthma have a deficient
innate immune response from type I antiviral IFNs, particularly
CONTRIBUTION OF STRUCTURAL CELLS TO of IFN-β release during rhinovirus infection. Changes of epi-
thelial cell structure and function occur early in disease patho-
ALLERGIC INFLAMMATION
genesis. These findings place the epithelium at the forefront of
While structural cells, such as epithelial, bone, smooth muscle asthma pathogenesis, and understanding the mechanisms that
cells, or fibroblast, have their proper function, they produce underlie these abnormalities will have short- and long-term
cytokines, chemokines, lipid mediators, and growth factors clinical significance for the treatment of this disease.
which control mobility of immune cells and local inflammatory When the epithelial cells are exposed to an external insult,
milieu. Symptoms of allergic airway disease, such as sneezing, such as allergens, pollutants, viruses, fungi, and bacterial tox-
rhinorrhea, unproductive coughing, episodic bronchospasm, ins, epithelial barrier is damaged, epithelial cytokines (TSLP,
and sensations of breathlessness, are neuronally mediated in IL-25, and IL-33) called alarmins are released. IL-25 and IL-33
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 19

activate ILC2s to produce IL-4, IL-5, and IL-13. Rhinovirus can Neuronal Control of Airway Function
also induce IL-33 and promote type 2 inflammation. The epithe- Both the immune system and the nervous system are critical
lial barrier is disrupted by Th2 cells, type 2 ILCs, and their cyto- to host defense within the airways. The immune system uses
kines IL-4 and IL-13 in human bronchial epithelium. Meanwhile, cellular and humoral mechanisms to protect the peripheral air
CPG-DNA administration strengthens the tight junction (TJ) spaces from invasion and colonization by microorganisms. The
integrity of the bronchial epithelial barrier. In addition, TSLP- nervous system protects the airways by orchestrating reflexes,
stimulated CD11c+ DCs can activate CRTH2+ Th2 effector mem- such as sneezing, coughing, mucus secretion, and broncho-
ory cells and undergo further Th2 polarization to magnify their spasm in response to inflammation. Therefore the nervous sys-
role in allergic inflammation. Periostin is secreted by stimulated tem serves as the principal transducer between immunologic
airway epithelial cells. It is an ECM protein and is considered a aspects of allergic inflammation and the symptomatology of
biomarker of type 2 inflammation. Periostin gene expression is immediate hypersensitivity.
increased by IL-13 and IL-4 in bronchial epithelial cells. Periostin Nerve–immune interactions can be inappropriate and dele-
functions on fibroblasts to promote airway remodeling, increase terious, as with allergy; the immune response triggered by aller-
mucus secretion, and recruit eosinophils.37 gen exposure can recruit the nervous system in a way that is not
Epithelial TJs seal the epithelia and form an essential part beneficial to the host and causes or exacerbates the symptoms of
of the barrier between the inner tissues and the external envi- allergic disease: irritation, pruritus, sneezing, coughing, hyper-
ronment. They control the paracellular flux and epithelial per- secretion, reversible bronchospasm, and dyspnea. Relatively
meability and prevent the entrance of foreign particles, such as little is known about the specific pharmacology of allergen–
allergens and toxins to subepithelial tissues. They form com- immune–nerve interactions, but the mediators likely include
plexes with members of the claudin family, the marvel family, histamine, arachidonic acid metabolites, tryptase, neurotroph-
and the junctional adhesion molecule (JAM) family spanning ins, chemokines, and cytokines. In addition, the allergic reac-
the membrane and forming homo- and heterodimeric con- tion in the respiratory tract is associated with overt activation,
nections between adjacent cells. Scaffold proteins, such as the increases in electrical excitability, as well as phenotypic changes
zonula occludens (ZO) family, link the TJ complex to the actin in sensory, central, and autonomic neurons. Future research
cytoskeleton. Epithelial barrier TJ defects are reported in several into the mediators and mechanisms of allergen-induced neu-
allergic and inflammatory disorders, such as AD, asthma, and romodulation will not only increase our basic understanding of
chronic rhinosinusitis, and a role for TJ in smooth muscle cells the pathophysiology of allergic disease but also suggest novel
is described in asthma pathogenesis.38–45 therapeutic strategies.47
Epithelial TJs are very sensitive to environmental factors.
A recent study with laundry detergents demonstrated the dev-
astating effects on TJ barrier integrity and cellular toxicity of CYTOKINE NETWORKS IN ALLERGIC
human bronchial epithelial cells, even at very high dilution,
without affecting epigenome and TJ gene expression.46
INFLAMMATION
Cytokines play a key role in the orchestration and perpetua-
Airway Smooth Muscle Cells tion of allergic inflammation and are now targeted in therapy
In asthma, the ASM contracts in response to multiple stimuli, (Fig. 1.13).48 Allergic inflammation is characterized by the
but it also produces ECM proteins, proteases that modulate secretion of Th2 cytokines, including IL-4, IL-5, IL-9, and
these proteins, and myriad growth factors and cytokines. These IL-13, which are secreted mainly by Th2 cells. The use of bio-
collectively lead to airway remodeling—the pathology that char- logic immune response modifiers that target and neutralize
acterizes asthma and consists of thickening of the airway wall, cytokines is beginning to shed new light on the role of indi-
increased angiogenesis, mucous cell hyperplasia, thickening of vidual Th2 cytokines. IL-4 and IL-13 play a key role in IgE syn-
the basement membrane, and increased bulk of muscle. It was thesis through isotype switching of B cells and appear to play
previously thought that remodeling was a response to chronic a critical role in animal models of asthma. Thus far, blocking
airway inflammation, but it seems more likely that inflamma- IL-4 and IL-13 or their common receptor IL-4Rα has not yet
tion and remodeling develop along separate pathways. This is been shown to be of clinical benefit in asthma, but many clini-
consistent with the finding that bronchoconstriction alone in cal trials are currently under way. IL-5 is of critical importance
the absence of an inflammatory or allergic stimulus can lead to in the differentiation, survival, and priming of eosinophils. A
airway remodeling. humanized monoclonal IL-5 neutralizing antibody, mepoli-
ASM is a functional part of the innate immune system. It zumab, induced a profound decrease in eosinophils in the blood
expresses messenger RNAs (mRNAs) for TLR1 through TLR10 and in induced sputum in patients with mild asthma but had
and functional TLR2 and TLR3, indicating ASM can respond to no effect on the response to inhaled allergen. Clinical trials of
bacterial and viral infections. ASM modulates leukocyte traffick- anti–IL-5 in unselected symptomatic asthmatic patients showed
ing and function in asthma by activating cell adhesion molecules no overall clinical improvement. Yet in highly selected patients
and secretion of chemokines and cytokines. When the response with severe asthma and sputum eosinophilia, despite high doses
from cells obtained from people with asthma and people without of inhaled or oral corticosteroids, mepolizumab decreased the
asthma were compared, higher levels of cytokines and profibrotic frequency of exacerbations and reduced requirements for oral
factors were observed in the asthma-derived cells. corticosteroids, although it did not lessen symptoms or AHR.
20 Allergy Essentials

Inhaled allergens have greater therapeutic potential. TSLP is an upstream IL-7–


like cytokine that may initiate and propagate allergic immune
responses and plays an important role in immune responses to
Epithelial cells
CCL11 helminths. TSLP is produced predominantly by airways and
Mast cell SCF nasal epithelial cells and by skin keratinocytes and also stimu-
lates immature mDCs, which express the heterodimeric TSLP
Histamine receptor to differentiate into mature DCs. TSLP-activated DCs
TSLP
cys-LTs promote naive CD4+ T cells to differentiate into a Th2 pheno-
PGD2 IL-33 type and promote the expansion of Th2 memory cells through
CCL17 Dendritic
CCL22 cell the release of Th2 chemotactic cytokines CCL17 and CCL22
Bronchoconstriction and expression of the costimulatory molecule OX40 ligand. In
IL-9
CCR4 ↓Tregs addition, TSLP suppresses the IL-12 p40 receptor in DCs and,
by suppressing Th1 responses, further enhances Th2 responses.
IL-4, Th2 cell TSLP also promotes allergic inflammation by activating the dif-
IgE IL-13 ferentiation IL-4 gene transcription in Th2 cells and the pro-
IL-5
CCR3 duction of IL-13 from mast cells, by recruiting eosinophils and
by amplifying responses of basophils. TSLP may therefore play
a pivotal role in the initiation of allergic asthma, rhinitis, and
B lymphocyte
AD. It is highly expressed in the airways of asthmatic patients,
Eosinophils
and its expression is correlated with disease severity and the
Fig. 1.13 Inflammation in allergy. Inhaled allergens activate expression of CCL17. TSLP is also expressed in epithelial cells
sensitized mast cells by cross-linking surface-bound immuno- of patients with AR and AD. Overexpression of TSLP in skin
globulin E (IgE) molecules to release several bronchoconstrictor keratinocytes of mice amplifies the inflammatory response of
mediators, including cysteinyl leukotrienes (cys-LTs) and prosta-
inhaled allergen in sensitized animals, thus providing a mecha-
glandin D2 (PGD2). Epithelial cells release stem cell factor (SCF)
(i.e., Kit ligand), which is important for maintaining mucosal mast nism for the “allergic march” whereby AD commonly precedes
cells at the airway or skin surface. Allergens are processed by the development of asthma in children.
myeloid dendritic cells, which are conditioned by thymic stro- IL-25 (IL-17E) is a member of the IL-17 family of cytokines
mal lymphopoietin (TSLP) secreted by epithelial cells and mast and induces allergic inflammation through increased produc-
cells to release the chemokines CCL17 and CCL22, which act on tion of Th2 cytokines. Although originally shown to be produced
CCR4 to attract T helper 2 (Th2) cells. Th2 cells have a central role by Th2 cells, it is now known to be released from many different
in orchestrating the inflammatory response in allergy through cells, including mast cells, basophils, eosinophils, macrophages,
the release of interleukin (IL)-4 and IL-13 (which stimulate B and epithelial cells. Blockade of IL-25 is effective in animal mod-
cells to synthesize IgE), IL-5 (which is necessary for eosinophilic els of allergic disease, and blocking antibodies are now in clinical
inflammation), and IL-9 (which stimulates mast cell proliferation). development. IL-33 is another upstream cytokine and a member
Epithelial cells release CCL11, which recruits eosinophils via
of the IL-1 family of cytokines, which is unusual in its localization
CCR3. Patients with allergic disease may have a defect in regula-
tory T cells (Tregs), which may favor further Th2 cell activation. within the nucleus, where it may regulate chromatin structure
CCL, C–C chemokine ligand; CCR, C–C chemokine receptor. and gene expression. It appears to be released only on damage to
epithelial or endothelial cells, presumably acting as an alarmin,
and is constitutively expressed at mucosal surfaces such as the air-
This observation suggests that blockade of individual cytokines ways. It signals through a receptor, ST2, that activates NF-κB and
may provide clinical benefit only in carefully selected patients. MAPK pathways. Its relevance to allergic inflammation is that it
Several proinflammatory cytokines have been implicated in enhances ILC2 and Th2 cell function, leading to eosinophilia, mast
allergic diseases, including IL-1β, IL-6, TNF-α, and GM-CSF, cell activation, and mucus hypersecretion, potentially acting as a
which are released from a variety of cells, including macro- bridge between innate and adaptive immunity in allergic inflam-
phages and epithelial cells, and may be important in amplifying mation. It also directly activates eosinophils, mast cells, epithelial
the allergic inflammatory response. Although available evi- cells, and DCs. It appears to switch alveolar macrophages to the
dence is persuasive that TNF-α may be important in patients alternatively activated form (M2) that has been found in animal
with severe asthma, and earlier small clinical studies with anti– models of asthma with increased secretion of CCL17, although
TNF-α therapies were promising, a large placebo-controlled whether this association is relevant to human allergic disease is
trial of an anti-TNF antibody (golimumab) in severe asthma uncertain. IL-33 shows increased expression in airway epithelium
showed no overall benefit. Some of the subjects may have been of asthmatic patients, and level of expression is related to disease
responders, however, and patients with greater bronchodilator severity. IL-33 is increased in the skin of patients with AD and is
reversibility showed an apparent reduction in exacerbations. released into the circulation during as well as mediating anaphy-
IL-17 also is increased in severe asthma, but anti–IL-17 anti- lactic shock. IL-33 also is expressed in mast cells after activation
bodies have not yet been tested in asthma patients. through IgE receptors and also activates mast cells, providing a
Interest has now focused on upstream regulatory cytokines means of maintaining mast cell activation. Antibodies that block
in the pathogenesis of asthma because it is thought that they may IL-33 or ST2 are now in clinical development.
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 21

the promotion of intestinal food allergy development in a Th2-


MICROBIOME AND IMMUNE SYSTEM
dependent manner before the establishment of immune toler-
Bacteria can stimulate or suppress inflammatory events in many ance.57,58 Despite the advancement in studies, the mechanism to
ways. Both bacterial cell wall components and some metabo- which allergic sensitization in the skin is able to disrupt oral
lites of the microbiome have been associated with immunoreg- tolerance and how it leads to the development of food allergy in
ulatory effects. Bifidobacterium, Lactobacillus, and Clostridium the gut remains unclear. It is proposed that the triggering effect
species have been shown to increase the proportion of Treg of food allergens stimulates TSLP, IL-33, and IL-25 production
cells in animal models. Moreover, Clostridia stimulates ILC3s in the skin keratinocytes and these alarmins in turn results in
to produce IL-22, which results in a strengthening of the epi- the activation of ILC2s and DCs.52,53,58–62 The migration of DCs
thelial barrier in the gastrointestinal tract. Bifidobacteria and to the lymph nodes triggers the proliferation of Th2 effector and
Lactobacilli increase the induction of Treg cells by promot- memory cells.32 Following the ingestion of sensitized food, these
ing metabolic processes such as vitamin A metabolism and Th2 cells are likely to migrate into the intestine and communi-
tryptophan metabolism in DCs. An exopolysaccharide from cate with ILC2s leading to the production of IL-13. Intestinal
Bifidobacterium longum has been shown to suppress Th17 epithelial cells also produce IL-33 and IL-25, further stimulating
responses in the gut and lung. Ingestion of B. longum by healthy ILC2s. As a result of this, an allergic immune response develops
human volunteers stimulated Foxp3+ Treg cells in peripheral towards the sensitized food.63,64
blood. Administration of this bacterial strain to patients with Tregs regulate the functions of ILC2s and suppress their type
chronic inflammatory diseases resulted in decreased levels of 2 cytokine production. Reciprocally, ILC2s secrete IL-4, which
serum proinflammatory biomarkers. Bacteria-derived metab- downregulates the Treg functions and increases the mast cell acti-
olites also have some effects on immunoregulatory processes. vation.65 In the steady state, this network functions towards the
Short-chain fatty acids (SCFAs) produced by the gut microbiota food tolerance side. However, some genetic and environmental
have been shown to affect DC and T cell functions by epigen- factors like microbiota dysregulation may stimulate alarmin pro-
etic mechanisms which are inhibition of histone deacetylases. duction from the intestinal epithelial cells, which results in ILC2
Biogenic amines produced by bacteria in the human gut can activation.66 Additionally, peanut allergens have been shown to
change immune and inflammatory responses. In recent studies, increase alarmin production leading to food allergy develop-
microbiota-originated taurine and histamine have been shown ment.67 ILC2s produce many Th2 cytokines like IL-4, IL-5, IL-9,
to influence host–microbiome interactions in different ways IL-13. IL-4, and IL-9 that amplifies the mast cells response.68 As a
such as co-modulating NLRP6 inflammatory signaling, produc- result, iTregs are inhibited by the effect of IL-4.65 After re-exposure
tion of epithelial IL-18, and suppressing the AMP production.49 to food allergens, activated mast cells can stimulate IL-33 produc-
tion and ILC2 activation. This forms a positive feedback loop on
mast cell activation and a negative feedback loop on iTregs, pro-
FOOD ALLERGY AS A MODEL FOR ALLERGIC moting the persistence of food allergy.69,70
Commensal bacteria also have some indirect effects on host
DISEASES immune responses towards food antigens. Some Clostridia
Food allergy frequently develops during infancy and this is strains induce the accumulation of Tregs in the colon.71,72
explained by the immaturity of the gastrointestinal mucosa.50 Clostridia also trıgger ILC3s to produce IL-22, strengthening
For this reason, exposure to food allergens early in life may the epithelial barrier. In mice, it was shown that Clostridia-
be protective towards the development of allergy, establishing containing microbiota suppresses the response to food allergy.73
oral tolerance before sensitization to the allergen. LEAP study In response to the microbial signals, macrophages secrete IL-1β
showed that early introduction of peanuts decreased the prob- which mediates GM-CSF release from ILC3s. GM-CSF induces
ability of peanut allergy development among children at high IL-10 and retinoic acid production by DCs and macrophages
risk and resulted in the induction of oral tolerance to peanuts.51 and subsequently promotes the induction of Tregs. Any inter-
Although GUT mucosa are continuously exposed to aller- ference with this crosstalk results in loss of oral tolerance to
gens and commensal microorganisms, the immune system food allergens.74
are mostly capable of tolerating these antigens. Induced Treg
cells (iTregs) and Tr1 lymphocytes play an important role in
this immune tolerance. Tolerogenic CD103+ DCs present the RESOLUTION OF ALLERGIC INFLAMMATION
luminal antigens and induce Foxp3+ Tregs in a TGF-β–depen-
dent and retinoic acid–dependent pathway.52,53 In children
AND MAJOR PATHWAYS
who outgrow or become tolerant towards cow’s milk allergy, Inflammation resolution is a tightly regulated and active pro-
the level of Tregs has been found to be higher than those with cess essential for the restoration of tissue homeostasis after an
active allergies.54 Genetic and environmental factors shape the inflammatory insult. Dysregulated resolution results in chronic
immune responses in the skin when an exposure to food aller- inflammation, tissue remodeling, and fibrosis. Granulocyte
gens occurs. In two epidemiologic studies, AD and the filag- apoptosis–mediated caspase family proteins are essential for the
grin gene mutation have been identified as potential risk factors clearance of these infiltrating inflammatory cells; cell survival
for the development of food allergy.55,56 In experimental food is increased during inflammation, and apoptosis is accelerated
allergy models, exposure of the skin to food allergens resulted in during the resolution phase.
22 Allergy Essentials

Macrophage
Therapies: recruitment
Survival factors Proapoptotic
(e.g., IL-5, IL-13, GM-CSF) • Glucocorticoids
• CDKi Eosinophil apoptosis

Phagocytosis
Th2 Eosinophils Proapoptotic factors
Allergen IL-10
response (e.g., IL-4, FAS, Therapies:
Siglec 8) TGF-β
Increased phagocytosis
• Proresolving lipids Proresolving
• Glucocorticoids lipids
Mast cells
Failure of resolution
Therapies:
• Exogenous
proresolving
T cell Tissue Fibrosis lipids
Migration
activation remodeling to lymphatics

Chronic inflammation Resolution of inflammation

Fig. 1.14 Inflammation resolution and therapeutic opportunities. After the initial Th2-mediated proinflammatory events that occur
in allergic inflammation and that are characterized by increased eosinophil recruitment, activation, and survival along with mast cell
degranulation, progression to the resolution phase of inflammation allows return of normal tissue structure and function. Increasing
proapoptotic factors drive eosinophil apoptosis for their timely clearance by macrophages, a process that is controlled by and
increases the production of proresolving lipids. Apoptosis can be enhanced through the use of glucocorticoids, which can also
increase the phagocytic capacity of macrophages and a variety of proresolving lipids. IL-10 released from a variety of cell types,
including macrophages, can indirectly attenuate eosinophil survival and promote resolution. CDKi, Cyclin-dependent kinase inhibitor;
GM-CSF, granulocyte-macrophage colony-stimulating factor; IL, interleukin; Siglec, sialic acid–binding immunoglobulin-like lectin;
TGF-β, transforming growth factor-β; Th2, T helper type 2 cell.

Phagocytosis of apoptotic cells by macrophages ensures the 3. Maeda K, Caldez MJ, Akira S. Innate immunity in allergy. Allergy.
safe disposal of dead and dying cells without release of toxic intra- 2019;74(9):1660–1674.
cellular mediators. Engulfment of apoptotic cells signals to the 4. Joiner KA, Brown EJ, Frank MM. Complement and bacteria:
phagocytosing macrophage that inflammation is coming to an chemistry and biology in host defense. Annu Rev Immunol.
end and alters macrophage mediator production from predomi- 1984;2:461–491.
5. Liu J, Cao X. Cellular and molecular regulation of innate
nantly proinflammatory to proresolution, with enhanced pro-
inflammatory responses. Cell Mol Immunol. 2016;13(6):711–721.
duction of cytokines with antiinflammatory properties, including 6. Palomares O, Ruckert B, Jartti T, et al. Induction and maintenance
IL-10 and TGF-β. This pattern contrasts with macrophage phago- of allergen-specific FOXP3+ Treg cells in human tonsils as
cytosis of necrotic eosinophils, which leads to enhanced proin- potential first-line organs of oral tolerance. J Allergy Clin
flammatory mediator production such as GM-CSF. Immunol. 2012;129(2):510–520, 520. e511–519.
Several proresolving lipids promote and control the resolution 7. Kaur BP, Secord E. Innate immunity. Pediatr Clin North Am.
phenotype. The delivery of exogenous protectins, lipoxins, and 2019;66(5):905–911.
resolvins has increased inflammation resolution and improved 8. Kubelkova K, Macela A. Innate immune recognition: an issue
clinical outcomes in a variety of allergic murine models. Advances more complex than expected. Front Cell Infect Microbiol.
in our understanding of proresolving lipids, granulocyte apopto- 2019;9:241.
sis, and phagocytic clearance of dead and dying cells are creat- 9. Mellor AL, Munn DH. Physiologic control of the functional status
of Foxp3+ regulatory T cells. J Immunol. 2011;186(8):4535–4540.
ing new avenues for generation of novel proresolving agents with
10. Pierce SK, Liu W. The tipping points in the initiation of B cell
which to tackle allergic inflammation (Fig. 1.14). signalling: how small changes make big differences. Nat Rev
Immunol. 2010;10(11):767–777.
REFERENCES 11. Macaubas C, DeKruyff RH, Umetsu DT. Respiratory tolerance
in the protection against asthma. Curr Drug Targets Inflamm
1. Castillo EF, Zheng H, Yang XO. Orchestration of epithelial- Allergy. 2003;2(2):175–186.
derived cytokines and innate immune cells in allergic airway 12. Metzger TC, Anderson MS. Control of central and peripheral
inflammation. Cytokine Growth Factor Rev. 2018;39:19–25. tolerance by Aire. Immunol Rev. 2011;241(1):89–103.
2. Lambrecht BN, Hammad H. The airway epithelium in asthma. 13. Komlosi ZI, Kovacs N, Sokolowska M, van de Veen W, Akdis
Nat Med. 2012;18(5):684–692. M, Akdis CA. Mechanisms of subcutaneous and sublingual
CHAPTER 1 Introduction to Mechanisms of Allergic Diseases 23

aeroallergen immunotherapy: what is new? Immunol Allergy 32. Wang YH, Ito T, Wang YH, et al. Maintenance and
Clin North Am. 2020;40(1):1–14. polarization of human TH2 central memory T cells by thymic
14. Globinska A, Boonpiyathad T, Satitsuksanoa P, et al. Mechanisms stromal lymphopoietin-activated dendritic cells. Immunity.
of allergen-specific immunotherapy: Diverse mechanisms of 2006;24(6):827–838.
immune tolerance to allergens. Ann Allergy Asthma Immunol. 33. Klunker S, Trautmann A, Akdis M, et al. A second step of
2018;121(3):306–312. chemotaxis after transendothelial migration: keratinocytes
15. Sindher SB, Long A, Acharya S, Sampath V, Nadeau KC. The use undergoing apoptosis release IFN-gamma-inducible protein
of biomarkers to predict aero-allergen and food immunotherapy 10, monokine induced by IFN-gamma, and IFN-gamma-
responses. Clin Rev Allergy Immunol. 2018;55(2):190–204. inducible alpha-chemoattractant for T cell chemotaxis toward
16. Penagos M, Durham SR. Duration of allergen immunotherapy epidermis in atopic dermatitis. J Immunol. 2003;171(2):
for inhalant allergy. Curr Opin Allergy Clin Immunol. 1078–1084.
2019;19(6):594–605. 34. Rebane A, Zimmermann M, Aab A, et al. Mechanisms of
17. Wang M, Yang IV, Davidson EJ, et al. Forkhead box protein IFN-gamma-induced apoptosis of human skin keratinocytes
3 demethylation is associated with tolerance induction in in patients with atopic dermatitis. J Allergy Clin Immunol.
peanut-induced intestinal allergy. J Allergy Clin Immunol. 2012;129(5):1297–1306.
2018;141(2):659–670. e652. 35. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell
18. Stanic B, van de Veen W, Wirz OF, et al. IL-10-overexpressing B populations (*). Annu Rev Immunol. 2010;28:445–489.
cells regulate innate and adaptive immune responses. J Allergy 36. Sonnenberg GF, Mjosberg J, Spits H, Artis D. SnapShot: innate
Clin Immunol. 2015;135(3):771–780. e778. lymphoid cells. Immunity. 2013;39(3) 622-622. e621.
19. Shamji MH, Durham SR. Mechanisms of allergen immunotherapy 37. Boonpiyathad T, Sozener ZC, Satitsuksanoa P, Akdis CA.
for inhaled allergens and predictive biomarkers. J Allergy Immunologic mechanisms in asthma. Semin Immunol.
Clin Immunol. 2017;140(6):1485–1498. 2019;46:101333.
20. Akdis M, Akdis CA. Mechanisms of allergen-specific 38. Schulzke JD, Gunzel D, John LJ, Fromm M. Perspectives on tight
immunotherapy: multiple suppressor factors at work in immune junction research. Ann N Y Acad Sci. 2012;1257:1–19.
tolerance to allergens. J Allergy Clin Immunol. 2014;133(3):621–631. 39. Matter K, Balda MS. SnapShot: epithelial tight junctions. Cell.
21. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, 2014;157(4) 992-992. e991.
differences, and implications of their inhibition. Am J Clin Oncol. 40. De Benedetto A, Rafaels NM, McGirt LY, et al. Tight junction
2016;39(1):98–106. defects in patients with atopic dermatitis. J Allergy Clin Immunol.
22. Fort MM, Cheung J, Yen D, et al. IL-25 induces IL-4, IL-5, 2011;127(3):773–786. e771–777.
and IL-13 and Th2-associated pathologies in vivo. Immunity. 41. de Boer WI, Sharma HS, Baelemans SM, Hoogsteden
2001;15(6):985–995. HC, Lambrecht BN, Braunstahl GJ. Altered expression of
23. Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin- epithelial junctional proteins in atopic asthma: possible role in
1-like cytokine that signals via the IL-1 receptor-related protein inflammation. Can J Physiol Pharmacol. 2008;86(3):105–112.
ST2 and induces T helper type 2-associated cytokines. Immunity. 42. Fujita H, Chalubinski M, Rhyner C, et al. Claudin-1 expression
2005;23(5):479–490. in airway smooth muscle exacerbates airway remodeling in
24. Khodoun MV, Tomar S, Tocker JE, Wang YH, Finkelman FD. asthmatic subjects. J Allergy Clin Immunol. 2011;127(6):
Prevention of food allergy development and suppression of 1612–1621. e1618.
established food allergy by neutralization of thymic stromal 43. Holgate ST. Epithelium dysfunction in asthma. J Allergy Clin
lymphopoietin, IL-25, and IL-33. J Allergy Clin Immunol. Immunol. 2007;120(6):1233–1244, quiz 1245–1236.
2018;141(1):171–179. e171. 44. Soyka MB, Wawrzyniak P, Eiwegger T, et al. Defective epithelial
25. Chen CC, Kobayashi T, Iijima K, Hsu FC, Kita H. IL-33 barrier in chronic rhinosinusitis: the regulation of tight
dysregulates regulatory T cells and impairs established junctions by IFN-gamma and IL-4. J Allergy Clin Immunol.
immunologic tolerance in the lungs. J Allergy Clin Immunol. 2012;130(5):1087–1096. e1010.
2017;140(5):1351–1363. e1357. 45. Xiao C, Puddicombe SM, Field S, et al. Defective epithelial barrier
26. Shen P, Roch T, Lampropoulou V, et al. IL-35-producing B cells function in asthma. J Allergy Clin Immunol. 2011;128(3):549–556.
are critical regulators of immunity during autoimmune and e541–512.
infectious diseases. Nature. 2014;507(7492):366–370. 46. Wang M, Tan G, Eljaszewicz A, et al. Laundry detergents and
27. Wang RX, Yu CR, Dambuza IM, et al. Interleukin-35 induces detergent residue after rinsing directly disrupt tight junction
regulatory B cells that suppress autoimmune disease. Nat Med. barrier integrity in human bronchial epithelial cells. J Allergy Clin
2014;20(6):633–641. Immunol. 2019;143(5):1892–1903.
28. Collison LW, Chaturvedi V, Henderson AL, et al. IL-35-mediated 47. Kipnis J. Multifaceted interactions between adaptive immunity
induction of a potent regulatory T cell population. Nat Immunol. and the central nervous system. Science. 2016;353(6301):766–771.
2010;11(12):1093–1101. 48. Barnes PJ. The cytokine network in asthma and chronic obstructive
29. Wirtz S, Billmeier U, McHedlidze T, Blumberg RS, Neurath pulmonary disease. J Clin Invest. 2008;118(11):3546–3556.
MF. Interleukin-35 mediates mucosal immune responses that 49. Sokolowska M, Frei R, Lunjani N, Akdis CA, O’Mahony L.
protect against T-cell-dependent colitis. Gastroenterology. Microbiome and asthma. Asthma Res Pract. 2018;4:1.
2011;141(5):1875–1886. 50. Sampson HA. Food allergy. Part 1: immunopathogenesis and
30. Vignali DA, Kuchroo VK. IL-12 family cytokines: immunological clinical disorders. J Allergy Clin Immunol. 1999;103(5 Pt 1):
playmakers. Nat Immunol. 2012;13(8):722–728. 717–728.
31. Layhadi JA, Eguiluz-Gracia I, Shamji MH. Role of IL-35 in 51. Du Toit G, Roberts G, Sayre PH, et al. Randomized trial of peanut
sublingual allergen immunotherapy. Curr Opin Allergy Clin consumption in infants at risk for peanut allergy. N Engl J Med.
Immunol. 2019;19(1):12–17. 2015;372(9):803–813.
24 Allergy Essentials

52. Schulz O, Jaensson E, Persson EK, et al. Intestinal CD103+, but 63. Shik D, Tomar S, Lee JB, Chen CY, Smith A, Wang YH.IL-9-
not CX3CR1+, antigen sampling cells migrate in lymph and serve producing cells in the development of IgE-mediated food allergy.
classical dendritic cell functions. J Exp Med. 2009;206(13): Semin Immunopathol. 2017;39(1):69–77.
3101–3114. 64. Lee JB.Regulation of IgE-mediated food allergy by IL-9 producing
53. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, et al. A mucosal mast cells and type 2 innate lymphoid cells. Immune
functionally specialized population of mucosal CD103+ DCs Netw. 2016;16(4):211–218.
induces Foxp3+ regulatory T cells via a TGF-beta and retinoic 65. Noval Rivas M, Burton OT, Oettgen HC, Chatila T. IL-4
acid-dependent mechanism. J Exp Med. 2007;204(8): production by group 2 innate lymphoid cells promotes food
1757–1764. allergy by blocking regulatory T-cell function. J Allergy Clin
54. Karlsson MR, Rugtveit J, Brandtzaeg P. Allergen-responsive Immunol. 2016;138(3):801–811. e809.
CD4+CD25+ regulatory T cells in children who have outgrown 66. Palmer G, Gabay C.Interleukin-33 biology with potential insights
cow's milk allergy. J Exp Med. 2004;199(12):1679–1688. into human diseases. Nat Rev Rheumatol. 2011;7(6):321–329.
55. Hill DJ, Hosking CS, de Benedictis FM, et al. Confirmation of 67. Li J, Wang Y, Tang L, et al. Dietary medium-chain triglycerides
the association between high levels of immunoglobulin E food promote oral allergic sensitization and orally induced
sensitization and eczema in infancy: an international study. Clin anaphylaxis to peanut protein in mice. J Allergy Clin Immunol.
Exp Allergy. 2008;38(1):161–168. 2013;131(2):442–450.
56. Lack G, Fox D, Northstone K, Golding J. Avon Longitudinal Study 68. Osterfeld H, Ahrens R, Strait R, Finkelman FD, Renauld JC,
of Parents and Children Study Team. Factors associated with Hogan SP.Differential roles for the IL-9/IL-9 receptor alpha-chain
the development of peanut allergy in childhood. N Engl J Med. pathway in systemic and oral antigen-induced anaphylaxis.
2003;348(11):977–985. J Allergy Clin Immunol. 2010;125(2):469–476. e462.
57. Noti M, Kim BS, Siracusa MC, et al. Exposure to food allergens 69. Lefrancais E, Duval A, Mirey E, et al. Central domain of IL-
through inflamed skin promotes intestinal food allergy through 33 is cleaved by mast cell proteases for potent activation of
the thymic stromal lymphopoietin-basophil axis. J Allergy Clin group-2 innate lymphoid cells. Proc Natl Acad Sci U S A.
Immunol. 2014;133(5):1390–1399. 1399. e1391–1396. 2014;111(43):15502–15507.
58. Tordesillas L, Goswami R, Benede S, et al. Skin exposure promotes 70. Hsu CL, Neilsen CV, Bryce PJ.IL-33 is produced by mast
a Th2-dependent sensitization to peanut allergens. cells and regulates IgE-dependent inflammation. PLoS One.
J Clin Invest. 2014;124(11):4965–4975. 2010;5(8):e11944.
59. Reese TA, Liang HE, Tager AM, et al. Chitin induces 71. Atarashi K, Tanoue T, Shima T, et al. Induction of colonic
accumulation in tissue of innate immune cells associated with regulatory T cells by indigenous Clostridium species. Science.
allergy. Nature. 2007;447(7140):92–96. 2011;331(6015):337–341.
60. Roediger B, Kyle R, Yip KH, et al. Cutaneous immunosurveillance 72. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by a
and regulation of inflammation by group 2 innate lymphoid cells. rationally selected mixture of Clostridia strains from the human
Nat Immunol. 2013;14(6):564–573. microbiota. Nature. 2013;500(7461):232–236.
61. Salimi M, Barlow JL, Saunders SP, et al. A role for IL-25 and IL- 73. Stefka AT, Feehley T, Tripathi P, et al. Commensal bacteria protect
33-driven type-2 innate lymphoid cells in atopic dermatitis. J Exp against food allergen sensitization. Proc Natl Acad Sci U S A.
Med. 2013;210(13):2939–2950. 2014;111(36):13145–13150.
62. Kim BS, Siracusa MC, Saenz SA, et al. TSLP elicits IL-33- 74. Mortha A, Chudnovskiy A, Hashimoto D, et al. Microbiota-
independent innate lymphoid cell responses to promote skin dependent crosstalk between macrophages and ILC3 promotes
inflammation. Sci Transl Med. 2013;5(170): 170ra116. intestinal homeostasis. Science. 2014;343(6178):1249288.
2
Precision Medicine
Gurjit K. Khurana Hershey, Michael G. Sherenian,
and Tesfaye B. Mersha

CHAPTER OUTLINE
Precision Medicine: A Brief History and Definition, 25 Nutrition, 30
Evidence-Based Versus Precision Medicine, 26 Vitamin D, 30
Components of Precision Medicine in Allergic Fatty Acids, 30
Disorders, 26 Respiratory Pathogens and Allergic Disease, 31
Endotypes/Biomarkers in Allergic Disorders, 26 Biologics and Precision Medicine, 31
Immune Profiling of Allergic Disorders, 27 Asthma, 31
Role of -omics, 27 Anti-IgE, 31
Genomics, 27 Anti–IL-5 or Anti–IL-5 Receptor, 31
Transcriptomics, 28 Anti–IL-4/IL-13, 32
Epigenomics, 29 Other, 32
Proteomics, 29 Atopic Dermatitis, 32
Metabolomics, 29 Chronic Rhinosinusitis With Nasal Polyposis, 32
Microbiome, 29 Eosinophilic Esophagitis (EoE), 33
Exposomics, 29 Food Allergy, 33
Comprehensive Integrative Omics, 29 Challenges in Precision Medicine, 33
Pharmacogenomics and Drug Dosing, 30 Summary, 33
Precision Medicine in the Management of Allergic References, 33
Disorders, 30

PRECISION MEDICINE: A BRIEF HISTORY U.S. National Research Council (US NRC) entitled Toward
Precision Medicine: Building a Knowledge Network for Biomedical
AND DEFINITION Research and a New Taxonomy of Disease.5,6 What all of these
Starting in the 19th century, developments in the basic and terms have in common is that they recognize that a “one-size-
clinical sciences allowed scientists to start understanding the fits-all” approach to medicine is not sufficient.
underlying causes and treatments of disease.1 In recent years, In January 2015, President Obama launched the Precision
the concept of personalized medicine has become part of the Medicine Initiative (PMI). PMI “is a long-term research
everyday language. However, medical practice has always endeavor, involving the National Institutes of Health (NIH) and
been about treating each patient based on personal character- multiple other research centers, which aims to understand how a
istics. Clinicians understand that patients respond differently person’s genetics, environment, and lifestyle can help determine
to specific therapies, and this makes it challenging to identify the best approach to prevent or treat disease.”5 The overarch-
what management strategy is appropriate for a given patient. ing goal of PM, despite its varied definitions,1,6–8 is to provide
Precision medicine (PM) offers the promise of improving prog- each patient with the most accurate therapy management based
nostication and diagnostic accuracy and enhancing individu- on genetics/genomics, environmental exposures, and lifestyle
alization therapy based on a patient’s genetics, environmental choices.9 This is being achieved through the national research
exposures, and lifestyle choices. The terms PM, personalized program called All of Us, which is currently recruiting at least
medicine, genomic medicine, stratified medicine, individual- one million volunteers from populations of diverse ancestry to
ized medicine, and P4 medicine (personalized, predictive, pre- help build a database of genetic information, biological samples,
ventive, and participatory) are often used interchangeably.2–4 and other health data that will be used to predict disease risk,
Although the term PM was first coined in 2008, it was not until understand how diseases occur, and improve the diagnosis
2011 that it became widely used following a report from the and treatment of medical conditions.10 The Trans-Omics for
25
26 Allergy Essentials

TABLE 2.1 Potential Benefits of Precision framework into allergic disorders, we allow for the potential to
Medicine generate similar outcomes as the cancer field. In this chapter,
we will briefly discuss the key components of the PMI. We will
Drug A national knowledge repository (with high-quality then focus on recent advancements made in PM for allergic dis-
developers genotype–phenotype data) would allow for the orders, including biologic therapies as a recent example of using
repurposing of existing drugs and the development of
PM approaches within allergic disorders.
novel therapeutics for specific subsets of populations
within allergic disorders.
Researchers Increasingly large integrated datasets of individual EVIDENCE-BASED VERSUS PRECISION
clinical and molecular data over time are becoming MEDICINE
available for interrogation. This will allow for
increased characterization of populations within Published practice parameters provide evidence-based medi-
allergic disorders. cine (EBM) guidelines for the diagnosis and management of
Medical Extensive information to identify specific subsets allergic disorders. For decades, the allergy community has col-
insurers of patients with allergic disorders will allow for lected demographic, immunologic, genomic, biomarker, and
the optimization of management algorithms that clinical data to generate disease phenotypes and endotypes.
mutually benefit the insurer and the patient. A Recently, novel technological advances enabled the rapid gen-
novel, potentially more expensive treatment might eration of increasing amounts of tissue- and cell-level molecular
be made available to patients who will more likely data. These data allow individuals with a given allergic disorder
respond to that specific treatment. This would allow to be placed into subgroups that define disease subsets based on
for increased cost-effectiveness of allergic disorder biologic pathways and pathogenic mechanisms. The synthesis of
management by (1) advocating use in patients who
this information allows PM to account for individual variability
are most likely to respond to the therapy and (2)
and then to apply the EBM principles to each allergic disorder
avoiding use in patients who are more likely to have
serious adverse effects. and its subtype. Expanding on an EBM approach, whereby the
best disease management strategy is determined for a hetero-
Healthcare New algorithms that tailor the development or use
geneous group of individuals with a given disorder, PM aims
providers of medical therapies to the key characteristics
to determine the best management strategy for each subset of
of population subsets will improve efficacy and
satisfaction while decreasing treatment failures that individuals stratified based on their molecular data (Fig. 2.1). In
result in healthcare utilization. contrast, by using the PM approach, clinical management and
prevention strategies can target the pathogenic mechanisms
rather than applying one type of treatment across heterogeneous
patients with different endotypes, but similar phenotypes, as is
Precision Medicine (TOPMed) Program is gathering omics data done using traditional EBM.18
across diverse populations, including those that have been tra-
ditionally underrepresented in research.11 The potential impact
of PM to improve the diagnosis of and alleviate the suffering COMPONENTS OF PRECISION MEDICINE IN
from allergic diseases is significant. This initiative and PM on
the whole is centered on four core values: predictive, preven-
ALLERGIC DISORDERS
tive, personalized, and participatory, which together are called Patients with allergic diseases present and respond to treatments
P4 Medicine.12 Some of the important potential short- and long- differently, which can create a major dilemma in providing
term benefits of PM are presented in Table 2.1. optimal management. Patients with asthma often share similar
While the initial goals of the PMI have focused primarily clinical symptoms, yet these similar presentations may opti-
on applying PM to cancer, its long-term goals include apply- mally respond to different treatments.19,20 Indeed, 40%–70% of
ing PM to all areas of health and healthcare, including allergic asthma treatments are considered to have absent or incomplete
diseases. Allergic disorders have significant potential to benefit efficacy,21 including responses to corticosteroids.21,22 This is due
from implementing PM practices. Its use in oncology and can- to underlying -omic (genetic, genomic, epigenomic, metabolo-
cer management provides a clear example of the successful use mic, etc.), immune, environmental, and microbiome differences
of the practice.13–16 For example, HER2-positive breast cancer among patients. Thus PM in allergy extends beyond genetic
patients are being targeted with specific therapies that reduce sequence analysis. Assays that measure multiple biomarkers
treatment time and speed recovery.14 Moreover, a meta-analysis can now be used to interrogate several key steps in the tran-
of phase II clinical trials of different cancers suggests that the scription (transcriptome), translation (proteome), regulation
use of a precision approach to select participants, based on sub- of gene expression (epigenome), host–microbial interactions
set characteristics, for specific trials resulted in increased thera- (immunome and microbiome), exposures and their metabolites
peutic response rates when compared with randomly selected (metabolome), and their combined synergies (multi-omics).
participants (p<0.001).16 The development of new precision
management strategies is made possible by integrating clinical, Endotypes/Biomarkers in Allergic Disorders
research, environmental, and lifestyle information—collectively Endotypes are characterized by the immunological, inflam-
known as information commons.17 By incorporating this matory, metabolic, and remodeling pathways that explain the
CHAPTER 2 Precision Medicine 27

Clinical
Phenotypes Patient Targeted
Endotyping Management

Environmental Metabolomics Epigenomics


factors Subgroup 1

Ancestry Proteomics Omics Genomics


Data Subgroup 2

Transcriptomics Subgroup 3
Heredity

Subgroup 4

Sex
Fig. 2.1 Conceptual diagram on how precision medicine can aid in generating targeted managements for allergic diseases. A patient’s
clinical phenotype will be integrated with -omics level data to identify their disease-specific endotype category and subgroup.
This endotype will then be used to identify which targeted management recommendations would lead to the best outcome(s) for a
given patient.

mechanisms underlying a clinical presentation (phenotype) of Th17 cells polarized with transforming growth factor-β (TGF-β)
a disease.23–26 In allergic disorders, endotypes are an important and IL-6 are nonpathogenic.57,58 Also, single-cell transcriptomic
tool for disease management. As part of determining endotypes, sequencing has recently been shown to identify unique subsets
biologic markers (i.e., biomarkers) are biological compounds of T cells associated with clinical disease.59,60 These findings
used to define different aspects of a disease such as severity or highlight the importance of accurate immune profiling in aller-
the likelihood of a response to a given treatment.24,27,28 Ideal bio- gic disorders. Characterizing patients by T cell phenotype will
markers share three important characteristics. First, they are likely become an important part of PM in allergic disorders.
stable over time. Second, they link disease endotypes and phe-
notypes to appropriately discern management strategies. Third, Role of -omics
they are detectable among populations with genetically distinct Through technological advances such as the use of high-
backgrounds.29 Several potential biomarkers have been identi- throughput assays that allow for hundreds of thousands of
fied for allergic disorders (Table 2.2).28 experimental samples to be processed simultaneously, next-
generation sequencing, computational biology, clinical bioin-
Immune Profiling of Allergic Disorders formatics, and genome-wide assay sequencing, these advances
The immune response is affected by several factors includ- are coming together in multi-omics form to increase the pre-
ing an individual’s immune predisposition based on underly- dictive accuracy of disease classification.61 The pathogenesis of
ing genetic characteristics, personal immune history, infection complex diseases such as allergy involves several cascades of
history, age, sex, and season. New technologies allow for the events at various levels of -omics as including transcriptomics
detailed study of the immune response in allergic disorders. of gene expression, epigenomics of gene regulation, proteomics,
These technologies can provide a means to determine “immune and metabolomics, which may have direct effects on disease
fingerprints” associated with the development of a given allergic endotypes. These methods are now being applied at the level
disease throughout an individual’s life spectrum. of the single cell, which is extremely powerful and reveals the
For example, the functional heterogeneity, and thus specific considerable heterogeneity within cell types.59
subpopulations, of T helper type 2 (Th2) cells drive specific Th2-
based pathologies.55 However, allergic disorders are increasingly Genomics
recognized as having additional T cell subtypes involved in their Genomic studies in allergy can be subdivided into different
pathogenesis. Allergic asthma is often characterized by a Th2 approaches: whole-genome sequencing (WGS), whole-exome
phenotype, with increased eosinophils and Th2 cytokine (e.g., sequencing, genome-wide association studies (GWAS), or can-
interleukin [IL]-4, IL-5, and IL-13) levels. Yet, severe cortico­ didate gene association studies. GWAS focus mainly on detecting
steroid-resistant asthma is often characterized by mixed Th2/ nucleotide polymorphisms (single-nucleotide polymorphisms)
Th17 responses.56 Moreover, like Th2 cells, Th17 cells also show across the whole genome. WGS is being increasingly applied to
considerable heterogeneity. Th17 cells that are polarized in vitro allergic disorders and yielding promising results for application
with IL-1β, IL-6, and IL-23 adopt a more pathogenic state, whereas as a key part of PM. The International HapMap Project,62 the
28 Allergy Essentials

TABLE 2.2 Established and Emerging Biomarkers for Major Allergic Disorders
Disease Source Marker Phenotype/Outcome Reference (s)
Asthma Blood, serum Serum IgE 30

Blood eosinophils T2-high asthma, lung function 31

Serum periostin T2-high asthma 32

Urine LTE4 Asthma severity, aspirin-sensitive asthma, 33

susceptibility to leukotriene receptor


antagonists
Metabolomic profile Asthma severity, corticosteroid-resistant 34–36

asthma, early-onset asthma


Exhaled breath Volatile organic compounds Eosinophilic asthma, neutrophilic asthma, 37,38

persistent asthma
Fractional exhaled nitric oxide Eosinophilic airway inflammation, response to 39

treatment
Sputum Eosinophils T2-high asthma, asthma severity, lung function, 31,40,41

predictor of exacerbations, response to


treatment
AD Blood, serum Serum TARC/CCL17 AD severity 42

DNA Filaggrin genotype Screening and prognostic biomarker for AD risk, 43

AD severity, early-onset AD
Skin Transcriptome profile Treatment response 44

Microbiome profile AD severity 45

Allergic rhinitis Skin prick test Allergen sensitization Diagnosis, distinguishes allergic from non- 46

allergic rhinitis
Nasal secretions TARC/CCL17, endothelin-1 Distinguishes allergic from non-allergic rhinitis 47

Nasal lavage following Eosinophils, IL-5, IL-6, Diagnosis, monitoring of treatment 48

nasal allergen challenge macrophage inflammatory


protein
Food allergy Skin prick test Allergen sensitization Diagnosis 49

Blood, serum Allergen-specific IgE levels Diagnosis 49

Blood Basophil activation test Diagnosis 50

Plasma Mast cell activation Diagnosis 51,52

Blood FOXP3 methylation in antigen- Predictive of response to oral immunotherapy 53

induced Treg cells


Tissue, serum Eotaxins Disease activity 54

Eosinophilic Tissue, serum IL-5, IL-13 Disease activity


esophagitis
Tissue, serum Eosinophil-derived neurotoxin Disease activity
Tissue, serum Transcriptomics Disease activity, remission
Tissue Mast cell quantification Disease activity
AD, Atopic dermatitis; IgE, immunoglobulin E; IL, interleukin; LTE4, leukotriene E4; TARC/CCL17, thymus- and activation-regulated chemokine/CC
chemokine ligand 17.

1000 Genomes Project,63 and the Exome Sequencing Project64 generally fail to take into account gene–gene and gene–environ-
along with publicly available databanks, including PubMed, ment interactions.68 In the future, integrating molecular bio-
EBI/Ensembl, University of California, Santa Cruz, Genome markers that help characterize patients into groups according
Browser, the National Human Genome and Research Institute, to pathogenic mechanisms of disease and more cost-efficient
among others, have helped shape a new era of research for many next-generation sequencing approaches will ultimately enable
diseases and disorders, including allergic disorders. A catalog genome-wide studies with the ability to yield more genetic
of almost all published GWAS can be accessed at https://www. causality.
ebi.ac.uk/gwas.65 According to the GWAS catalog, 85 studies
and more than 600 associations are related to asthma alone.66 Transcriptomics
Recently, an increasing number of GWAS involve other aller- Transcriptomic approaches in allergy encompass two main
gic disorders.67,68 One drawback of GWAS results is that they techniques: gene expression microarrays and RNA-sequencing
CHAPTER 2 Precision Medicine 29

(RNA-seq). The study of how genes are expressed during spe- (MS)-based methods (e.g., tandem MS, electron capture, or
cific conditions (e.g., health status, exposures, and disease state) electron-transfer dissociations), and protein microarray meth-
in different tissues and/or cells is known as transcriptomics. The ods. Like transcriptomics and epigenomics, it is important to
transcriptome varies from cell to cell, and to precisely analyze note that proteome collected from a single body compartment/
how the disease affects gene expression it is necessary to select site or at a certain moment (e.g., exacerbation, stable, etc.) will
tissues/cells directly affected from disease to be studied. In the not provide the information on the complete dynamic proteome
case of allergic disorders, transcriptomes from whole blood, neu- linked to the disease process. Thus comparative evaluation of the
trophils, CD4+ T cells, lung and airway tissues, airway smooth information collected from different sample sites and in differ-
muscle cells, induced sputum, and nasal lavage fluids have been ent disease conditions and/or times may be needed to get more
studied.69 For example, genome-wide profiling of bronchial epi- comprehensive information about the asthma–proteome link.
thelial brushings in individuals revealed two different asthma
phenotypes: “Th2-High” and “Th2-Low” based on microarray Metabolomics
expression of IL-5 and IL-13.22 These findings have been repli- Metabolomics concerns the study of low molecular weight
cated in nasal epithelial brushings,70 opening the possibility of organic compounds (50–1500 Da) that originate from human-/
using nasal airway gene profiles as biological markers for asthma microorganism-related metabolism and are involved in biologi-
diagnostics and treatment,70 which is very attractive, as this sam- cal processes.69,72 The techniques used in metabolomics belong
ple can be easily obtained without anesthesia at a clinical point mainly to gas or liquid chromatography coupled with MS.
of care. Recent approaches utilizing single-cell RNA-seq enable Another method is nuclear magnetic resonance (NMR) spec-
the characterization of all cell subpopulations. For example, a troscopy, which has lower sensitivity and specificity compared
recent study of asthmatic patients found that suboptimal asthma to MS-based techniques and therefore requires higher analyte
control was associated with signatures of eosinophilic and granu- concentrations.
locytic inflammatory signals, while optimal asthma control sig-
natures were associated with immature lymphocytic patterns.71 Microbiome
This study highlights the existence of specific, reproducible tran- The human body comprises at least 10 times more bacteria in
scriptomic components in the blood that vary with the degree numbers than human cells. Microbiome investigations are
of asthma control.71 Thus transcriptomic signatures from easily divided into two main approaches: 16S ribosomal RNA sequenc-
accessible tissue (nasal cells, blood, skin) could potentially be uti- ing and shotgun metagenomics. The former is less costly and
lized to monitor disease control and determine responsiveness to less computationally intensive compared to metagenomics, and
treatments including corticosteroids. therefore used more frequently. However, it has a lower potential
to detect microbial taxa up to species level. Data strongly sup-
Epigenomics port that dysbiosis contributes to the mechanistic underpinnings
Epigenetics study the changes in genetic functions that are not of the hygiene hypothesis that was first described in the 1980s.73
related to genetic alterations. Epigenetic variations are dynamic Indeed, site-specific dysbiosis has been shown to affect all aller-
and affected by environmental factors such as diet, chemical gic disorders.74 For example, skin bacteria dysbiosis is associated
compounds (including the use of medication), air pollution, with atopic dermatitis (AD) pathogenesis and outcomes.45,75–78 In
smoking, and others. Epigenetic studies in allergy often focus addition, airway dysbiosis has been associated with both asthma79
on changes in DNA methylation patterns, which can be exam- and allergic rhinitis.80 This dysbiosis can extend beyond the pri-
ined by epigenome-wide association studies. Other epigenetic mary affected organ of an allergic disease as well. Several stud-
studies may include the investigation of patterns of histone ies have shown that intestinal dysbiosis can lead to detrimental
modifications or noncoding RNAs. Together, these epigenetic immune-mediated outcomes including asthma and allergies.81,82
mechanisms regulate the gene expression program of a cell by
being responsive to changes in the environment of a cell. A com- Exposomics
pelling hypothesis is that environmental cues associated with Environmental exposure factors, which might influence asthma
diseases might initiate or influence the epigenetic processes of risk, are numerous, ranging from lifestyle-related factors such
host cells, leading to epigenetic reprogramming of host cells to as diet, smoking, exercise, and stress to environmental expo-
favor their pathogenic function and contributing to the devel- sures related to pollution, allergens, occupational factors, as well
opment of the disease.24,25 as many others. Linking the exposome to other-omics layers is
now a focus of allergy research. An example is the EXPOsOMICS
Proteomics consortium that investigates short- and long-term exposures
Proteins play a significant role in cellular processes, and their lev- to air and water contaminants and their biological effects
els reflect the momentary state of tissues/cells at the time of the on chronic diseases (including asthma) using a multi-omics
investigation. Analytical proteomic techniques allow the charac- approach.83
terization, identification, and quantification of proteins and their
associated functions. Different methods of proteomics are avail- Comprehensive Integrative Omics
able, which can roughly be subdivided into immunoassay-based Integrative omics (multi-omics) in allergy is the process of com-
methods (e.g., enzyme-linked immunosorbent assay [ELISA], bining the information of multiple-omics layers, to get more
immunohistochemistry, and Western blot), mass spectrometry insight into the disease process. Each omics data type typically
30 Allergy Essentials

provides a list of differential factors potentially associated with innate92,93 and adaptive immune function.94–96 For example,
the disease. These data can be useful as disease markers while vitamin D is also known to enhance skin barrier function,97
providing insight as to which biological pathways or processes antimicrobial peptide expression,98 and IL-10 production by
are different between the disease and control groups. However, regulatory T (Treg) cells.94,95
analysis of only one-omic(s) data type is limited to correlations Importantly for the clinician, multiple studies have investi-
and provides a partial view of the biological system. Integrating gated the influence of vitamin D (25OH-D3) levels on allergic
different-omics data types is often used to elucidate the poten- disorders. As a result, insufficient vitamin D levels have been
tial causative changes that lead to disease or can be used to iden- attributed to a variety of outcomes and pathogenic mechanisms
tify potential therapeutic targets for further molecular studies. in allergic diseases. In subjects with AD, seasonal low levels of
A large number of publicly available tools have been developed vitamin D have been associated with worse disease severity.99
for omics data integration.84 Moreover, AD severity has been associated with vitamin D lev-
els, whereby mild AD has higher levels than moderate or severe
Pharmacogenomics and Drug Dosing AD.100 Indeed, correcting vitamin D insufficiency among indi-
Pharmacogenomics is the study and application of genetic fac- viduals with AD has been shown to improve disease severity.101
tors relating to the body’s response to drugs. By predicting the In children, insufficient vitamin D levels have been correlated
drug response of an individual, the goal is to increase the suc- with asthma,102 including an inverse association between the
cess of therapies and reduce the incidence of adverse side effects. level of vitamin D and inhaled corticosteroid dose.103 Vitamin D
Currently, there are more than 130 medications that have labels supplementation in children with asthma has also been shown
from the Food and Drug Administration (FDA) with warnings to reduce the frequency of seasonal asthma exacerbations101,104
about possible serious implications based on genotypes for drug and the risk for respiratory tract infections.105 Studies have
response.85 There are at least 35 companies offering pharmaco- also reported a prenatal role for vitamin D in allergic disease
genetics (PGx) testing panels on different genes associated with development as decreased maternal vitamin D levels are cor-
the metabolism of drugs for various diseases (e.g., cardiovascu- related with an increased risk of childhood wheezing.106,107 Yet,
lar, diabetes, neurological, psychiatry).86,87 While no PGx testing other studies have shown the inverse correlation,108 suggest-
exists for allergic disorders currently, pharmacogenomic stud- ing a potential dynamic relationship between vitamin D and
ies targeting drugs used for the treatment of asthma are being prenatal asthma risk. Similarly, both increased and decreased
conducted and were summarized in a recent review.88 One rea- levels of vitamin D have been associated with increased immu-
son that pharmacogenomics has not already been successful noglobulin E (IgE) levels,109 although levels may vary with vita-
in allergic disorders is because it is difficult to interpret differ- min D supplementation.108 Vitamin D sufficiency in children
ential drug responses by “groups” when the “group” definition is associated with decreased odds of having allergen-specific
is imprecise, fluid, and time-dependent, such as the case with sensitizations.110 However, several studies have also shown that
asthma and other allergic disorders. vitamin D supplementation leads to an increased risk of aller-
gic disease development.111,112 With the increasing amount and
complexity of data, PM has the potential to guide management
PRECISION MEDICINE IN THE MANAGEMENT based on a patient’s vitamin D level in the context of other
OF ALLERGIC DISORDERS individual factors.

Nutrition Fatty Acids


Nutrition is a key area that can enhance PM in allergic disorders. PUFAs have also been shown to impact allergic disease. Omega-6
Vitamin D and omega-3 polyunsaturated fatty acids (PUFAs) (n-6) PUFAs and omega-3 (n-3) PUFAs have been shown to
are two examples of where PM can influence allergic disorders. have differential effects on the immune system.113,114 In general,
The following discussion is not meant to be a complete survey n-6 PUFAs contribute to a proinflammatory state113,114 and come
of the effects of nutrition on allergic disease, but rather to high- from sources such as corn, soybean, and sunflower oils—includ-
light key findings and convey where PM can make an impact on ing products made from these oils,115 whereas n-3 PUFAs are
patient care. considered antiinflammatory,113,114 with the major source com-
ing from oily fish.115 Indeed, n-3 PUFAs have direct immuno-
Vitamin D regulatory effects113,114,116 and have been shown to downregulate
One of the most studied nutrition factors within allergic disease both Th1 and Th2 responses.117 n-3 PUFAs can also decrease
has been vitamin D. Vitamin D3 is the most common nutri- antigen-presenting cell production of cytokines.118
tional source of vitamin D and is determined by measuring PUFAs have been studied on their impact on allergic dis-
serum 25-hydroxyvitamin D (25OH-D3) levels.89 However, the orders, particularly during childhood. For example, children
most biologically active form of vitamin D is that of 1,25-dihy- who regularly eat oily fish are significantly less likely to develop
droxyvitamin D (1,25[OH]2-D3) obtained through several asthma compared with their peers.118 Conversely, increased
mechanisms including sun exposure. Sufficient vitamin D levels consumption of n-6 PUFAs is associated with an increased risk
depend on several factors including age and race.90,91 Serum lev- of allergic disease.119 A diet high in n-6 PUFAs has been linked
els less than 30 ng/mL are considered insufficient.91 Vitamin D with increased risk of allergic diseases including asthma,120–122
affects several aspects of the immune system including both AD,121 and allergic rhinitis120,121,123 prevalence. While these data
CHAPTER 2 Precision Medicine 31

have been compelling, there is uncertainty around the true infection, one could anticipate the utility of PM in a situation
benefit of n-3 PUFA supplementation, as others have shown where this is possible, whereby PM identifies those individuals
an increased risk of allergic disease with increased n-3 PUFA with the highest risk of viral-induced wheeze based on genetic
intake.122,124 Several studies have shown that dietary supplemen- risk factors and identifies appropriate strategies to mitigate the
tation with fish oil decreases inflammatory mediators125,126 and risk of future asthma and allergic disease development.
leukocyte chemotaxis.126–128 In children, fish oil supplementa-
tion has been associated with improved asthma symptom scores
and reduced need for rescue medication.128,129 Furthermore,
BIOLOGICS AND PRECISION MEDICINE
dietary n-3 PUFA supplementation has been associated with Biologics are discussed briefly here in the context of PM.
improvement in peak expiratory flow measurements.129 Despite Briefly, a biologic is a targeted therapy manufactured using
data suggesting that n-3 PUFA intake may reduce the risk of living organisms rather than via chemical synthesis. Biologics
or improve outcomes related to allergic disease, current opin- are large, complex molecules consisting of proteins, peptides,
ion on best practice related to management remains unclear nucleic acids, sugars, other cellular structures, or a combina-
as other studies show no long-term benefit.120,130,131 Moreover, tion of these, produced within living cells or microorganisms.
a meta-analysis has shown no association between n-3 PUFA Biologics represent an increasingly important means to use PM
supplementation on allergic disease pathogenesis.132 PM can in allergic disorders (Fig. 2.2). Both small- and large-molecule
help provide the tools to appropriately make management deci- drugs interact with a patient’s biology; however, small-molecule
sions regarding those patients who will benefit the most from drugs work as inhibitors disrupting the process through pen-
n-3 PUFA supplementation. etrating cells, whereas large biologic drugs are designed to
There is also evidence that n-3 PUFA supplementation may bind to specific targets with extreme precision. Biologics target
act in disease prevention and that the immunomodulatory ben- inflammatory modulators that are important within a given
efits are relevant before disease onset. Children who receive fish allergic disease. Thus far asthma has the most available biologic
oil supplementation have shown a decrease in wheezing inci- therapies. A key area where PM could be useful in the rapidly
dence.133 Also, fish oil supplementation in pregnant women has developing landscape of biologic therapy for allergic diseases is
been shown to decrease IL-13 levels in umbilical cord plasma134 to identify which patients would benefit most from a given bio-
as well as IL-5, IL-10, and interferon-γ.135 Conversely, increases logic by using key patient biomarkers and other characteristics.
in dietary n-6 PUFA have been associated with increases in The discussion below presents a brief overview of biologics in
the above cytokines from umbilical cord blood.134 Infants with several allergic diseases and what biomarkers could be used in a
AD whose mothers received fish oil supplementation have also PM approach to patient management.
been shown to have less severe disease and less egg sensitization
than those whose mothers did not have supplementation.134 PM Asthma
could provide clearer insight into those populations who would Anti-IgE
benefit most from increased n-3 PUFA maternal dietary supple- Omalizumab was the first biologic used to treat allergic dis-
mentation to help reduce the risk of allergic disease develop- ease by targeting circulating IgE and preventing binding to its
ment in infants and children. receptor.143 Omalizumab significantly improves asthma out-
comes143,144; however, these effects are stronger in certain indi-
Respiratory Pathogens and Allergic Disease viduals based on their underlying characteristics. For example,
Early life wheezing due to viruses and other respiratory patho- patients with high fractional exhaled nitric oxide (FeNO >19.5–
gens is associated with later onset of asthma and allergic sensiti- 25 ppb) are more likely to have a positive response to omali-
zation.136–138 Indeed, preschool children who develop wheezing zumab.145 Also, patients with asthma and a baseline peripheral
during either viral or bacterial respiratory illnesses have an absolute eosinophil counts of at least 300 cells/μL are more
increased asthma risk. Respiratory syncytial virus (RSV) and likely to have reduced asthma exacerbations by 67% relative to
rhinovirus are two of the largest contributors to viral wheezing placebo and those with 400 cells/μL had reductions of 74%.146 In
illnesses in children.139 Treatment using RSV-specific mono- contrast, an observational study compared omalizumab treat-
clonal antibodies (mAbs) in preterm infants who have RSV ment with placebo and found that blood eosinophils, as well
infection has also been shown to reduce the incidence of later as FeNO and total IgE levels, did not predict the more likely
wheezing.140,141 Rhinovirus management presents a key oppor- response to omalizumab.145 This lack of evidence for eosinophil
tunity for PM using patient genotyping. Cadherin-related family levels was also observed in a second observational study.147 In
member 3 (CDHR3) is an entry factor for airway epithelial cells situations with conflicting results like this, PM could serve as
for rhinovirus C,142 one of the main types of serious rhinoviral a means to determine and then identify those individuals who
infections in infants.140 A CDHR3 polymorphism increases the would benefit from eosinophil-guided omalizumab therapy.
amount of CDHR3 present on cell surfaces and is thus associated
with an increased risk of childhood asthma.142 Therefore using Anti–IL-5 or Anti–IL-5 Receptor
PM techniques could identify these individuals as high risk for Anti–IL-5 antibodies such as mepolizumab, reslizumab, and
asthma development before disease onset and potentially alter benralizumab have also shown efficacy in treating asthma.148–150
management. Moreover, while no current preventative mea- These antibodies act by either binding and depleting IL-5 (mepo-
sures exist to ameliorate the risk of later wheeze after rhinovirus lizumab, reslizumab)151 or blocking the IL-5 receptor alpha chain
32 Allergy Essentials

Uncontrolled
Dupilimab CRSwNP Non-eosinophilic
disease Dupilimab
asthma
Increased
Reslizumab nasal Il-5 • FeNO >19.5–25
secretions Asthma • Peripheral eosinophils > 300 cells/µL Omalizumab

Peripheral
Comorbid eosinophils >
Benralizumab asthma EoE Mepolizumab
500 cells/µL
Pediatric Reslizumab
patients
• Nasal polyposis
Omalizumab • Prebronchodilator FEV1 <65% Benralizumab
• Maintenance oral steroid use
Food Allergy • ≥3 exacerbations in a year
• Asthma onset >18 years of age
Atopic
Dupilimab Peanut and milk oral
Dermatitis
immunotherapy

Omalizumab

Fig. 2.2 Phenotypic and endotypic influences on biologic therapies in patients with allergic disease. This figure illustrates select
examples of different endotypes and phenotypes and their associations with improved outcomes using a given biologic for several
allergic diseases. CRSwNP, Chronic rhinosinusitis with nasal polyposis; EoE, eosinophilic esophagitis.

(benralizumab).152 Either mechanism leads to a decrease in found that dupilumab improves AD outcomes.162 Dupilumab
eosinophil count. Importantly, the patient biomarker that best has also been shown to alter the AD transcriptome by upregu-
predicts response to any of these agents is blood eosinophilia, lating structural proteins, lipid metabolism proteins, and epi-
rather than sputum eosinophilia.153 Moreover, patients with dermal barrier proteins as well as downregulating inflammatory
eosinophil counts of 500 cells/μL or more show better improve- mediators, and markers of epidermal proliferation.44
ment in both pre- and post-bronchodilator FEV1 compared
with lower eosinophil counts.154 Also, in patients with high blood Chronic Rhinosinusitis With Nasal Polyposis
eosinophils, several other markers were predictive of benrali- While not yet standard-of-care in clinical practice, several bio-
zumab responsiveness: nasal polyposis, prebronchodilator FEV1 logic therapies have been developed for chronic rhinosinusitis
<65% predicted, maintenance oral corticosteroid use, three or with nasal polyposis (CRSwNP). Dupilumab has been shown
more exacerbations in the last year, and age of asthma onset of to improve outcomes in patients with uncontrolled CRSwNP.163
18 years or older.155 Therefore when considering a PM approach After treatment with dupilumab, individuals have been shown
to biologics, the provider would likely find more benefit from to have an improvement in sinonasal symptom scores, sense of
using blood eosinophil counts over those from patient sputum. smell, sinonasal imaging, nasal polyp size, and a decreased need
for systemic corticosteroids or sinus surgery.163,164 Omalizumab
Anti–IL-4/IL-13 has also been shown to be an effective biologic therapy for
Dupilumab binds to the IL-4 receptor alpha chain, which leads CRSwNP and can lead to improvements in nasal congestion, rhi-
to both IL-4 and IL-13 receptor signaling blockade156 and has norrhea, and a sense of smell.165 These findings were increased
shown to be effective in treating asthma.157 in subjects without atopic disease.166 Anti–IL-5 mAb therapies
have also shown efficacy in treating CRSwNP.167 Mepolizumab
Other has shown particular promise and has been found to decrease
Anti–IL-33 mAbs have shown promising results for the improve- nasal polyp size, decrease in need for sinus surgery, and improved
ment of asthma outcomes.158 Similarly, anti–thymic stromal congestion, postnasal drip, and sense of smell.167 Reslizumab has
lymphopoietin mAbs have shown promise in reducing asthma also been shown to reduce total nasal polyp scores, particularly
exacerbations, particularly in individuals with noneosinophilic among those individuals with increased IL-5 nasal secretions.168
asthma.159,160 Thus mAbs top the list when compared to other bio- Benralizumab is most effective in patients with both CRSwNP
logical drug types, such as proteins, enzymes, and vaccines. and asthma, with improvements in both diseases over patients
who only have either asthma or CRSwNP.169
Atopic Dermatitis Each of these therapies has the potential to be a part of PM
The only currently approved biologic therapy for AD is dupil- management. PM approaches could use patient biomarkers and
umab, an IL-4 receptor alpha-antagonist. IL-4 and IL-13 are two medical history to determine the medicine most likely to pro-
key cytokines involved with AD pathogenesis.161 Studies have duce beneficial results.
CHAPTER 2 Precision Medicine 33

Eosinophilic Esophagitis (EoE) they have applications for a wide range of diseases that are not
Mepolizumab has been trialed for use within EoE and is an being adequately addressed with existing medicines. The concept
effective treatment for decreasing overall eosinophilia and of clinical care practice to fit specific treatment to the individual
improving symptoms in adults with disease resistance to other patient in PM paradigm is an evolving concept becoming a reality.
therapies.170,171 However, while esophageal eosinophils decreased
in children, mepolizumab appears to not have a significant
effect on decreasing clinical symptoms in children.172 Similarly,
REFERENCES
reslizumab therapy in EoE was shown to decrease esophageal 1. US Food and Drug Administration. Paving the Way for
eosinophil counts but did not affect patient symptom scores.173 Personalized Medicine: FDA’s Role in a New Era of Medical
While one study showed no benefit of omalizumab therapy in Product Development. Silver Spring, MD: US Food and Drug
EoE,174 a second showed potential benefit, particularly in chil- Administration; 2013.
dren.175 These results, whereby a greater benefit is seen using 2. Khoury MJ, Gwinn ML, Glasgow RE, Kramer BS. A population
omalizumab in children, highlight the potential role for a PM approach to precision medicine. Am J Prev Med. 2012;42:639–645.
3. Hood L, Flores M. A personal view on systems medicine and
approach using biologics within EoE.
the emergence of proactive P4 medicine: predictive, preventive,
personalized and participatory. N Biotechnol. 2012;29:613–624.
Food Allergy 4. Roden DM, Tyndale RF. Genomic medicine, precision medicine,
The most promising biologic at this time for use within food personalized medicine: what’s in a name? Clin Pharmacol Ther.
allergy is omalizumab. Omalizumab has been shown to increase 2013;94:169–172. https://doi.org/10.1038/clpt.2013.101.
the tolerability of cow’s milk176,177 and peanut oral immuno­ 5. Genetics Home Reference. https://ghr.nlm.nih.gov/. Accessed
therapy.178 Further investigation is needed on relevant biomark- September 10, 2020.
ers within these populations to help guide PM approaches. 6. Redekop WK, Mladsi D. The faces of personalized medicine:
a framework for understanding its meaning and scope. Value
Health. 2013;16:S4–S9.
7. Ginsburg GS, Willard HF. Genomic and personalized medicine:
CHALLENGES IN PRECISION MEDICINE foundations and applications. Transl Res. 2009;154:277–287.
PM has significant potential for providing improved manage- 8. Abrahams E, Silver M. The history of personalized medicine.
ment in allergic disorders, but it is in its infancy. Notably, many Integr Neurosci Personal Med. 2010:3–16.
9. Juengst E, McGowan ML, Fishman JR, Settersten Jr. RA. From
challenges exist facing the progress and implementation of PM
“Personalized” to “Precision” medicine: the ethical and social
in everyday healthcare. Incredibly large amounts of data are implications of rhetorical reform in genomic medicine. Hastings
being accumulated creating a vast knowledge network, but con- Cent Rep. 2016;46:21–33. https://doi.org/10.1002/hast.614.
siderable work remains in mining and understanding the data 10. Denny JC, Rutter JL, Goldstein DB, et al. The “All of Us”
and finally interpreting it for the benefit of the healthcare prac- Research Program. N Engl J Med. 2019;381:668–676. https://doi.
titioner and patients and families. Although great progress has org/10.1056/NEJMsr1809937.
been made to technologies and platforms for use in the clinic, 11. Kowalski MH, Qian H, Hou Z, et al. Use of >100,000 NHLBI Trans-
there are still diagnostic, technical, and therapeutic challenges Omics for Precision Medicine (TOPMed) Consortium whole
that need to be taken into account and resolved.179 One of the genome sequences improves imputation quality and detection
major selling points of PM is that it is participatory, meaning of rare variant associations in admixed African and Hispanic/
that the patient will play a big role in the decision-making pro- Latino populations. PLoS Genet. 2019;15:e1008500. https://doi.
org/10.1371/journal.pgen.1008500.
cess. Thus additional tools are needed to discuss with patients
12. Hood L, Friend SH. Predictive, personalized, preventive, participatory
the individual-level data clearly and effectively. This presents (P4) cancer medicine. Nat Rev Clin Oncol. 2011;8:184–187.
a challenge in the sense that doctors will need to develop suf- 13. Hoberg-Vetti H, Bjorvatn C, Fiane BE, et al. BRCA1/2 testing
ficient skills and knowledge in genomics, pharmacogenomics, in newly diagnosed breast and ovarian cancer patients without
and related fields.180 There is also the challenge of economic fea- prior genetic counselling: the DNA-BONus study. Eur J Hum
sibility; while the costs are decreasing, they remain high. Many Genet. 2016;24:881–888. https://doi.org/10.1038/ejhg.2015.196.
people are opting to pay out of pocket to companies that sell 14. Incorvati JA, Shah S, Mu Y, Lu J. Targeted therapy for HER2
genomic and other tests directly to the consumer. Moreover, positive breast cancer. J Hematol Oncol. 2013;6:38.
other patients discard the possibility of testing completely 15. Druker BJ, Guilhot F, O’Brien SG, et al. Five-year follow-up of
because they are afraid they will be discriminated against based patients receiving imatinib for chronic myeloid leukemia. N Engl
on their genetic predispositions.181 Lastly, many ethical aspects J Med. 2006;355:2408–2417.
16. Schwaederle M, Zhao M, Lee JJ, et al. Impact of precision
including individual privacy issues have to be taken into consid-
medicine in diverse cancers: a meta-analysis of phase II clinical
eration when applying PM to practice. trials. J Clin Oncol. 2015;33:3817–3825. https://doi.org/10.1200/
JCO.2015.61.5997.
SUMMARY 17. Council NR. Toward Precision Medicine: Building a Knowledge
Network for Biomedical Research and a New Taxonomy of
Biologics are becoming the primary focus for a growing number Disease. Washington, DC: National Academies Press; 2011.
of therapeutic developers in the era of PM. Although they are both 18. Muraro A, Lemanske Jr RF, Hellings PW, et al. Precision medicine
challenging (produced in living cells) and expensive to develop, in patients with allergic diseases: airway diseases and atopic
34 Allergy Essentials

dermatitis-PRACTALL document of the European Academy of Immunol Allergy Clin North Am. 2018;38:599–610.
Allergy and Clinical Immunology and the American Academy https://doi.org/10.1016/j.iac.2018.06.011.
of Allergy, Asthma & Immunology. J Allergy Clin Immunol. 34. Carraro S, Bozzetto S, Giordano G, et al. Wheezing preschool
2016;137:1347–1358. https://doi.org/10.1016/j.jaci.2016.03.010. children with early-onset asthma reveal a specific metabolomic
19. Fassl BA, Nkoy FL, Stone BL, et al. The Joint Commission profile. Pediatr Allergy Immunol. 2018;29:375–382. https://doi.
Children’s Asthma Care quality measures and asthma org/10.1111/pai.12879.
readmissions. Pediatrics. 2012;130:482–491. https://doi. 35. Park YH, Fitzpatrick AM, Medriano CA, Jones DP. High-
org/10.1542/peds.2011-3318. resolution metabolomics to identify urine biomarkers in
20. Akinbami L, Centers for Disease Control and Prevention corticosteroid-resistant asthmatic children. J Allergy Clin
National Center for Health Statistics. The state of childhood Immunol. 2017;139:1518–1524.e1514. https://doi.org/10.1016/j.
asthma, United States, 1980-2005. Adv Data. 2006;381:1–24. jaci.2016.08.018.
21. Woodruff PG, Boushey HA, Dolganov GM, et al. Genome-wide 36. Papamichael MM, Katsardis C, Erbas B, Itsiopoulos C, Tsoukalas
profiling identifies epithelial cell genes associated with asthma D. Urinary organic acids as biomarkers in the assessment
and with treatment response to corticosteroids. Proc Natl Acad of pulmonary function in children with asthma. Nutr Res.
Sci U S A. 2007;104:15858–15863. https://doi.org/10.1073/ 2019;61:31–40. https://doi.org/10.1016/j.nutres.2018.10.004.
pnas.0707413104. 37. Cavaleiro Rufo J, Paciência I, Mendes FC, et al. Exhaled breath
22. Woodruff PG, Modrek B, Choy DF, et al. T-helper type condensate volatilome allows sensitive diagnosis of persistent
2-driven inflammation defines major subphenotypes of asthma. asthma. Allergy. 2019;74:527–534. https://doi.org/10.1111/
Am J Respir Crit Care Med. 2009;180:388–395. https://doi. all.13596.
org/10.1164/rccm.200903-0392OC. 38. Schleich FN, Zanella D, Stefanuto P-H, et al. Exhaled volatile
23. Lotvall J, Akdis CA, Bacharier LB, et al. Asthma endotypes: organic compounds are able to discriminate between neutrophilic
a new approach to classification of disease entities within the and eosinophilic asthma. Am J Respir Crit Care Med.
asthma syndrome. J Allergy Clin Immunol. 2011;127:355–360. 2019;200:444–453. https://doi.org/10.1164/rccm.201811-2210OC.
https://doi.org/10.1016/j.jaci.2010.11.037. 39. Menzies-Gow A, Mansur AH, Brightling CE. Clinical utility
24. Agache I, Akdis CA. Endotypes of allergic diseases and of fractional exhaled nitric oxide (FeNO) in severe asthma
asthma: an important step in building blocks for the future of management. Eur Respirat J. 2020;1901633. https://doi.
precision medicine. Allergol Int. 2016;65:243–252. https://doi. org/10.1183/13993003.01633-2019.
org/10.1016/j.alit.2016.04.011. 40. Hastie AT, Moore WC, Meyers DA, et al. Analyses of asthma severity
25. Desai M, Oppenheimer J. Elucidating asthma phenotypes and phenotypes and inflammatory proteins in subjects stratified by
endotypes: progress towards personalized medicine. Ann Allergy sputum granulocytes. J Allergy Clin Immunol. 2010;125:
Asthma Immunol. 2016;116:394–401. https://doi.org/10.1016/j. 1028–1036. e1013. https://doi.org/10.1016/j.jaci.2010.02.008.
anai.2015.12.024. 41. Licari A, Castagnoli R, Brambilla I, et al. Asthma endotyping
26. Anderson GP. Endotyping asthma: new insights into key and biomarkers in childhood asthma. Pediatr Allergy
pathogenic mechanisms in a complex, heterogeneous disease. Immunol Pulmonol. 2018;31:44–55. https://doi.org/10.1089/
Lancet. 2008;372:1107–1119. https://doi.org/10.1016/s0140- ped.2018.0886\\192.168.60.5\Public\Hari
6736(08)61452-x. 42. Thijs J, Krastev T, Weidinger S, et al. Biomarkers for atopic
27. Akdis CA, Ballas ZK. Precision medicine and precision health: dermatitis: a systematic review and meta-analysis. Curr
building blocks to foster a revolutionary health care model. Opin Allergy Clin Immunol. 2015;15:453–460. https://doi.
J Allergy Clin Immunol. 2016;137:1359–1361. https://doi. org/10.1097/ACI.0000000000000198.
org/10.1016/j.jaci.2016.03.020. 43. Carpagnano GE, Scioscia G, Lacedonia D, et al. Looking
28. Berry A, Busse WW. Biomarkers in asthmatic patients: for airways periostin in severe asthma: could it be useful for
has their time come to direct treatment? J Allergy Clin clustering type 2 endotype? Chest. 2018;154:1083–1090. https://
Immunol. 2016;137:1317–1324. https://doi.org/10.1016/j. doi.org/10.1016/j.chest.2018.08.1032.
jaci.2016.03.009. 44. Hamilton JD, Suárez-Fariñas M, Dhingra N, et al. Dupilumab
29. Agache I, Rogozea L. Asthma biomarkers: do they bring improves the molecular signature in skin of patients with
precision medicine closer to the clinic? Allergy Asthma moderate-to-severe atopic dermatitis. J Allergy Clin Immunol.
Immunol Res. 2017;9:466–476. https://doi.org/10.4168/ 2014;134:1293–1300.
aair.2017.9.6.466. 45. Kong HH, Oh J, Deming C, et al. Temporal shifts in the skin
30. Sonntag H-J, Filippi S, Pipis S, Custovic A. Blood biomarkers microbiome associated with disease flares and treatment in
of sensitization and asthma. Front Pediatr. 2019;7:251. https:// children with atopic dermatitis. Genome Res. 2012;22:850–859.
doi.org/10.3389/fped.2019.00251. https://europepmc.org/ https://doi.org/10.1101/gr.131029.111.
articles/PMC6593482, https://europepmc.org/articles/ 46. Fornadley JA. Skin testing for inhalant allergy. Int Forum Allergy
PMC6593482?pdf=render. Rhinol. 2014;4(Suppl 2):S41–S45. https://doi.org/10.1002/alr.21393.
31. Tiotiu A. Biomarkers in asthma: state of the art. Asthma Res 47. Tsybikov NN, Egorova EV, Kuznik BI, Fefelova EV, Magen
Pract. 2018;4:10. https://doi.org/10.1186/s40733-018-0047-4. E. Biomarker assessment in chronic rhinitis and chronic
32. van Rensen EL, Straathof KC, Veselic-Charvat MA, et al. Effect rhinosinusitis: endothelin-1, TARC/CCL17, neopterin, and
of inhaled steroids on airway hyperresponsiveness, sputum α-defensins. Allergy Asthma Proc. 2016;37:35–42.
eosinophils, and exhaled nitric oxide levels in patients with 48. Badorrek P, Müller M, Koch W, Hohlfeld JM, Krug N. Specificity
asthma. Thorax. 1999;54:403–408. https://doi.org/10.1136/ and reproducibility of nasal biomarkers in patients with allergic
thx.54.5.403. rhinitis after allergen challenge chamber exposure. Ann Allergy
33. Hoffman BC, Rabinovitch N. Urinary leukotriene E(4) as a Asthma Immunol. 2017;118:290–297. https://doi.org/10.1016/j.
biomarker of exposure, susceptibility, and risk in asthma: an update. anai.2017.01.018\\192.168.60.5\Public\Hari.
CHAPTER 2 Precision Medicine 35

49. Oriel RC, Wang J. Diagnosis and management of food identifies new risk loci for atopic dermatitis. Nat Genet.
allergy. Pediatr Clin North Am. 2019;66:941–954. https://doi. 2015;47:1449–1456. https://doi.org/10.1038/ng.3424.
org/10.1016/j.pcl.2019.06.002. 68. Meyers DA, Bleecker ER, Holloway JW, Holgate ST. Asthma
50. Hemmings O, Kwok M, McKendry R, Santos AF. Basophil genetics and personalised medicine. Lancet Respir Med.
activation test: old and new applications in allergy. Curr Allergy 2014;2:405–415.
Asthma Rep. 2018;18:77. https://doi.org/10.1007/s11882-018- 69. Kan M, Shumyatcher M, Himes BE. Using omics approaches to
0831-5. understand pulmonary diseases. Respir Res. 2017;18:149.
51. Bahri R, Custovic A, Korosec P, et al. Mast cell activation test in 70. Poole A, Urbanek C, Eng C, et al. Dissecting childhood asthma
the diagnosis of allergic disease and anaphylaxis. J Allergy Clin with nasal transcriptomics distinguishes subphenotypes of
Immunol. 2018;142:485–496.e416. https://doi.org/10.1016/j. disease. J Allergy Clin Immunol. 2014;133:670–8.e612.
jaci.2018.01.043. 71. Croteau-Chonka DC, Qiu W, Martinez FD, et al. Gene
52. Santos AF, Couto-Francisco N, Bécares N, Kwok M, Bahnson expression profiling in blood provides reproducible molecular
HT, Lack G. A novel human mast cell activation test for peanut insights into asthma control. Am J Respir Crit Care Med.
allergy. J Allergy Clin Immunol. 2018;142:689–691.e689. https:// 2017;195:179–188.
doi.org/10.1016/j.jaci.2018.03.011. 72. Turi KN, Romick-Rosendale L, Ryckman KK, Hartert TV. A
53. Syed A, Garcia MA, Lyu S-C, et al. Peanut oral immunotherapy review of metabolomics approaches and their application in
results in increased antigen-induced regulatory T-cell function identifying causal pathways of childhood asthma. J Allergy Clin
and hypomethylation of forkhead box protein 3 (FOXP3). Immunol. 2017;141:1191–1201.
J Allergy Clin Immunol. 2014;133:500–510. https://doi. 73. Strachan DP. Hay fever, hygiene, and household size. BMJ: Br
org/10.1016/j.jaci.2013.12.1037. Med J. 1989;299:1259–1260.
54. Bhardwaj N, Ghaffari G. Biomarkers for eosinophilic esophagitis: 74. Huang YJ, Marsland BJ, Bunyavanich S, et al. The microbiome
a review. Ann Allergy Asthma Immunol. 2012;109:155–159. in allergic disease: current understanding and future
https://doi.org/10.1016/j.anai.2012.06.014. opportunities-2017 PRACTALL document of the American
55. Wambre E, Bajzik V, DeLong JH, et al. A phenotypically and Academy of Allergy, Asthma & Immunology and the European
functionally distinct human TH2 cell subpopulation is associated Academy of Allergy and Clinical Immunology. J Allergy Clin
with allergic disorders. Sci Transl Med. 2017;9. https://doi. Immunol. 2017;139:1099–1110. https://doi.org/10.1016/j.
org/10.1126/scitranslmed.aam9171. jaci.2017.02.007.
56. Lloyd CM, Hessel EM. Functions of T cells in asthma: more 75. Kennedy EA, Connolly J, O’B Hourihane J, et al. Skin
than just T(H)2 cells. Nat Rev. 2010;10:838–848. https://doi. microbiome before development of atopic dermatitis: early
org/10.1038/nri2870. colonization with commensal staphylococci at 2 months is
57. Gaublomme JT, Yosef N, Lee Y, et al. Single-cell genomics associated with a lower risk of atopic dermatitis at 1 year.
unveils critical regulators of Th17 cell pathogenicity. Cell. J Allergy Clin Immunol. 2017;139:166–172. https://doi.
2015;163:1400–1412. https://doi.org/10.1016/j.cell.2015.11.009. org/10.1016/j.jaci.2016.07.029.
58. Wang C, Yosef N, Gaublomme J, et al. CD5L/AIM regulates 76. Baurecht H, Rühlemann MC, Rodríguez E, et al. Epidermal lipid
lipid biosynthesis and restrains Th17 cell pathogenicity. Cell. composition, barrier integrity, and eczematous inflammation are
2015;163:1413–1427. https://doi.org/10.1016/j.cell.2015.10.068. associated with skin microbiome configuration. J Allergy Clin
59. Papalexi E, Satija R. Single-cell RNA sequencing to explore Immunol. 2018;141:1668–676.e1616. https://doi.org/10.1016/j.
immune cell heterogeneity. Nat Rev Immunol. 2018;18:35–45. jaci.2018.01.019.
https://doi.org/10.1038/nri.2017.76. 77. Nakatsuji T, Chen TH, Narala S, et al. Antimicrobials from
60. Seumois G, Ramírez-Suástegui C, Schmiedel BJ, et al. Single-cell human skin commensal bacteria protect against Staphylococcus
transcriptomic analysis of allergen-specific T cells in allergy and aureus and are deficient in atopic dermatitis. Sci Transl Med.
asthma. Sci Immunol. 2020;5:eaba6087. https://doi.org/10.1126/ 2017;9:eaah4680. https://doi.org/10.1126/scitranslmed.aah4680.
sciimmunol.aba6087. 78. Meylan P, Lang C, Mermoud S, et al. Skin colonization by
61. Li CX, Wheelock CE, Sköld CM, Wheelock Å. M. Integration of Staphylococcus aureus precedes the clinical diagnosis of atopic
multi-omics datasets enables molecular classification of COPD. dermatitis in infancy. J Invest Dermatol. 2017;137:2497–2504.
Eur Respir J. 2018;51. https://doi.org/10.1183/13993003.01930- https://doi.org/10.1016/j.jid.2017.07.834.
2017. 79. Chung KF. Airway microbial dysbiosis in asthmatic patients:
62. Gibbs RA, et al. The international HapMap project. Nature. a target for prevention and treatment? J Allergy Clin
2003;426:789–796. Immunol. 2017;139:1071–1081. https://doi.org/10.1016/j.
63. Siva, N. Nature Publishing Group, 2008. jaci.2017.02.004.
64. Auer PL, Johnsen JM, Johnson AD, et al. Imputation of exome 80. Chiu C-Y, Chan Y-L, Tsai Yu-S, et al. Airway microbial diversity
sequence variants into population-based samples and blood- is inversely associated with mite-sensitized rhinitis and asthma
cell-trait-associated loci in African Americans: NHLBI GO in early childhood. Scientific Rep. 2017;7:1820. https://doi.
Exome Sequencing Project. Am J Hum Genet. 2012;91:794–808. org/10.1038/s41598-017-02067-7.
65. Hindorff L, Junkins H, Mehta J, Manolio T. A catalog of 81. Arrieta MC, Stiemsma LT, Dimitriu PA, et al. Early infancy
published genome-wide association studies 2010. Ref Type: microbial and metabolic alterations affect risk of childhood
Generic. 2011 asthma. Sci Transl Med. 2015;7:307ra152. https://doi.
66. Welter D, MacArthur J, Morales J, et al. The NHGRI GWAS org/10.1126/scitranslmed.aab2271.
Catalog, a curated resource of SNP-trait associations. Nucleic 82. Fujimura KE, Sitarik AR, Havstad S, et al. Neonatal gut
Acids Res. 2014;42:D1001–D1006. microbiota associates with childhood multisensitized atopy and
67. Paternoster L, Standl M, Waage J, et al. Multi-ancestry genome- T cell differentiation. Nat Med. 2016;22:1187–1191. https://doi.
wide association study of 21,000 cases and 95,000 controls org/10.1038/nm.4176.
Another random document with
no related content on Scribd:
tree, King Sound
XLIII 1. Carved boabab nut, King Sound. 2.
“Wanningi” from north-western Australia. 3.
Slate scrapers used by the extinct
Adelaide tribe for trimming skins 336
XLIV 1. Hand marks in cave, Port George IV, Worora
tribe. 2. Foot marks in cave, Port George
IV, Worora tribe 344
XLV 1. Cave drawings, Forrest River, north-western
Australia. 2. Decorating the body with
pipe-clay, Humbert River, Northern
Territory 352
XLVI Wordaman native with his body and head
decorated in imitation of skeleton and
skull, Victoria River, Northern Territory 356
XLVII 1. Cave drawings (kangaroo, etc.), Forrest
River, north-western Australia. 2. Cave
drawing of kangaroo, Forrest River, north-
western Australia 360
XLVIII Rock-drawings of archer fish (Toxotes),
Katherine River, Northern Territory 364
XLIX Ochre-drawings, Katherine River 368
L 1. Cave drawing of camel, north of Musgrave
Ranges, central Australia. 2. Cave drawing
of human figure, Glenelg River, north-
western Australia 376
LI 1. Ochre-drawings of mythic semi-human
creatures, Forrest River, north-western
Australia. 2. Sacred “Utnguringita” or
witchedy grub drawings, Emily Gap,
MacDonnell Ranges 384
LII Aluridja man rendering a musical
accompaniment with boomerangs 388
LIII Wordaman youth playing on the “drone pipe” or
“bamboo trumpet” 392
LIV 1. Making “vegetable down” by pounding grass 396
between two stones, Humbert River,
Northern Territory. 2. Worora native
making a stone spear-head, Northern
Kimberleys, Western Australia
LV 1. Wongapitcha man shaping a spear-thrower
with an adze. 2. Aluridja man scraping a
boomerang with a sharp stone flake 400
LIST OF FIGURES IN THE TEXT
No. Page
1. Map of Australia showing geographical distribution of
tribes 4
2. Peculiar “hand-like” feet of Berringin tribesman 11
3. Berringin women netting fish 130
4. Two Arunndta carvings of scenes in a dagger-duel 172
5. Types of spears 191
6. Sacred sun-design of the “Ilpalinja” ceremony 266
7. Stone phallus, Northern Kimberleys, Western Australia 284
8. Ochre drawing of “Kukadja” men, north of Wickham
River, Northern Territory 286
9. Charcoal drawing of a Kukadja man named
“Mongarrapungja” dancing at a sacred fire with an
ancestral female, Pigeon Hole, Victoria River 293
10. Rock carvings at Port Hedland 300
11. Rock carvings at Port Hedland 301
12. Rock carvings at Port Hedland 301
13. Sketch of reconstructed manus of Diprotodon
compared with tracing of carving of supposed
Diprotodon track at Yunta 307
14. Carved grave posts of Melville and Bathurst Islanders 310
15. Ochre drawing, Glenelg River, Western Australia 312
16. Carved crocodile design on boabab nut, Derby district,
Western Australia 313
17. “Dangorra,” the great emu in the southern sky 315
18. Boomerang with a number of emu designs carved upon
it, Pidunga tribe, Broome 317
19. Charcoal sketch of crows, Pigeon Hole, Victoria River 319
20. Pipe-clay cave-drawings of dancing figures, Humbert
River, Northern Territory 320
21. Charcoal drawing of hopping kangaroos, Pigeon Hole, 321
Victoria River
22. Bark-drawing depicting an eagle-hawk clawing and
tearing the carcass of a wallaby, Port Darwin 323
23. Pipe-clay drawing of man and dogs, Humbert River 324
24. Charcoal sketch of native hunting buffalo, Pigeon Hole,
Victoria River 325
25. Charcoal sketch of native spearing kangaroo, Pigeon
Hole, Victoria River 326
26. Carving depicting a quarrel between a man and his gin,
Arunndta tribe 328
27. Ochre-drawing of spear-boomerang duel, Arunndta
tribe 330
28. Charcoal sketch of ceremonial dance, Pigeon Hole,
Victoria River 332
29. Remarkable cave drawing, Glenelg River, N.W.
Australia 333
30. Pictograph of lizard, natural and conventional form 334
31. Normal, conventional, and emblematic representations
of turtle 335
32. Normal, conventional, and emblematic representations
of frog 335
33. Normal, conventional, and emblematic representations
of echidna 336
34. Conventionalized “Ladjia” or yam tjuringa pattern 337
35. A dog track 338
36. A kangaroo track 338
37. A rabbit track 339
38. Emu tracks 339
39. Pictographic representation of nesting emu 340
40. A lizard track 340
41. A snake or snake track 341
42. Human foot-prints and trail 342
43. “A man is tracking a rabbit.” Simple example of
pictography 344
44. Pictographic representation of emu hunt 344
45. Flying fox pattern 345
46. Conventional representation of hopping kangaroo 346
47. Crossed boomerangs, the symbolic representation of a
fight 347
48. Witchedy grub tjuringa, Arunndta tribe 348
49. Symbolic pictograph of kangaroo tjuringa, Arunndta
tribe 349
50. Symbolic pictograph of caterpillar tjuringa, Arunndta
tribe 350
51. Symbolic drawing of “native-pear totem,” Arunndta tribe 351
52. Ochre drawing and tree-carving of man with shield,
Humbert River 352
53. Human chain-pattern 353
54. Camps consisting of a man and his wife, and of eight
men 353
55. Anthropomorphous designs, carved on spear-throwers 354
56. Anthropomorphous design, carved on pearl-shell,
Sunday Island 355
57. Sign language of Arunndta tribe 391
CHAPTER I
INTRODUCTION TO AN AUSTRALIAN TRIBE

The lonely bush of Australia—The silence is broken—A mysterious call-note—A


human figure in the distance—Coo-ee!—A voluntary but cautious escort—The
official approach and salutation—Friendship established—Tribal introductions.

Let us imagine that we are travelling with a caravan over a lonely


tract of ground, in a remote district of the Australian bush, which has
not been contaminated by any disturbing influence of our civilization.
In consequence of the serenity and the deserted aspect of the scene
around us, we would be wondering whether the place holds any
mortal creatures but our party. We might even have resigned
ourselves to the inevitable desolation.
Suddenly the spell is broken by a faint sound falling upon our ears
—a long-drawn, shrill, yet melodious note—then all is silent again.
What could it have been? We are in doubt as to whether this was
the call of a bird or animal, or a phenomenon unknown to us; being
so far away from any centre of industry, a locomotive or factory
whistle is quite out of the question. Eagerly we wait for a possible
repetition of the singular sound.
Soon, indeed, it comes again; and, with the attentiveness our
expectation has solicited, we now recognize the human character of
the note. Presently it is repeated, then again, and yet again. But
where does it come from? So far we know not.
In the meantime we continue forging through the sand, and, being
now on the alert, we in due course espy, in the direction whence the
sound is coming, but a considerable distance off, a slim, dark figure
gliding from the cover of a rock to that of a bush. Presently it again
shows itself a short distance on; and our attention is further attracted
by the appearance of one or two other dark bodies running from
cover to cover in a line parallel to our course.
The calling is continued at frequent intervals; and, as near as we
can represent it by our alphabet, it sounds like the word “Coy!” with
the “y” specially emphasized and spun out. One has no difficulty in
recognizing in this call-note, which is met with all over Australia, the
derivation of the familiar “Coo-ee!”
It is, of course, assumed that we in no way betray a feeling of
uneasiness or give these dusky fellows the impression that we are
preparing for hostilities. Having satisfied themselves in respect of
this, and after manoeuvring for a considerable time in the manner
described, our uninvited escort become more trusting, even daring.
They run or walk for longer distances away from cover and gradually
bring their line of travelling closer in to ours. When eventually they
realize that we seem more like friend than foe, they drop behind our
caravan, and, at a measured distance, in our trail. Whenever we
move, they move, and when we stop, they stop. If any of our party
go towards them, they turn to the side and cleverly disappear into
the bush (Plate I).
When we have arrived at the end of our day’s journey and camp,
they pull up short and squat for a while; although it may not appear
so to the inexperienced traveller, they are taking stock of all our
doings.
Before very long, two men stand up and beckon to a small boy
among their party, who immediately jumps to his feet and walks to a
place in front of his elders. Each of the men now holds one of his
hands upon the lad’s shoulder, and, in that position, the little group
moves towards our camp.
In the hands not so used, both men are now seen to be carrying
small branches of Cassia bush, which they occasionally lift towards
us. Thus they walk to within fifty or sixty paces of our encampment
and again squat on the ground, arranging themselves in the same
order as they walked in. It is apparent they want us to approach
them; and we do so.
Statuary could not be more rigid than the persons we behold, upon
arriving at the little group. They sit silently, with downcast eyes, and it
is not until we address them that they rise to their feet. The old men,
for such we now recognize them to be, start a meek conversation,
nervously pushing the boy towards us. We are informed that this is
an uninitiated boy, a child in fact, a piccaninny in the tribal
relationship of son to the speakers. We are implored not to hurt this
innocent being, and to extend that friendship to themselves and to all
the members of the tribe. It is pointed out that they are unarmed and
that this is the guarantee of their goodly intentions towards us, the
unknown wanderers.
The men now advance and pat us on the chest, and instinctively
we return the compliment—for such it is intended to be—which is
akin to any ordinary European method of salutation. The bonds of
friendship have thus been sealed, and the men continue to jabber
profusely on the more intricate tribal relationships existing between
themselves and the rest of their party.
When the genealogical explanations have been concluded, the
men turn towards their company, who are still squatting in the
distance, and call aloud to them to come along: “Pitchai, ngalla
pitchai, waipella tami pu!” which in the Wongapitcha dialect stands
for “Come along, the white fellow is good.” The invitation is quickly
responded to, and ere many minutes have passed the whole group
has arrived, which includes other men as well as women and
children. The new arrivals, without hesitation, and with seeming
confidence, join in the conversation.
So this is our introduction to the aboriginal, the primitive hunting
man of Australia, and his family!
Fig. 1. Map of Australia showing geographical distribution of tribes.
PLATE I

Wordaman natives on the march.

“... they drop behind our caravan, and, at a measured distance, in our trail.”
CHAPTER II
RACIAL CHARACTERISTICS

Straightness of figure—Angular contours—Absence of fatty tissue—Nature’s


economy—Abnormal obesity—Straight spinal column—Flat and long dorsal
curve—Ensellure—Strong cervical curve—Uniformity of sacro-lumbar curve—
Flexibility of spine due to thick cartilaginous discs—Racial comparisons
between length of vertebræ and that of intervertebral discs—Influence of
upright attitude—Smallness of bones composing the spinal column and the
explanation—Exceptions to rule—Narrow sacral bone—Peculiarities of fifth
lumbar vertebra—Long extremities of equal length—Foot suggests tree-
climbing—Evolution of foot—So-called “hand-footed” men—Feet used for
lifting and carrying purposes—Function of peroneus muscle—Flatfoot rare—
Correct placement of foot when walking—Tree-climbing and its effects—
Peculiarity of Tasmanian’s foot—The shape and skeleton of lower limbs
—“Boomerang” legs—Shoulder and arm bones—Bodily height of male and
female.

Let us study these interesting-looking people more closely and


endeavour to find out their characteristic features, and in what
respects they differ from ourselves. At the same time, let us in
passing ascertain to what degree they resemble us and other
peoples, past or present, and what peculiarities they might share
with the man-apes or lower forms of the animal kingdom.
What immediately appeals to our critical eye is the strong contrast
in the general outline of the figure when we compare it with our own.
The round, full contours and shapely exterior of the European are
replaced by an angularity and straightness in the aboriginal. The
surface-padding or, more correctly, the subcutaneous deposition of
fatty tissue, which makes the lines and curves of our bodies so
uniform—and the female figure so beautiful—is, to a large extent, the
result of long selective culture and of the comforts which civilized life
has brought with it. In the case of the aboriginal, however, the
forethought of Nature has not allowed the development of such
paddings of fat to any considerable amount; they would only tend to
impede the agility of the hardy desert roamer. He does not need a
thick layer of fat beneath his skin. From an artistic point of view, the
leanness of his body is quite becoming to himself. He lives in a
country whose climate is hot, and his healthy hunting-life makes him
immune from many of the ills to which the city dweller is heir. As a
reserve storage of heat and nourishment, therefore, which might be
called upon to aid his physiological constitution in times of need, the
quantity of superfluous fat can safely be reduced to a minimum.
Nature has given enough, but not a measure in excess. Thus,
without any indication of unhealthy emaciation, the integumentary
accumulation of fat is so scant that parts of the internal anatomy of
an aboriginal can readily be deciphered topographically. We can
follow the shape of the superficial muscles and of the skeleton, and
can palpate the outline of the abdominal organs with comparative
ease. Although the muscles are small, they are, nevertheless,
strong, firm, and wiry; this is particularly noticeable in the extremities.
It is a curious fact, however, that there is a natural predisposition in
the aboriginal to produce fatty tissue once he gives up his active
hunting career, like a sportsman out of training, and to develop a
perceptible obesity when he lives under conditions which supply him
daily with an abundance of nourishment. Under such circumstances,
which are of course abnormal and only brought about by European
influence, his skin is very apt to accumulate locally masses of fat
known medically as lipomas. The Arunndta natives call these
tumours “lurra,” and connect their appearance upon their bodies with
heavy weight-carrying. It is, indeed, a noteworthy fact that these fatty
tumours frequently occur upon the shoulders of aboriginal wood-
carriers, who are in the habit of collecting logs of timber for camp or
station purposes.
Associated with the angularity and flatness of the bodily form, we
notice, when looking upon the figure of an aboriginal in profile, and
comparing it with that of an European in a similar position, a
straightness of the spine. If, for instance, we were to make an
accurate drawing of the spinal curvatures of the two subjects shown
in Plate II, we would find that the line representing the spinal column
of the aboriginal gin would be very straight in the centre of the back,
that is, in the dorsal segment; in fact it would be found that the dorsal
curve is very slight. Careful comparisons have been made with
frozen corpses of different races and the man-apes, bisected in the
mesial plane, and it has been determined that this portion of the
spine is flatter in the Australian aboriginal than in any other race of
man; and indeed it is flatter than in the chimpanzee. Moreover, a
larger number of vertebræ are involved in the dorsal curve of the
Australian than there are in the other cases.
In the European subject of our illustration—a young Australian
lady—the lumbo-sacral curve, known usually as the ensellure, is
unusually prominent.
If now we examine the curvature of that portion of the backbone
which constitutes the neck, we shall find it less pronounced in the
European but strongly developed in the Australian aboriginal.
But perhaps the greatest difference between the two types is the
manner in which the curvature breaks from the lumbar to the sacral
portions of the spine. In the European this break is sharp and
angular; in the Australian it is very gradual on account of a peculiar
oblique position of the last lumbar vertebra. Should we, again,
extend our observation to the chimpanzee, we would find that a
number of the sacral vertebræ are included in the lumbar curve.
Consequently the aboriginal’s spine seems to occupy an interesting
position in which the last lumbar vertebra stands almost as a
connecting link between the lumbar curve above and the sacral
curve immediately below it.
The lumbar curve is greater in the European than in the Australian,
but it is decidedly greater in the chimpanzee. The difference in
curvature is brought about mainly by the discs of cartilage which
exist between the vertebræ, and that is why there is a considerable
movement possible in the lumbar portion of the spine of the lower
races of man; and it is quite possible that the lumbar curvature alters
according to the position adopted by the individual, that is, according
to whether he be in a standing or in his favourite squatting posture.
In the European the corresponding portion of the spine is much more
rigid.
The proportional lengths of cervical, dorsal, and lumbar spinal
sections are much the same in both European and Australian, but
there are considerable differences in the two spines so far as the
proportions of the bony vertebræ and the intervertebral discs of
cartilage are concerned, especially in the lumbar region. The lumbar
vertebræ of the European are shorter than are those of the
Australian, and the latter again are shorter than those of the man-
apes. In other words, the lengths of the bones, which build up the
lumbar portion of the spinal column, increase (i.e. in proportion to the
size of the column as a whole) as one passes from the most highly
cultured European through the primitive human stages, like the
aboriginal of Australia, to the anthropoid apes, and finally to the
lower types of monkeys. At the same time, as the length of the
vertebræ increases, a reduction in the thickness of the cartilaginous
discs takes place.
There is no doubt this phenomenon depends to some extent upon
the acquisition of the upright attitude by man, since the cartilage
between the bone acts as an effective shock-absorber—the
percussion produced by the impact of the heel against the ground
when walking being reduced before it reaches the brain. When the
brain-box does not rest immediately above the point of percussion,
as for instance in the semi-erect posture of the apes, the dangers of
concussion are not so great.
While we are discussing the vertebral column of the Australian
aboriginal, we might draw attention to the comparative smallness of
the bones composing it. If we were, for instance, to compare the
column of an Australian with that of a European of similar height, we
would find that the vertebræ of the former are appreciably the
smaller—their volumes being almost in the ratio of one to one-and-a-
half. This is the more striking since we shall learn later that the
Australian aboriginal often is quite as tall in stature as the European.
The skeleton of the African negro, on the other hand, is decidedly
more massive than that of the European.
The smallness of the bones composing the vertebral column
undoubtedly favours the flexibility and agility which characterize the
Australians as a hunting people. There are, it is true, certain
variations in the structure of the spinal column of the Australian,
which seem to contradict this general rule, as, for instance, a slightly
stronger development of the vertebræ of the neck and a greater
volume of the lumbar vertebral bones in the female. The former of
these features is no doubt a primitive characteristic throwing back to
the quadrupedal ancestry of the human species, the latter having to
do with the processes of birth.
The sacral bone at the lower end of the vertebral column varies
slightly in size, but is, generally speaking, much narrower than that of
the European or of any other living race. It is principally on this
account that the hip-bones of the Australians seem remarkably close
together in both sexes.
The fifth lumbar vertebra of the Australian often exhibits certain
sacral characters, which remind one of the orang outang; in that
anthropoid the fifth lumbar bone is often fused to the os sacrum and
in reality becomes the first sacral body. Occasionally this vertebra is
asymmetrical, being normal, i.e. lumbar, on the one side and sacral
on the other. Its posterior arch is at times wanting, the spines having
failed to join, as ordinarily they do, in a median line behind the main
body of the bone. The last named feature is, however, not
infrequently observed in the skeletons of other races as well.
Another very striking feature, connected with the anatomy of the
Australian, is the great length of his arms and legs. This length of
extremities is taken in a conjoint sense, and with regard to the height
of the individual. The aboriginal is often said to have very much
longer arms than legs. This is incorrect. In point of fact, no human
type is known, living or fossil, with such a disproportion in the limbs.
All types of mankind, individually considered, have arms and legs
more or less equal in length; from this original condition the
elongated arms, so typical of apes, have evolved, by secondary
processes, in all probability through the acquisition of arboreal
habits. There is, of course, no doubt that the length of the
extremities, both upper and lower, so characteristic of the Australian,
together with the relative slenderness of the vertebræ, points to an
early evolutional stage, which was common to the ancestral forms of
both man and ape. The monkey has brought tree-climbing to such a
degree of perfection that it practically lives in the branches. Primitive
man, too, has not neglected the art, and, although the normal
proportions of his extremities do not directly suggest tree-climbing,
there is another development which does, especially in the
Australian; and that is his foot.
When we consider the likely transformations which the human foot
has undergone from an original hand-like form, resembling that of
certain monkeys and lower primates, to its present condition, we
shall find that two processes have been at work in the modelling of
this important part of man’s anatomy. Firstly, the big toe (originally a
thumb) has taken up a position adjacent to that of the next digit
(originally an index finger), and, by lying in the same plane with it,
has forfeited its power of opposition. Secondly, the big toe has grown
appreciably stronger, while the other digits have become smaller and
weaker. That the big toe, in its ancient evolution, once stood in the
same relation to the other toes as the thumb does to the fingers of
the hand, is evident from the arrangement of the blood-vessels and
nerves in this part of the foot, corresponding exactly to that of the
hand, even though the gap originally existing between the first and
second digits has been filled by fleshy tissue.
It is of considerable scientific interest to note that cases are
occasionally observed among the Australian tribes in which
indications of this ancestral condition are retained. In the Fig. 2 we
see the feet of an aboriginal of the Berringin tribe in the north of
Australia, whose big toes are remarkable for their shortness when
compared with the second.
Fig. 2. Peculiar “hand-like” feet of the Berringin tribesmen. Tracing
from a photograph.

It is, of course, a well-known fact that the newly-born European


baby possesses a wonderful mobility in its feet; and such might also
be acquired by people who have lost their arms; but the wearing of
boots usually deprives modern nations of this freedom of movement.
The aborigines of Australia make frequent use of their toes. A
considerable lateral flexibility of the end phalanges enables them to
lift small objects off the ground between the big and second toes.
Spears are carried by warriors, between the toes of either foot, to
conceal the weapons in the grass; and so the enemy is led to believe
that the men are unarmed.
When collecting firewood, the gins never stoop to pick up the
pieces, but lift them with their toes to the level of their hands. The
hands then pile the fuel upon the head and hold it there until
sufficient has been collected to carry back to camp.
The power of being able to use the toes in the manner described
depends upon the development of a muscle, which arising from the
outer side of the fibula and terminating in a long tendon, passes
obliquely across the sole of the foot, to insert itself into the
metatarsal bone of the great toe. This is the long peroneus muscle,
the function of which, in the monkeys at any rate, is to keep the big
toe in opposition. In man, moreover, this muscle helps considerably
to maintain the arch of the foot. Flatfoot is eminently rare among the
aborigines; only one or two cases have come under observation.
When walking, the aboriginal carries his foot so that it points
directly ahead of him, and not, as has been written, “with his toes
well turned out.” If anything, the sole of the foot is slightly tilted so
that the outer border touches the ground a little in advance of the
ball.
There is no doubt the outer surface and the ball of the foot play an
important role in the art of tree-climbing, as it is practised by the
Australians and other primitive peoples. Several methods are in
vogue; they will be described later. Suffice it, for the time being, to
refer to one: In pursuit of small marsupials, young birds, honey, nuts,
fruits, or any other things good to eat, the aboriginal often has
occasion to ascend the tall smooth trunks of trees, which harbour
such articles in abundance. This is done, often without the aid of any
implement, in the following way: The hunter faces the tree and
applies the palms of his hands to the opposite side of the butt. As he
tightens his hold with his fingers, he springs from the ground and
clutches the butt between the soles of his feet. The arches adjust
themselves to the convexity of the trunk, whilst the pressure of the
outer edges and balls of the feet prevent the limbs from slipping. In
this posture, the hunter is virtually hanging by his arms, which are
hooked by the hands, and is sitting upon his heels, which are fixed
firmly against the tree, as described. Holding his head well back
between the shoulder-blades, he suddenly lifts his body upwards
with his thighs, while his hands, momentarily relaxing their hold, are
pushed upwards also. Now the fingers again tightly clasp the trunk,
and the feet are quickly lifted and tucked under the buttocks, to again
support the weight of the body as before. The same actions are
repeated, time after time, and it is not long before the climber
reaches the nearest branches, when progress is simplified. Vide
Plate XIX, 2.
PLATE II

Comparison of European with Aboriginal figure.

“We notice, when looking upon the figure of an aboriginal in profile, and
comparing it with that of an European in a similar position, a straightness of the
spine.”
This ancient custom of tree-climbing is not peculiar to the
Australians, but is adopted by most primitive races. It is very
probable, too, that the prehistoric races were to a large extent
arboreal, and made use of similar methods of tree-climbing. When
considering the evolution of the human foot, therefore, we will have
to remember that it has been to some extent influenced by the tree-
climbing factor, which, indeed, must be considered in the light of a
forerunning stage in the acquisition of the upright attitude by man.
In this primitive method of ascending trees, by which the head is
thrown so far behind, we see also a likely explanation of the greater
cervical curvature we have noticed in the aboriginal’s spine, when
one compares it with the European’s. We might even venture to say
that these processes originally brought about the lumbar curvature,
and thereby laid the foundation to the acquisition of the erect
posture, by means of which man learned to balance his head upon
the vertical spinal column. Then the foot, which had been to a great
extent modelled through his arboreal activities, stood man in good
stead, and he began to walk erect between the trees.
The foot skeleton of the Tasmanian shows a peculiarity, in which it
differs from that of the Australian on the mainland. Under normal
conditions, the heel-bone of the Australian, and of the European as
well, has a small elongation or process on the anterior side which
separates the two adjoining small bones, the cuboid and the talus,
from each other. But in the case of the Tasmanian the two small
bones named lie in juxtaposition. This phenomenon is only
occasionally noticed in Australian skeletons, and is quite exceptional
in European; it is abnormal even in the anthropoids.
The Australian’s legs are often the subject of comment, if not
ridicule; they are so thin and lanky. Even when the proportions of the
chest and trunk as a whole are good, the legs usually remain
unshapely. Even under the best of conditions, there is a paucity of
flesh both in thigh and calf; the lower portions of the limbs are in the
true sense of the word spindle-shanks.
Even the gluteal musculature is only moderately developed.
Sedentary life and cosmetic culture seem to have been the principal

You might also like