Tissue factors are disease determinants in allergy

advertisement
UNIVERSITY MEDICAL CENTER UTRECHT
DEPARTMENT OF DERMATOLOGY AND ALLERGOLOGY
AND
DEPARTMENT OF IMMUNOLOGY
Tissue factors are disease
determinants in allergy
Jessica Wijngaarden
3734730
Infection and Immunity
Supervised by Dr Edward Knol
December 2012
Abstract
Over 40% of the western population is atopic however, only a limited percentage develops
allergic disease. Many chromosomal loci containing immune related genes have been associated
with allergic disease, but immune dysregulation alone cannot explain disease development. In
recent years the contribution of tissue restricted factors to development of allergic disease has
become a research focus. The skin and the airway epithelium are highly structured barriers as the
first line of defense against allergen entry and sensitization. Keratinocytes and epithelial cells are
also now recognized to play a driving role in allergic inflammation. Epidermal barrier breakdown due
to genetic polymorphisms and/or environmental factors can lead to the development of atopic
dermatitis (AD). Genetic polymorphisms in filaggrin, a key component of the stratum corenum and
lipid lamellae barriers of the skin, are associated with a higher risk of AD. Dysregulation of
desquamation due to Kallikrein-related peptidase-7, Lymphoepithelial Kazal-type-related inhibitor and
cystatin A polymorphisms and breakdown of the tight junction barrier due to claudin-1
polymorphisms are also associated with AD. Airway epithelial barrier integrity is essential in
protection against asthma development. This barrier depends on cell-cell adhesion, primarily
through tight and adherens junctions. Many adhesive proteins including claudin-1, occludin, zonula
occludens-1, e-cadherin, α-catenin and protocadherin-1 have been associated with asthma
development. Airway remodelling, a common feature of asthma, was believed to be caused by
chronic airway inflammation in the asthmatic lung; however there are recent indications that it also
has a genetic component. ADAM33 and claudin-1 have been associated with airway remodelling.
With the evidence that tissue dysregulation is at the base of allergic disease, therapeutic approaches
have shifted to address the underlying tissue irregularities rather than simply suppressing
inflammation. This shift opens the door for development of preventative therapies.
-2-
Index
Abstract .................................................................................................................................................. 2
Introduction ............................................................................................................................................ 4
Allergy and Atopy ....................................................................................................................................... 4
Tissue Role in Allergy .................................................................................................................................. 6
Role of the Skin in Allergy .............................................................................................................................. 7
Atopic Dermatitis ................................................................................................................................... 9
Introduction ................................................................................................................................................ 9
Tissue Factors ........................................................................................................................................... 11
Filaggrin ........................................................................................................................................... 11
Kallikrein-related peptidases........................................................................................................... 13
Lymphoepithelial Kazal-type-related inhibitor ............................................................................... 13
Cystatin A ........................................................................................................................................ 14
Claudin-1 ......................................................................................................................................... 15
Role of the Lung Epithelium in Allergy ........................................................................................................ 17
Asthma .................................................................................................................................................. 19
Introduction .............................................................................................................................................. 19
Tissue Factors ........................................................................................................................................... 20
ADAM33 .......................................................................................................................................... 20
Tight and Adherens Junctions ......................................................................................................... 22
Claudin-1............................................................................................................................ 22
Occludin ............................................................................................................................. 22
Zonula Occludens-1 ........................................................................................................... 23
E-cadherin .......................................................................................................................... 23
α-catenin............................................................................................................................ 24
Protocadherin-1 .............................................................................................................................. 24
Filaggrin ........................................................................................................................................... 25
Serine protease inhibitor, Kazal-type, 5 ....................................................................................... 25
Current Therapies and Future Perspective.......................................................................................... 26
Atopic Dermatitis ...................................................................................................................................... 26
Asthma ...................................................................................................................................................... 26
Conclusions ........................................................................................................................................... 28
References ............................................................................................................................................ 29
-3-
Introduction
Allergy and Atopy
Over the past 50 years the prevalence of allergic diseases in the developed world has
significantly risen.1-3 The hygiene hypothesis is commonly used to explain this phenomenon. Infants
naturally have a Th2 dominated immune response. As they develop and encounter microbial
antigens, their immune systems switch to a Th1 dominated response. The hygiene hypothesis states
that the lack of microbial encounters in the developed world results in a failure to induce a Th1
regulated immune system and allows for the development of Th2 dominated allergic diseases. This is
supported by decreased incidence of allergic disease in children who grow up on a farm or with
older siblings and presumably encounter more microbes.1,3
The classical view of allergic disease is based on immune dysregulation and atopy: a
predisposition to produce IgE antibodies in response to innocuous environmental stimuli.1,3 Allergic
diseases progress in three stages: (1) allergen sensitization followed by recurrent cycles of (2) acute
phase and (3) late/chronic phase responses upon allergen re-exposure. In atopic individuals, initial
allergen encounters lead to a sensitization response which primes the immune system to over-react
to subsequent encounters. The allergen is also processed by antigen presenting cells (APCs) and
presented on MHC-II molecules to induce a Th2 response. Polarization to a Th2 response requires an
initial Th2 environment, particularly IL-4; the source of this initial IL-4 is unknown, though it is
postulated to be produced either by naive CD4+ T cells in the absence of a strong Th1 inducer or by
infiltrating basophils.2 Th2 cell interactions with B cells through MHC-II, co-stimulatory molecules
such as CD40/CD40L and through secreted IL-4 and IL-13 drive the B cell isotype switch to produce
one of the primary hallmarks of allergic disease: IgE antibodies.1,4 IgE binds to the high affinity FcєRI
on mast cells, arming them to respond to subsequent allergen encounters. IgE also binds to FcєRI on
APCs where it functions to optimize antigen presentation.1
The acute phase response begins within minutes of antigen re-encounter due to crosslinking of mast cell bound IgE which triggers the release of pre-formed granules containing
histamine, cysteinyl leukotrines, proteases such as tryptase, and lipid mediators such as PGD2.1,4 IgE
cross linking also stimulates mast cell production of Th2 cytokines and various chemokines to recruit
Th2 cells, basophils and eosinophils: the mediators of the late phase response.1,3 Tissue resident
APCs take up, process and present allergens on MHC-II to memory Th2 cells activating them to
secrete a myriad of Th2 cytokines.5 Together these initiate the inflammatory immune response and
recruit basophils, eosinophils and more Th2 cells.1,3
The late phase response, which typically peaks within 6-9 hours of allergen exposure, is
characterized by an influx of immune effector cells, primarily basophils, eosinophils, CD8+ T cells and
CD4+ Th2 cells.1,3 Similar to mast cells, basophils are activated by cross-linking of surface bound IgE to
release pre-formed granules containing various inflammatory mediators, lipid mediators, proteases,
cytokines and chemokines. Basophil activation also triggers continued production of inflammatory
cytokines and chemokines.3 Eosinophils contain pre-formed granules which, upon activation, release
toxic proteins, inflammatory mediators and activators of basophils, mast cells, neutrophils and
eosinophils. Some of the eosinophil produced factors may also play a role in tissue repair.6 Th2 cells
can be activated at the site of allergen entry by tissue resident APCs or by activated APCs in the
lymph nodes. They are then recruited to the site of allergic inflammation by mast cell and basophil
secreted chemokines. Activated Th2 cells produce IL-3, IL-4, IL-5, IL-9, IL-13 and GM-CSF, which
perpetuate the inflammatory conditions2 (see Table 1 for a description of the soluble factors in
allergic disease).
Many atopic children initially present with atopic dermatitis and develop other atopic
diseases such as asthma and allergic rhinitis as they age. This progression of atopic disease is
referred to as the atopic march.7 Immune dysregulation is thought to be the driver behind the atopic
march. In support of this, many immune related genes have been associated with atopy and
-4-
Table 1 Key soluble factors in allergic disease
Factor
Produced by
Immunoglobulins
IgE
B cells (under Th2 env’t)
Phase in allergic disease
Function in allergic disease
Produced during
sensitization and
initiates acute phase
reaction
Recognizes allergens and initiates immune response
through mast cell and basophil activation and enhances
antigen processing by APCs1,4,9
Critical inflammatory mediator in anaphylaxis, urticaria
and rhinoconjunctivitis, but insignificant in asthma and
atopic dermatitis. Can cause an increase in vasodilation
to aid in effector cell recruitment.3,10
Th1,Th2 and Th17 attractant,5 causes vasodilation,
increased vascular permeability and in asthma: induces
smooth muscle contraction and increased mucous
production1
Upregulates epithelial and endothelial cell adhesion
molecule expression to attract basophils and
eosinophils1,3
Mediators
Histamine
Mast cells and basophils
Acute phase
Leukotrines
Mast cells and basophils
Acute phase
Tryptase
Mast cells
Acute phase
Th2 cells
Basophils, eosinophils
and Th2 cells
Late phase
Required during
sensitization, production
initiated in acute phase.
Primarily mediates late
phase response
Late phase
Cytokines
IL-3
IL-4
IL-5
Th2 cells
IL-9
Th2 cells, mast cells,
basophils and
eosinophils
Late phase
IL-13
Mast cells, basophils,
eosinophils and Th2 cells
Production initiated in
acute phase. Primarily
mediates late phase
response
IL-25
Late phase
GM-CSF
Th2 cells, mast cells,
basophils, eosinophils,
epithelial cells and
endothelial cells
Stored intra-cellularly in
epithelial cells and
produced by mast cells
Epithelial cells,
fibroblasts,
keratinocytes and mast
cells
Th2 cells
Chemokines
PGD2
Mast cells
Acute phase
Mast cells
Fibroblasts,
keratinocytes and
airway epithelial cells
Epithelial cells and mast
cells
Epithelial cells and mast
cells
Keratinocytes
Acute phase
Acute phase
IL-33
TSLP
CCL1
CCL5 (RANTES)
CCL17
CCL22
CCL27
Late phase
Late phase
Late phase
Involved in eosinophil survival1
Important in the initiation of the allergic Th2
response,10-11 drives isotype switch to IgE. Th2 survival
factor2 and mast cell development factor1,11 In asthma:
promotes mucous over-production1
Important eosinophil growth, differentiation and
survival factor1
Important eosinophil development factor and mast cell
development factor. In asthma: important in lung
inflammation, promotes airway hyper responsiveness,
and mucous over-production1-2,11
Drives isotype switch to IgE. Mast cell differentiation
and maturation factor. Eosinophil maturation and
survival factor, and involved in basophil recruitment.2 In
asthma: promotes airway hyper responsiveness and
mucous over-production1,11
Activates eosinophils and T cells.11
Activates mast cells (without triggering degranulation),
basophils, eosinophils, Th2 cells and B cells. Also aids in
skewing the T cell response to Th2.11
Important in activating DCs to polarize to Th2 response.
Enhances mast cell IL-13 production, and Th2 IL-4
production. Also involved in eosinophil recruitment and
basophil activation.11
Involved in eosinophil survival. In asthma: enhances
antigen presentation and recruitment of macrophages1
Th2 chemo-attractant and activator, also involved in
eosinophil and basophil recruitment and activation2,5
Th2 chemo-attractant5
Eosinophil and memory CD4+ T cell
chemo-attractant12-14
Acute phase
Th2 chemo-attractant2,5
Acute phase
Th2 chemo-attractant2,5
Constitutively expressed
Th1 and Th2 chemo-attractant15
-5-
allergic diseases. One of the most consistent associations is with chromosomal locus 5q23-33, which
contains the IL-4 cytokine gene cluster including IL3, IL4, IL5, IL9 and IL13.3,8
Tissue Role in Allergy
Approximately 40% of the western population is atopic but only a fraction develops allergic
disease.2 This, combined with evidence of non-atopic forms of allergic diseases points to something
other than only immune dysregulation at the core of these disorders.16-17 The lack of complete
responses to immune targeted therapies such as anti-IL-4, anti-IL-5, anti-IL-13 and anti-IgE antibody
treatments, recombinant soluble IL-4 receptor, and chemokine receptor antagonists has shifted the
focus from immune dysregulation to the role of the tissue in allergic disease.2,4,18 Recent evidence
has shown that tissue restricted factors play a central role in initiation and perpetuation of allergic
diseases. The skin and the lung epithelium provide the primary physical and chemical barriers
against allergen entry and sensitization.17,19 Barrier disruptions allow allergen entry and interaction
with immune effector cells initiating the allergic immune response. Allergen interaction with innate
immune receptors on epidermal keratinocytes and lung epithelial cells propagates allergic
inflammation.19-20 As discussed below, many epidermal and epithelial restricted genetic
polymorphisms have been associated with the development and severity of atopic dermatitis and
asthma respectively, reinforcing the importance of an intact barrier in the protection against allergic
disease.
-6-
Role of the Skin in Allergy
Skin structure
The skin is a multi-level barrier against water loss and entry of unwanted or harmful matter,
such as pathogens or allergens. The barrier includes the stratum corneum (SC), the lipid lamellae,
the acid mantle and the tight junction barrier (see Figure 1).17,21 The stratum corneum, the outer
most layer of the skin, acts as the air-liquid barrier. It is composed of corneocytes, which are dead,
terminally differentiated keratinocytes. These corneocytes no longer contain intracellular organelles,
a nucleus or cell membranes .21 Their keratin skeleton has been bundled together by filaggrin to
flatten the cells into squames. Corneodesmosomes join the cells to form the SC barrier. Surrounding
the squames is a cornified envelope composed of cross-linked structural proteins including loricrin,
involucrin, filaggrin and small proline-rich proteins.17 The cornified envelope is linked to the
surrounding lipid lamellae through the keratin matrix to further enhance the barrier.17, 21-22 The lipid
lamellae is important in maintaining skin hydration and preventing the entry of soluble irritants. It is
composed of ceramides, cholesterol, fatty acids and cholesterol esters which are secreted during
differentiation from lamellar granules at the interface of the stratum granulosum (SG) and SC layers
of the skin.17,22 Lamellar granules also secrete anti-microbial peptides such as LL-37 and β-defensin
2.23 The acidic pH in the upper layers of the skin, termed the acid mantle, is an important component
of the skin’s barrier function. It is anti-microbial, helps maintain the lipid lamellae and regulates
desquamation: the shedding of the uppermost corneocyte layers.17 Desquamation is tightly
controlled by multiple pH-dependent KLK serine proteases, which breakdown the
corneodesmosomes to release the corneocytes from the lipid lamellae, and by protease inhibitors
which prevent excess barrier breakdown.17,22 The final line of defense in the skin is the tight junction
barrier joining the cells of the SG.21,24
Keratinocytes
Keratinocytes react to mechanical stress and environmental stimuli through surface toll-like
receptors (TLRs) and NOD-like receptors (NLRs) to produce a number of cytokines and chemokines
which, under certain conditions, can initiate an allergic immune response (see Table 2).25 Proteolytic
allergens can cause cell damage which immediately triggers keratinocyte secretion of potent
inflammatory cytokines IL-1, IL-18, TNF-α, and GM-CSF and upregulation of cell adhesion molecules
to attract immune effector cells.20,25 One of the major keratinocyte derived factors contributing to
the allergic response is TSLP. Allergen derived injury or allergen recognition by TLR2/6 and TLR5
induces TSLP secretion and its production can be enhanced by other TLR or NLR ligands, viral or
microbial co-morbidities, smoke, exhaust or chemicals.24,26 TSLP primes skin DCs to activate Th2 cells
and is therefore one of the main drivers of the Th2 allergic response.20,25-26 TSLP can also act
synergistically with keratinocyte produced IL-1 and TNF-α to induce mast cell production of Th2
cytokines.20 In addition, keratinocytes and skin fibroblasts produce eotaxin, RANTES (CCL5) and
CCL27 to recruit eosinophils to further enhance the allergic response.12,15
Following the induction of the allergic response, keratinocytes are involved in its
maintenance. They express histamine receptors, which upon stimulation increase inflammatory
cytokine production.20 The allergic inflammatory response negatively impacts keratinocyte barrier
function and increases production of TSLP and T cell recruiting cytokines.20,26 IFN-γ (produced in the
Th1 phase of atopic dermatitis) is a potent keratinocyte activator which induces cytokine and
chemokine production, up-regulation of antigen presenting molecules, costimulatory molecules and
adhesion molecules as well as the expression of Fas. Fas interacts with FasL on infiltrating T cells
causing keratinocyte apoptosis and breakdown of the skin barrier.12,20 Th2 inflammatory cytokines IL4 and IL-13 inhibit keratinocyte anti-microbial peptide synthesis which further disrupts the skin
-7-
barrier.12,22 See Table 2 for a list of keratinocyte produced factors and their function in allergic
disease.
Table 2 Keratinocyte derived factors contributing to the allergic immune response
Soluble factor
Production stimulated by
Function in allergic disease
Cytokines
IL-1(α and β)
Typically stored intra-cellularly but
Pro-inflammatory.25 Also promotes keratinocyte
released upon mechanical trauma and
differentiation and activates LCs27
20,27
barrier disruption
IL-18
mechanical trauma and barrier
Pro-inflammatory25 involved in Th1 cell proliferation and
disruption25
differentiation27
TSLP
IL-4, IL-13 and TNF-α. Barrier
directs skin DCs to induce a Th2 response20,25-26 also acts
disruption, TLR (TLR2/6 and TLR5) or
synergistically with IL-1 and TNF-α to induce mast cell
NLR ligands, viruses, microbes,
production of Th2 cytokines.20
20,26
chemicals, exhaust, smoke
TNF-α
Mechanical trauma and skin barrier
Increases adhesion molecule expression on endothelial
disruption20 and IFN-y and IL-1.27
cells. Can induce keratinocyte apoptosis leading to
barrier damage and increased immune response.27
GM-CSF
Chemokines
CXCL10
CCL5 (RANTES)
CCL17
CCL22
CCL27
CXCR3 ligands
Eotaxin
Autocrine IL-1α, TNF-α and T cell
produced IFN-γ and IL-4.12 Mechanical
trauma and skin barrier disruption20
Important skin DC survival and maturation factor. Upregulates CD80 and CD86 expression on LCs and may aid
in antigen presentation. Stimulates keratinocyte
proliferation. Production is increased in atopic
individuals.27
Th2 cytokines20
Allergen challenge13 and Th2 cytokines28
Th2 cytokines20
Th2 cytokines20
Constitutively expressed15
INF-y activation27
Th2 cytokines13
T cell recruitment20
Eosinophil recreuitment12-13
T cell recruitment20
T cell recruitment20
Binds CCR10 to recruit skin homing Th1 and Th2 cells15
attract activated T cells27
Eosinophil recruitment12
-8-
Atopic Dermatitis
Atopic dermatitis (AD) is a chronic inflammatory disease of the skin that typically appears
early in childhood and affects 15-25% of children worldwide.7,24,29 Approximately 40-50% of
childhood cases resolve with age but the remaining cases persist into adulthood.7 AD is
characterized by dry, itchy, erythematous lesional patches of skin concentrated on the face, neck
and extensor/flexural surfaces.7,17,29 In early childhood AD presents with increased edema leading to
an oozing and crusting appearance. After about one year the skin dries out and presents as more
typical AD.7
AD progresses from an acute, recurrent lesional-phase disease to chronic lesional disease.
During the acute phase encountered allergens are captured by Langerhans cell (LC) bound IgE. LC
bound IgE enhances allergen uptake and presentation to tissue resident memory Th2 cells activating
them to secrete Th2 cytokines and initiate allergic inflammation.28 Allergens can also trigger
keratinocyte production of inflammatory cytokines and chemokines.12,20 The Th2 allergic
environment leads to an upregulation of cell adhesion molecules, inflammatory cytokines and
chemokines which perpetuates recruitment of inflammatory cells such as basophils, eosinophils and
Th2 cells. Infiltrating basophils and eosinophils also contribute to the inflammatory environment.
Infiltrating T cells are activated by tissue resident APCs which drive a Th2 response leading to
increased secretion of IL-4, IL-5 and IL-13.12,20,29
Initially, AD is a Th2 mediated disease; however, during the chronic phase it switches to a Th1
mediated disease. This switch is mediated by IL-12 which drives T cell production of IFN-γ leading to
a Th1 dominated response. Although the source of IL-12 is unknown, it may be produced by
keratinocytes, Th2 stimulated eosinophils, or inflammatory dendritic epidermal cells.6,20 The
increased IFN-γ in the chronic phase induces keratinocyte expression of Fas which interacts with FasL
on the infiltrating T cells, triggering keratinocyte apoptosis. This initiates increased production of Th1
chemokines resulting in increased keratinocyte apoptosis, perpetual skin barrier damage and chronic
Th1inflammation.12,20 Macrophages and eosinophils continue to infiltrate the lesional tissue and
maintain the inflammatory environment.12 Lesional tissue in chronic AD undergoes remodelling in
which the skin thickens and becomes lichenified.7,20 In acute and chronic AD the non-lesional skin is
also affected; it is characterized by mild hyperplasia and T cell infiltration as well as increased
numbers of tissue resident mast cells.12,20
Similar to other allergic diseases, AD is highly heritable. Genome wide scans have identified
many chromosomal regions linked to disease risk; however, untangling the specific underlying genes
is a challenge.24,29 More than 100 candidate gene studies have reported associations with AD.
However, these studies are often under powered and determining true associations is difficult. Many
immune related genes, including IL4, IL4RA, IL5, IL13, RANTES, IL18, CD14, have been repeatedly
associated with AD, indicating the importance of a misdirected immune response in allergic
disease.24 Environmental factors also play an important role in AD. Proteolytic allergens can
breakdown the skin barrier and trigger an inflammatory response through keratinocyte innate
PRRs.25,30 Bacterial and viral infections complicate AD. Over 90% of AD patients are infected with
Staphylococcus aureus, compared to only 5-30% of non-AD patients. This is likely due to genetic and
environmental breakdown of the skin’s anti-microbial barrier.24 S. aureus toxins can act as
superantigens which potently activate tissue resident mast cells and T cells, further perpetuating the
inflammatory environment and epidermal barrier breakdown.7,12,24,29,31
The course and severity of AD is determined by a combination of underlying genetic factors,
the immune response mounted against a specific set of allergens, and various environmental factors;
however, the tissue specific genetic factors are pivotal in the initiation of disease. AD skin is
intrinsically defective as a physical and chemical barrier against allergen entry which allows allergen
sensitization and perpetuation of the allergic inflammatory response.24 The following is a summary
of some of the common tissue factors that have been associated with AD in recent years (see Figure
1).
-9-
A
SC Barrier
FLG
KLK7
SPINK5
CSTA
SC
SG
TJ Barrier
CLDN1
Stratum spinoza
Stratum basale
Basement membrane
Dermis
NMFs
Filaggrin degradation
Filaggrin monomers
B
Corneodesmosome
Desquamation
SC
pH
(approx)
pH mantle
FLG
KLK
LEKTI
SG
TJ Barrier
Lipid Lamellae
FLG
Figure 1 Schematic representation of the skin barrier and the tissue factors associated with atopic dermatitis. (A) Genetic
polymorphisms in genes involved at each layer of the skin have been associated to AD. These polymorphisms result in
barrier dysfunction. (B) The epidermis is a highly stratified, multi-layer barrier. (i) The tight junction barrier is located at the
SG-SC interface. (ii) Lamellar granules are secreted outside of the TJ barrier and play a role in the SC barrier. (iii) SG1 cells
lose their TJs as they undergo (iv) terminal differentiation into corneocytes. (v) The mature corneocytes are surrounded by
a cornified envelope and the lipid lamellae (both part of the SC barrier). (vi) Corneodesmosomes anchor the corneocytes
together adding to the barrier strength. (vii) pH differences in the upper SC control the activation of KLKs and inhibition of
LEKTI allowing controlled barrier breakdown in the form of desquamation. Adapted from Kubo et al (2012)21
- 10 -
Filaggrin
Filaggrin is a multi-functional barrier protein found in the skin, oral and nasal mucosa and in
the conjunctiva.32-33 FLG is initially translated to profilaggrin, a large highly-phosphorylated
polypeptide composed of an N-terminal domain, 10-12 filaggrin monomer repeats and a C-terminal
domain.34-35 Profilaggrin localizes to the keratohyalin granules in the stratum granulosum
keratinocytes.32,34 During keratinocyte differentiation, secreted profilaggrin is dephosphorylated and
broken down by extracellular serine proteases. This process is highly regulated by pH and the
presence of protease inhibitors.35 The N-terminal domain likely contributes to keratinocytes
differentiation; it contains an S100-like calcium binding domain, which may be involved in calcium
dependent steps of terminal differentiation or profilaggrin processing, and a nuclear localization
domain, which may be involved in keratinocytes enucleation.34 The function of the C-terminal
domain is still unknown, however it seems to be necessary for profilaggrin processing to filaggrin.34-36
The filaggrin monomers are essential to the epidermal barrier. They bind to constituents of the
keratin cytoskeleton and other filament proteins to form tight bundles during terminal
differentiation of keratinocytes into corneocytes. Filaggrin is also an important part of the cornified
cell envelope which acts as a barrier to water loss by cross-linking the corneocytes. Excess filaggrin in
the skin is degraded into its constituent amino acids. The most abundant breakdown products,
histidine, argenine and glutamine, make up the majority of the natural moisturizing factor (NMF)
which helps maintain skin hydration and may help regulate the pH balance.34-35 Histidine is further
metabolized to urocanic acid (UCA) and glutamine is further metabolized to pyrrolidone-5-carboxylic
acid (PCA), both of which are also important parts of the NMF and help maintain the pH gradient in
the epidermis.34,37 There is some evidence that filaggrin is also involved in protection against UVmediated damage.34 Despite the importance of filaggrin, it has a relatively short half-life of six
hours.35
Filaggrin polymorphisms
Homozygous or compound heterozygous mutations in the FLG gene have been causally
linked to ichthyosis vulgaris (IV).34,38 Since IV has similar symptoms and often co-presents with AD,
the R501X (a non-sense mutation) and 2282del4 (a 4 base pair deletion causing frameshift) null
mutations were also investigated for a role in AD.32 Using the initial IV family-based study patients,
Palmer et al (2006)32 found an association between the two FLG mutations and AD. They repeated
these results with a cohort study of Irish pediatric AD patients, a case-control cohort study of
Scottish school children and a longitudinal study of Danish children with AD.32 These results were
subsequently confirmed in a German family-based study by Weidinger et al (2006)39 and have since
been confirmed in over 20 case-control studies and 8 family based studies.33-34,36,40-41
To date, over 40 null mutations in the FLG gene have been identified in European and Asian
populations (see Figure 2). The expression patterns of the mutations vary/segregate across ethnic
groups.36 Approximately 9-10% of the European population carry one of the two most common FLG
null-mutations, whereas 25-50% of AD patients have FLG mutations.32,36,42 As such, FLG mutations
account for approximately 15% of the population attributable risk of AD development.43 R501X and
2282del4 null-mutations are semi-dominant, with homozygous or compound heterozygous
mutations correlating to the highest risk of developing IV, AD or other skin diseases.32,44
FLG null mutations in AD lead to a number of barrier defects including impaired corneocyte
differentiation due to impaired filament/keratin aggregation. Cell-cell adhesion is also affected by
FLG mutations which result in decreased corneodesmosin, an important component of
corneodesmosomes and decreased numbers of tight junctions. FLG deficiencies also lead to the
development of a defective lipid lamellae, and lack of NMF leading to water loss and increased
epidermal pH.37,42 Increased SC pH can lead to a blockade of lamellar body secretion, increased
- 11 -
desquamation due to activation of proteases and deactivation of protease inhibitors, and increased
bacterial colonization.17,22,42 Together, these barrier function deficiencies likely allow for increased
allergen sensitization and contribute to the inflammatory response. This is supported by the
association of FLG mutations with increased IgE levels.36,39 FLG mutations have also been weakly
associated with AD severity, and AD patients with FLG mutations tend to have increased skin
infections with herpes virus, and greater risk of other allergies and asthma than those without
mutations.41-42
A
B
Normal skin barrier
Filaggrin staining
in normal skin
Ichthyosis vulgaris and
atopic dermatitis
Filaggrin
granules
Defective skin barrier
No filaggrin
granules
Figure 2 FLG in atopic dermatitis (A) Schematic representation of the FLG gene and mutations associated with AD in
European and Asian populations. Mutations in red are recurrent in the population and mutations in black are rare or
family-specific. (B) Filaggrin immunostaining in healthy skin shows filaggrin concentration in the upper SC and in lamellar
granules (left panel). Filaggrin immunostaining of patient skin with a homozygous FLG loss-of-function mutation and
suffering from IV and AD shows a complete lack of filaggrin in the SC and the lamellar granules (right panel). Adapted from
Irvine et al (2011)42 and Irvine and Mclean (2006)45
Other factors influencing filaggrin expression levels
Although FLG mutations are the leading predictive factor for the development of AD, only
40-60% of FLG mutation carriers develop AD.35,42 Factors other than genetic polymorphisms also
affect filaggrin expression levels. The number of filaggrin monomer repeats in the FLG gene naturally
varies from 10 to 12. An increased number of filaggrin repeats has been associated with a decreased
risk of dry skin and AD.32,34,46
Filaggrin expression can also be influenced by the inflammatory environment. IL-4, IL-13, IL22, and IL-25 have been shown to decrease FLG expression.34,42 IL-22 may also down-regulate the
profilaggrin processing enzyme cathepsin D and other Th2 cytokines can down-regulate caspase 14,
which deiminates the filaggrin monomers before they can be degraded to individual amino acids and
- 12 -
metabolized to the NMF components.34,47 Disease severity has also been found to affect NMF levels,
indicating that disease severity and the inflammatory environment play a role in FLG expression.37
In addition, FLG has been shown to interact with an eczema risk variant on the 11q13.5
chromosomal region and with polymorphisms on the IL-10 and IL-13 genes.34 These interactions
highlight the complex role of FLG in the skin barrier and that a combination of mutations in
interacting genes, in combination with environmental factors, may be responsible for the resulting
phenotype.
Kallikrein-related peptidases
Kallikrein-related peptidases (KLKs) are a family of 15 serine proteases encoded by genes on
chromosome 19q13.4.22,48-50 KLKs are synthesized as inactive pre-pro-enzymes with a 15-30 aminoacid pre-sequence on the N-terminal end that is cleaved off before secretion into the SC.17,49-50 At
neutral pH, as in the lower SC, pro-KLK5 zymogens can self activate through trypsin digestion to
remove the pro-peptide and initiate an activation cascade of other KLKs.17,50 KLK5, KLK7 and KLK14
are most abundant and the only KLKs that are found in their active form in the SC.49-50 KLKs primarily
function in desquamation (see Figure 1).17,22,49,51 They initiate corneodesmosome degradation
through the cleavage of key constituent proteins desmoglein-1 (DSG-1) by KLK5 and KLK14, and
corneodesmosin (Cdsn) and desmocollin-1 by KLK7.17,22,48 KLKs also affect the skin barrier through
degradation of lipid processing enzymes that work at the SG-SC interface to create the lipid lamellae
from the lamellar granule secretions.22,51-52 KLK5 and KLK14 can signal through PAR2 to downregulate lamellar granule secretion,22,53 and increase inflammatory cytokine secretion, particularly
TSLP.17,26,48-49,51,53 In addition, KLKs are involved in profilaggrin processing both directly and through
activation of ELA-2, another important profilaggrin processing enzyme.48,51
Kallikrein-related peptidase-7 polymorphisms
AD patients exhibit abnormally high levels of KLK7 and increased activity of KLK5, KLK7 and
KLK14 in lesional skin.50 In an initial study, Vasilopoulos et al (2004)54 screened for KLK7
polymorphisms and their role in AD. They found a 4 base-pair insertion (AACC) in the 3’ UTR which,
they hypothesize, increases KLK7 mRNA stability thereby increasing KLK7 activity. In their study, the
two-insertion repeat allele (AACCAACC) was significantly associated with AD.54 However, these
results have not been replicated. In a large study Weidinger et al (2008)55 found no association
between KLK7 3’UTR insertions and AD. They conclude that although the 3’ UTR insertion variant of
KLK7 does not increase the risk of AD development, the KLKs are important skin barrier regulatory
enzymes and other undiscovered polymorphisms may play a role.55
Lymphoepithelial Kazal-type-related inhibitor
Lymphoepithelial Kazal-type-related inhibitor (LEKTI) is a serine-protease inhibitor encoded
by serine protease inhibitor, Kazal-type, 5 (SPINK5) on chromosome 5q31-33 at the distal end of the
IL-4 cytokine gene cluster.14,52,56-58 LEKTI is composed of an initial peptide sequence and 15 inhibitory
domains.49,58 Variable transcriptional processing results in three LEKTI variants differing at the Cterminal end. LEKTI-FL is the most abundant and contains all 15 domains, LEKTI-Sh is shorter,
containing only the first 13 domains and LEKTI-L is elongated by a 30 amino-acid insertion between
domains 13 (D13) and D14.49,51 D2 and D15 contain true Kazal motifs (6 cysteine residues) whereas
the other domains contain Kazal-like motifs (4 cysteine residues). These cysteine residues, which
form two or three disulfide bonds forcing a loop structure within the domain to mimic serine
protease substrates, are responsible for the inhibitory action.49 In the skin LEKTI is packaged in
lamellar granules, where it is processed into polypeptides by furin cleavage of the argenine or lysine
rich linker sequences and secreted at the SG-SC interface.48 Each of the three LEKTI variants undergo
- 13 -
similar processing to produce D1, D5, D6, D7, D6-D9, D7-D9, D8-D9, D10-15, and D10-D13
polypeptides.49-50,58
Although LEKTI can control filaggrin expression through inhibition of profilaggrin proteolytic
enzymes,51 it primarily functions as a KLK serine protease inhibitor regulating desquamation in the
SC.49-50 LEKTI secretion from lamellar granules occurs before KLK secretion to prevent protease
activity in the lower SC.48-49,59 The secreted domain fragments exhibit variable patterns of inhibition
on the different KLKs. All domains except D1 inhibit KLK5 with varying strength; D5, D6, D8-11 and
D9-15 inhibit KLK5, KLK7 and KLK14. D6-9 and D9-12 inhibit KLK5 and KLK14 and D10-D15 fragments
primarily inhibit KLK7.49-50 LEKTI function is regulated by the pH gradient in the SC. At the SG-SC
interface the pH is near neutral allowing optimal LEKTI function and inhibition of KLK activity. The
progressive pH decrease in the outer SC layers inhibits LEKTI function to allow KLK activity and
desquamation in the outermost SC.17,21,50
Serine protease inhibitor, Kazal-type, 5 polymorphisms
Autosomal recessive SPINK5 loss-of-function mutations result in the rare skin disease
Netherton syndrome.22,57 Netherton patients completely lack LEKTI resulting in unrestricted
desquamation and severe barrier defects.52 Since Netherton syndrome patients always also present
with AD-like symptoms, SPINK5 mutations were investigated for an association to AD.53
In two independent panels of British families Walley et al (2001)58 found a maternally
derived association of the E420K polymorphism to AD, atopy, and elevated IgE. These results were
further confirmed by Kato et al (2003)60 who found an association between seven SPINK5
polymorphisms, including E420K, and AD. Nishio et al (2003)61 also found an association of the E420K
polymorphism to AD. In a large study of German children, the E420K polymorphism was associated
with AD when presenting with asthma but not to AD alone.62 A second study of a German population
and a French study also failed to find an association between AD and the E420K polymorphism.52
Since then, however, there have been more studies linking the polymorphism to AD: Weidinger et al
(2008)55 found an association of maternally inherited E420K polymorphisms to AD and a Chinese
study by Zhao et al (2012)52 found an association of E420K polymorphisms to AD.52,55
Although the E420K polymorphism has been repeatedly associated with AD, until recently its
function was entirely unknown.52,63 It lies near the furin cleavage site within the D6-D7 linker region
causing preferential cleavage. This altered fragmentation pattern results in a complete lack of the
D6-D9 fragment, the most potent inhibitor of KLK5 activity.51 Fortugno et al (2012)51 found that a
420KK genotype increases KLK5 and KLK7 activity resulting in increased DSG1 degradation and
decreased barrier function. They also found a correlation between increased TSLP levels and the
420KK variant, indicating that an imbalance between serine proteases and their inhibitors leads to
increased PAR-2 activation and TSLP secretion driving a Th2 response.51 These results show for the
first time how the 420K LEKTI polymorphism may be involved in the drive towards AD.
LEKTI 420KK variants have also been associated with increased ELA-2 activity, weaker
inhibition of profilaggrin proteolytic enzymes and decreased profilaggrin and filaggrin monomer
expression.51 Increased SC pH due to decreased filaggrin levels leads to KLK activation, decreased
skin barrier and initiation of the inflammatory cascade through PAR-2.22 Due to the functional links
between KLKs, LEKTI and filaggrin, and the potential association of each of these to AD, Weidinger et
al (2008)55 investigated the interaction between KLK7, SPINK5 and FLG polymorphisms. However,
they found no effect of KLK7 polymorphisms on AD risk and no interaction of KLK7 or SPINK5
polymorphisms with FLG polymorphisms.55
Cystatin A
CSTA, located on the AD susceptibility locus of chromosome 3q21, encodes the cysteine
protease inhibitor cystatin A.17 Cystatin A is expressed in the SC and in sweat. In the SC it functions
- 14 -
as a cathepsin inhibitor. It inhibits the cysteine proteases cathepsins B, H and L which are involved in
corneodesmosomal degradation during desquamation.64 In sweat, cystatin A forms a protective
barrier against exogenous proteases. It is a potent inhibitor of common house dust mite allergens,
Der p 1 and Der f 1, and S. aureus derived proteases.64-65 It is estimated that 50% of IgE antibodies in
allergic diseases are against Der p 1 and Der f 1.65 These allergens are not only able to breakdown
the corneodesmosomal barrier, but they can also activate keratinocytes to produce IL-8 and GM-CSF
potentially leading to enhanced allergen sensitization.65-66 Cystatin A is an important inhibitor of
endogenous and exogenous cysteine proteases to prevent epidermal barrier breakdown and
allergen sensitization.
Cystatin A polymorphisms
Seguchi et al (1996)67 reported decreased cystatin A levels in lesional skin of AD patients, but
no reduction in non-lesional AD skin. Recently, Vasilopoulos et al (2007)64 analyzed three CSTA
single nucleotide polymorphisms (SNPs): T-190C, T+162C and C+344T. They found a significant
association between the +344C variant and AD. Interestingly the rare +344T allele seemed to be
protective against AD development. They performed functional analysis on the +344C variant and
found that +344C mRNA has a much higher degradation rate, resulting in lower cystatin A levels.64
Together these data indicate that CSTA polymorphisms may predispose to AD and that the AD
phenotype may regulate cystatin A levels. Although there have not been many studies on cystatin A,
it appears to be an important barrier protein involved in regulating desquamation and preventing
exogenous protease damage and allergen sensitization.
Claudin-1
Tight junctions are important connective structures between cells. They act as a barrier
against unwanted material and control trans-epithelial water loss (TEWL) and trans-epithelial electric
resistance (TEER).68-69 Tight junctions are composed of two types of proteins: trans-membrane
proteins which anchor neighboring cells and scaffold proteins which anchor the trans-membrane
proteins to the intracellular matrix.68 Occludins and claudins are the most abundant and conserved
tight junction trans-membrane proteins (see Figure 3 for TJ structure).30 Claudin-1 is encoded by
CLDN1 on chromosome 3q28-q29.68 It has four trans-membrane domains and two extracellular loops
that interact with the neighbouring cell.69 Claudin-1 is known to increase barrier function in tight
junctions, but beyond this little is known of its expression patterns and function in humans.68 Tight
junctions were thought to be primarily involved in simple epithelial barriers; however, Furuse et al
(2002)70 demonstrated the functional importance of tight junctions and claudin-1 in stratified
epithelia through the generation of CLDN1-/- mice. These mice had wrinkled skin and a severely
damaged epidermal barrier and died within one day after birth.70
Claudin-1 polymorphisms
De Benadetto et al (2011)68,71 have found an approximate 50% decrease of claudin-1 mRNA
and protein levels in non-lesional AD skin. They also found functional defects in claudin-1 deficient
lesional and non-lesional AD skin. Interestingly, in their system IL-4 and IL-13 caused claudin-1 upregulation. Together these data point to an intrinsic claudin-1 deficiency in AD, rather than a
disease-driven deficiency. Analysis of 24 CLDN1 SNPs common to both an African-American and
European-American population revealed several significant associations in the African-American
population. SNP rs9290927 was associated with increased risk of AD. Two SNPs (rs893051 in intron 1
and rs9290929 in the promoter) were associated with increased disease severity and, interestingly,
SNP rs17501010 was associated with decreased AD risk. They were only able to find modest
associations in the European-American population.68
- 15 -
In a second study, De Bernadetto et al (2011)71 investigated the potential association of
CLDN1 polymorphisms and herpes simplex virus-1 (HSV-1) infections. HSV-1 infections in patients
with AD can lead to eczema herpaticum (EH), a rare and severe widespread skin infection. Previous
studies have shown that susceptibility of keratinocytes to HSV-1 infection is inversely correlated to
the amount of cell-cell contact. They found that claudin-1 silencing by siRNA significantly increased
HSV-1 infection levels. They subsequently reanalyzed the African-American and European-American
populations from their previous study for CLDN1 SNPs associated to EH. Initially, they were only able
to find modest associations; however, when they excluded patients with FLG mutations, they found
a significant association of two CLDN1 SNPs (rs3774032 and rs3732923) to EH in the EuropeanAmerican population. After the exclusion of FLG polymorphisms the African-American population
was too small to analyze.71
Although both of these studies are quite small and require replication, they indicate the
importance of tight junction integrity in resistance to development of AD and prevention of viral
exacerbations.
Adhesion genes associated to AD
CLDN1
Adhesion genes associated to asthma
CLDN1
OCLN
ZO1
PCHD1
CDH1
CTNNA1?
CTNNA
Figure 3 Tight junction, adherens junction and desmosome structure and genes associated with adhesion
abnormalities and atopic diseases. Adapted from Nawijn et al (2011)72
- 16 -
Role of the Lung Epithelium in Allergy
Airway epithelium structure
The airway epithelium is the largest surface that is in continual contact with the
environment and serves as the first line of defense against inhaled particles.19,73 The airway is
equipped with a mucous barrier, a physical barrier and a catabolic barrier to prevent entry of
harmful substances. The epithelial cell layer is composed of ciliated columnar cells, mucous secreting
goblet cells, and surfactant secreting Clara cells. The mucous traps inhaled particles, allergens and
microbes and the ciliated cells act as a conveyor belt to sweep away unwanted materials. The
physical barrier is formed by tight junctions joining the apical ends of the epithelial cells, to tightly
bind the cells and prevent soluble factors from passing between them.13,19,73-74 Other cell-cell and
cell-extracellular matrix interactions, such as adherens junctions, desmosomes and hemidesmosomes, further strengthen the physical barrier of the airway epithelium (see Figure 3).19,73-74
The basement membrane lies directly under the epithelial cell layer and acts as a barrier between
the epithelium and the underlying mesenchyme.75 It is composed of collagen, laminin, fibronectin,
proteoglycans and other extracellular matrix (ECM) proteins.75-77 Fibroblasts in the lamina propria
and airway smooth muscle cells produce the extracellular matrix proteins found in the basement
membrane.76 See Figure 4 for airway barrier structure. Finally, the epithelial cells form a catabolic
barrier through the expression of surface peptidases to degrade damage-inducing peptides, and the
secretion of protease inhibitors to limit the effects of allergen or pathogen derived proteases.13 To
protect against reactive oxygen species (ROS)-induced tissue damage from biological or chemical
insults, the epithelia contains many anti-oxidant enzymes including superoxide dismutase and
glutathione peroxidase.19,73 Epithelial cells also produce antimicrobial peptides such as defensins,
cathelicidins, and collectins which are toxic to invading pathogens and recruit immune cells.78
Epithelial cells
Epithelial cells (ECs) play an important role in the initiation of airway allergic inflammation.
They sample the environment through various pattern recognition receptors (PRRs), including TLRs,
NLRs, C-type lectins and protease activated receptors (PARs).79 Many allergens interact with TLRs
and PARs activating the ECs to produce cytokines, such as IL-25, IL-33 and TSLP, and chemokines,
such as CCL17 and CCL22, that drive the allergic Th2 inflammatory response (see Table 3 for a more
extensive list).2,13,19,79-80 IL-25, IL-33 and TSLP potently promote a Th2 immune response by inducing
Th2 cell, mast cell and basophil production of IL-4 and/or IL-13. They also promote eosinophil survival
and inflammatory cytokine production. TSLP also primes DCs to initiate a Th2 response.78 Inhaled TLR
ligands can act as adjuvants further activating the ECs to produce Th2 inducing cytokines and
chemokines.2
Epithelial cells are also involved in the perpetuation of airway allergic inflammation. IL-4 and
IL-13 stimulated ECs increase production of GM-CSF, CCL11 and CCL17 to increase recruitment of
eosinophils and Th2 cells to the airways. In addition, IL-4 and IL-13 disrupt the epithelial barrier
which activates the EC immune response and maintains allergic inflammation.79 IL-25, IL-33 and TSLP
may mediate secretion of IL-13 and TGF-β resulting in increased fibroblast production of
extracellular matrix proteins. This contributes to basement membrane thickening, one of the key
features of airway remodelling.78 EC derived GM-CSF may also play a role in airway remodelling.73
- 17 -
Table 3 Epithelial cell derived factors contributing to the allergic immune response
Soluble factor
Production stimulated by
Function in allergic disease
Cytokines
IL-6
Pro-inflammatory cytokine
Stimulates Th2 response, IgA mucosal production and B cell
stimulation, fungal or dust mite
differentiation13
proteases, or respiratory
viruses13
IL-11
Pro-inflammatory cytokine
Involved in B cell activation (dependent on T cells). Increased
stimulation or respiratory
levels observed in severe asthma patients13
13
viruses
IL-16
T cell and eosinophil attractant13
2
IL-25
TLR stimulation
enhances Th2 memory response, interacts with mast cells,
basophils, eosinophils and endothelial cells to enhance the
inflammatory response and important in DC activation2,79
2
IL-33
TLR stimulation
induces Th2 proliferation and cytokine production interacts
with mast cells, basophils, eosinophils and endothelial cells to
enhance the inflammatory response and activates DCs to
induce Th2 response2,79
GM-CSF
Pro-inflammatory cytokines, TLR activates neutrophils, eosinophils and macrophages.
stimulation2
eosinophil survival factor13 Important in DC activation and
promotes Th2 response2,79
TSLP
TLR stimulation2
primes DCs to induce Th2 response13,79 interacts with mast
cells, basophils, eosinophils and endothelial cells to enhance
the inflammatory response.2 important in DC activation.
stimulates bronchial EC proliferation and IL-13 production.
Important in basophil growth and differentiation. May also be
involved in epithelial repair mechanisms 79
Chemokines
CXC chemokines
mast cell recruitment13
CCL11, CCL24, CCL26
(eotaxins 1-3)
CCL5 (RANTES)
CCL17
CCL22
CCL20
CCL28
IL-4 and IL-1313
IL-4 and allergens, TLR
stimulation2 and epithelial
damage73
TLR stimulation2 and epithelial
damage73
inflammatory cytokines and
ambient particles13
inflammatory cytokines13
Bind to CCR3 on eosinophils and basophils13
eosinophil attractant13
Th2 recruitment13,19
Th2 recuitment19
recruitment of immature DCs13
recruitment of eosinophils and T cells13
- 18 -
Asthma
Asthma is a chronic inflammatory disease of the conducting airways that affects 5-10% of
the developed world.2,16 Allergic asthma typically begins in early childhood with allergen-triggered
episodes of airway hyper-responsiveness and inflammation. As it progresses to a chronic state it
spreads to the distal airways and is associated with airway remodelling.19,81 Asthma is characterized
by symptom free periods and periods of exacerbation caused by viral infections or allergen
exposure.82 During periods of exacerbations patients experience coughing, wheezing, chest tightness
and difficulty breathing.79 Asthma is caused by the interaction of genetic and environmental factors.
It has been estimated that 36-79% of asthma is heritable. Over 120 genes have been associated with
asthma but without a clear inheritance pattern. Chromosomal regions 5q23-31, 5p15 and 12q1424.2, which contain immune related genes such as IL3, IL4, IL5, IL9, IL13, IFNγ and FcєRIβ are
consistently associated with asthma.83 Early life viral infections also seem to play a driving role in
asthma development. Persistent viral infections in the lower respiratory tract, particularly with
rhinovirus (HRV) or respiratory syncytial virus (RSV), are associated with an increased risk of asthma
development.19,84 In contrast, early life microbial exposure may play a protective role.2 Recent
studies also indicate that early life changes in lung or gut microbiome compositions can increase risk
of asthma development.85-86
The asthmatic lung is characterized by infiltration of mast cells, eosinophils and Th2 cells in
the epithelium and the lamina propria.3,19,79,81 Allergen recognition by IgE sensitized mast cells and
basophils triggers degranulation releasing histamine, leukotrines, inflammatory cytokines and
chemokines. This triggers acute bronchial smooth muscle contractions and recruits inflammatory
immune cells.19,87 The infiltrating eosinophils and Th2 cells perpetuate the bronchial contractions and
inflamed conditions through the secretion of lipid, peptide and protein mediators and Th2 cytokines
such as IL-4, IL-5, and IL-13.13,81,87 Although most asthma is primarily Th2 mediated, in some patients
the primary infiltrates are neutrophils. High INF-γ responses are also observed during acute asthma
attacks in some patients.16,79,87 These, and similar observations, have led to the view of asthma as a
range of diseases with distinct phenotypes.16
Bronchial hyperresponsiveness (BHR) and airway remodelling are key features of asthma.
BHR is characterized by airway obstruction in response to a non-allergic physical or chemical
stimulus. This may be due to underlying airway Th2 or eosinophilic inflammation.79,88 Airway
remodelling is characterized by thickened basal lamina, increased smooth muscle around the airway
walls, goblet cell hyperplasia and metaplasia and angiogenesis (see Figure 4).73,89 Increased fibroblast
proliferation and differentiation into myofibroblasts results in increased deposition of ECM proteins
including collagen, fibronectin, laminin and proteoglycan.27,77 Airway smooth muscle (ASM)
undergoes hyperplasia and hypertrophy contributing to airway constriction and breathing
difficulties.76,79 ASM is also known to produce ECM proteins contributing to basal laminar
thickening.76 Increased production of highly viscous mucous due to goblet cell hyperplasia and
metaplasia also contributes to breathing difficulties.19,79 Angiogenesis allows for increased infiltration
of immune effector cells and perpetuation of the allergic inflammatory response.90 The cause of
airway remodelling is unknown, however both Th2 cytokines and genetic factors have been
implicated.76,89-90 An emerging theory is that an aberrant epithelial repair response leads to continual
activation of the epithelial mesenchymal trophic unit (EMTU) resulting in airway remodelling.89 The
EMTU is defined as the bidirectional communication between the epithelium and the underlying
mesenchyme resulting in cytokine and growth factor production.73
In recent years the suspected origins of asthma have been re-evaluated. Atopy-driven
chronic inflammation was thought to be the driving force behind airway remodelling. However,
airway remodelling is also found in young children and in the relative absence of inflammation.13,73 In
light of this, the contribution of the epithelium to both the development and perpetuation of asthma
is a major research focus.2,19 The epithelium acts as the primary barrier against allergen entry.
Intrinsic barrier dysfunction can increase the risk of allergen sensitization and asthma
development.13,19 Recently tissue restricted genes have been associated with increased asthma risk.
- 19 -
As discussed below, many of these genes are involved in the maintenance of the epithelial barrier
and some may play a role in airway remodelling (see Figure 4).
Healthy airway wall
TJ and AJ barrier
CLDN1? OCLN
ZO1
CDH1
CTNNA1?
CTNNA
Asthmatic airway wall
Legend
Barrier breakdown
Th2 inflammation
Airway remodelling
Lymphocyte
Macrophage
Eosinophil
Neutrophil
Epithelial cell
Other genes associated
with asthma
PCHD1 FLG? SPINK5?
Goblet cell
Smooth muscle cell
(myo)fibroblast
Blood vessel
Airway remodelling
ADAM33 CLDN1
Extracellular matrix
Basement membrane
Tight junction
Figure 4 Schematic representation of the airway epithelial structure in healthy tissue and asthmatic tissue. Genetic
polymorphisms have been associated with the epithelial barrier breakdown and airway remodelling in asthma. Adapted
from Rydell-Törmänen et al (2012)76
ADAM protein family
The ADAM (a disintegrin and metalloprotease) family is a sub-group of the zinc-dependent
metalloprotease superfamily.91 ADAMs are large membrane bound proteases with eight domains: an
N-terminus signal sequence, a pro-domain, a metalloprotease domain, a disintegrin-like domain, a
cysteine rich domain, an EGF-like domain, a trans-membrane domain and a cytoplasmic domain.91-93
They are unique because of the presence of both a disintegrin domain and a metalloprotease
domain. The disintegrin-like domain contains a 14 amino acid RGD motif in the disintegrin loop that
is necessary for integrin binding to facilitate cell-cell or cell-matrix interactions.92,94 The
metalloprotease domain contains a histidine-rich zinc-binding consensus sequence. Loss of the
histidine residues in this consensus sequence results in an inability to bind zinc and a loss of
proteolytic function.94-95 The metalloprotease domain is thought to be involved in extracellular
matrix breakdown and in the shedding of membrane bound cytokines, growth factors and
receptors.92 ADAMs are expressed in a variety of tissues and are thought to play a role in multiple
processes including cell adhesion, proliferation, differentiation, signalling, apoptosis and the
inflammatory response. 83,92,94 There are six ADAM subfamilies each with distinct characteristics.94
- 20 -
ADAM33 polymorphisms
ADAM33 belongs to subfamily E which also includes ADAM8, 12, 15, and 19.94-95 All the
members of this subfamily have active metalloprotease domain.91 ADAM33 is located on
chromosome 20p13 and exists in multiple splice variants, differing in the number of domains
expressed.93-94,96 The function of ADAM33 is largely unknown. Van Eerdewegh et al (2002)91 were the
first to associate ADAM33 with asthma. Genome wide scans of 460 Caucasian families identified an
association between the 20p13 gene region and asthma. Analysis of 135 SNPs in 23 genes in this
region identified a significant association of ADAM33 to asthma and bronchial hyperresponsiveness.
Van Eerdewegh et al (2002)91 found six SNPs that were significantly associated with asthma in a US
population and seven SNPs that were significantly associated with asthma in a UK population, some
of which were in linkage disequilibrium with SNPs in adjacent genes.91 Howard et al (2003)97
confirmed the association of ADAM33 to asthma in four populations: African-American, US white, US
Hispanic and Dutch white. They typed eight SNPs in the 3’ region and found a significant association
of at least one SNP in each population.97 Lind et al (2003)98 studied six individual SNPs and
combinations thereof in US Puerto Rican and Mexican populations; however they found no
association of any individual SNP or combination of SNPs with asthma risk in either population.98
Raby et al (2004)99 tested 17 SNPs (9 of which were found to be significant by Van Eerdewegh et al
(2002)91) in North American children. They were not able to find an association of any SNP to asthma
and only found a slight association of one haplotype to asthma.99 Since then the association of
ADAM33 with asthma has not gotten clearer. Positive associations have been found in German,
Chinese, Thai and Indian populations, however other studies failed to find associations in Chinese,
Australian, Colombian, and Indian populations.83,100-101 The lack of consistent SNP associations across
multiple populations may indicate that the major causative SNP in ADAM33 has not yet been found,
or that the true association of chromosome 20p13 is caused by a different gene, which may be in
linkage disequillbrium with ADAM33.97 Interestingly, a study by Holgate et al (2006)102 found an
association of three ADAM33 SNPs with early life lung function and asthma severity in later life
indicating that ADAM33 may not directly affect asthma risk but rather may be associated to
decreased lung function which predisposes to asthma development.102
In the lungs ADAM33 is primarily found in smooth muscle cells, myofibroblasts and
fibroblasts.83,91,95 There are some reports of ADAM33 expression in lung epithelial cells, however as
ADAM33 has been found to be silenced by DNA methylation in epithelial cells of both asthma
patients and healthy controls it is considered functionally absent.96,103-104 Due to its localization,
ADAM33 is also postulated to be involved in airway remodelling.83,91,95 As ADAM33 has a functional
metalloprotease domain it likely functions as a sheddase, releasing cytokines, growth factors and
receptors from the cell surface. The release of these factors may increase proliferation and
differentiation of fibroblasts leading to increased deposition of ECM components and may influence
smooth muscle growth leading to the smooth muscle thickening seen in asthma patients.83,95,102 Lee
et al (2006)105 identified a soluble variant of ADAM33 (sADAM33) in the bronchoalveolar lavage fluid
of asthma patients which correlated with decreased lung function. Puxeddu et al (2008)106
discovered that sADAM33 is produced from ectodomain shedding of membrane bound ADAM33. As
such, it contains the metalloprotease domain but has lost the cytoplasmic domain. This allows
sADAM33 to access proteolytic targets the membrane bound form was excluded from, potentially
increasing its function. They were able to confirm that sADAM33 promotes angiogensis, likely
through increased shedding of angiogenic factors in the ECM. Many of the ADAM33 SNPs associated
with asthma are in the transmembrane or cytoplasmic domains. These SNPs may play a role in
sADAM33 shedding or localization.106 This study shows, for the first time, a function of ADAM33 and
proposes a potential effect of ADAM33 polymorphisms.
- 21 -
Tight and Adherens Junctions
The airway epithelial barrier is primarily maintained through tight junctions (TJs), adherens
junctions (AJs) and desmosomes which tightly bind neighbouring cells but still allow water and solute
passage between them. Tight junctions are primarily composed of trans-membrane proteins,
including claudins, occludins and junctional adhesion molecules (JAM), and scaffold proteins, the
zonula occludens (ZO-1, -2, -3) which link the trans-membrane proteins to the actin
cytoskeleton.72,107 Adherens junctions are primarily composed of e-cadherin which is bound to the
keratin and actin cytoskeleton by p120 catenin, α-catenin, β-catenin and γ-catenin.72,108
Desmosomes are primarily composed of desmoleins and desmocollins.77 See Figure 3 for a schematic
representation of these adhesion complexes. Recently asthma has been recognized as a disease of
compromised epithelial barrier. Tight junctions are intrinsically deficient in asthmatic tissue and
many of these junctional proteins have been associated with a risk of developing asthma (see Figure
3 and Figure 5).73
Figure 5 Tight junction distribution is abnormal in asthmatic epithelium. TJ component ZO-1 is stained green. Left panel
represents healthy bronchial tissue. Right panel represents asthmatic bronchial tissue. Adapted from Holgate (2007)73
Claudin-1
Claudin-1 is a trans-membrane protein which, in epithelial cell tight junctions, regulates
plasma membrane permeability.72 Interestingly, claudin-1 is also expressed in the nucleus and
cytoplasm of ASM cells where it seems to play a role in cell proliferation. Fuijta et al (2011)69 found
that over-expression of claudin-1 in ASM cells results in increased cell proliferation and production
of VEGF, an important angiogenic factor. IL-1β and TNF-α seemed to upregulate claudin-1 expression
in ASM whereas Th2 cytokines IL-4 and IL-13 down-regulated expression. They found increased
claudin-1 expression in asthma patients ASM cells. These results indicate that claudin-1 may also
play an important role in airway remodelling.69 Future testing is needed to determine if genetic
polymorphisms are responsible for the increased expression of claudin-1 in asthmatic ASM. It would
also be interesting to examine if CLDN1 is increased in the TJs of these patients and the effects of
potential CLDN1 polymorphisms on TJ integrity.
Occludin
Occludin is a trans-membrane TJ protein that is involved in de novo TJ assembly.72 Xiao et al
(2011)107 found decreased protein levels of occludin in asthma patients. The mRNA levels, however,
were normal suggesting epigenetic down-regulation. They were able to determine that occludin
down-regulation was not caused by the Th2 cytokine milieu found in asthma.107 Ahdieh et al (2001)109
however, found that IL-4 and IL-13 induced decreased expression of occludin resulting in decreased
epithelial barrier function. These studies indicate the potential role of both genetic (or epigenetic)
polymorphisms and local environment in epithelial barrier disruption.
- 22 -
Zonula occludens-1
Zonula occludens-1 (ZO-1) acts primarily as a scaffold protein in TJs to secure claudins,
occludins and JAMs to the intracellular actin cytoskeleton. It can, however, also act as a transcription
factor when bound to ZO-1-associated nucleic acid binding protein (ZONAB).108 de Boer et al
(2008)108 were the first to report significantly reduced levels of ZO-1 in asthmatic epithelia. This was
confirmed by Xiao et al (2011)107 who also reported significant reductions in ZO-1 protein levels in
asthmatic bronchial epithelium. Although they found no reductions in ZO-1 mRNA levels, indicating
epigenetic or environmental control, cultures of asthmatic bronchial cells in the absence of Th2
cytokines did not revert back to wild type ZO-1 expression indicating intrinsic deficiencies.107 Ahdieh
et al (2001)109 found that Th2 cytokines, IL-4 and IL-13, are also able to decrease expression of ZO-1.
Intrinsic defects in ZO-1 and disease induced down-regulation may contribute to the disrupted
epithelial barrier in asthma (see Figure 5).
E-cadherin
E-cadherin is the adhesion protein that has most consistently been associated with asthma.
E-cadherin, encoded by CDH1 on chromosome 16q22.1, is a trans-membrane glycoprotein with
extracellular domains that form Ca+2 dependent interactions with cadherins on neighboring cells.
These interactions maintain a mechanical connection to surrounding cells, playing an important role
in the AJ epithelial barrier. The intracellular tail is highly conserved and interacts with scaffold
proteins to anchor e-cadherin to the cytoskeleton. In addition to its role in cell adhesion, e-cadherin
can regulate cell proliferation and differentiation through interactions with ZONAB, and can
negatively regulate multiple signalling cascades.72,110-111 As a CD103 ligand, E-cadherin may play a
role in Treg retention in the lungs and may limit DC activation during bronchial lumen sampling.72
de Boer et al (2008)108 and Hackett et al (2011)112 reported significantly decreased levels of
e-cadherin in asthma patient epithelia. Ierodiakonou et al (2011)111 repeated these results and
investigated the role of CDH1 polymorphisms. They found five SNPs (rs8056633, rs16958383,
rs2276330, rs3785078 and rs7203904) that were associated with significantly decreased e-cadherin
expression levels in asthma patients without inhaled corticosteroid (ICS) use. Recent evidence
shows potential of ICS to repair the epithelial barrier and increase e-cadherin expression, so
Ierodiakonou et al (2011)111 also looked at CHD1 polymorphisms in asthma patients with ICS use. In
these patients they found 7 SNPs associated with airway remodelling, 3 with CD8+ T cell counts, 2
with eosinophil counts and 7 with lung function decline. These results indicate that ICS use may alter
the functional effects of CHD1 polymorphisms.111
E-cadherin expression levels are also influenced by environmental factors such as RSV
infection and pro-inflammatory cytokines.77,112 Trautmann et al (2005)77 found that IFN-γ and TNF-α
cause e-cadherin down-regulation. T cell produced IFN-γ sensitizes epithelial cells to eosinophil TNFα induced apoptosis, which may result in epithelial cell shedding, a characteristic feature of asthma.
They also found significantly decreased e-cadherin levels in asthma patients with epithelial shedding
indicating a potential role of e-cadherin loss in epithelial shedding.77
Heijink et al (2007)110 investigated the signalling role of e-cadherin. E-cadherin is known to
reduce EGFR ligand binding and co-localize with EGFR to limit its movement in the plasma
membrane. E-cadherin knock-down by siRNA resulted in increased EGFR phosphorylation and
increased activation of downstream transcription factor inducers, ERK and p38. Increased EGFR
signalling, due to decreased e-cadherin, resulted in increased production of TARC (CCL17), a Th2
recruiting chemokine and TSLP, a potent Th2 inducing cytokine.110
E-cadherin is a multi-functional protein that plays an important role in the development and
pathogenesis of asthma. Future studies will continue to unravel the functional role of e-cadherin
variants in allergic disease.
- 23 -
α-catenin
α-catenin is a scaffolding protein which regulates AJ structure through binding e-cadherin to
the actin cytoskeleton. de Boer et al (2008)108 found significantly reduced levels of α-catenin in the
bronchial epithelia of asthma patients. The α-catenin gene, CTNNA1, is found on chromosome 5q31
near the cytokine cluster which is repeatedly associated with atopy and allergic diseases.108 Together
these indicate a potential role for CTNNA1 in asthma AJ defects.
Protocadherin-1
Protocadherin-1 is a member of the δ1-protocadherin subfamily.113 PCDH1 is located on
chromosome 5q31.1, which has been previously associated with atopy, asthma and atopic
dermatitis.114 PCDH1 has five exons, which can be differentially processed resulting in multiple
protein variants. The two most common variants are a three exon form and a longer five exon form.
Although the 3 exon variant of protocadherin-1 lacks the majority of the intracellular domain it
expresses an additional sequence in exon 2.114-115 Protocadherin-1 is a membrane spanning protein
whose extracellular domain contains seven cadherin repeats and intracellular domain contains three
conserved motifs (CM1, CM2 and CM3), an RVTF consensus domain and a PDZ-domain binding site
at the C-terminal end. The RVTF consensus motif can interact with protein phosphatase-1α (PP1α)
which is a signalling molecule reportedly involved in lung morphogenesis.113-115 There have been
reports that the CM2 and CM3 domains also interact with PP1α, but otherwise their function is
unknown.114-115 PDZ-containing proteins generally bind to the cytoskeleton and function in cell
signalling, either directly or as scaffold proteins.115 Protocadherin-1 is expressed in the brain, skin,
lung and epithelial tissues in mice. In the lungs it is found in fibroblasts and terminally differentiated
bronchial epithelial cells, where it primarily locates to the cell-cell boundaries.115-116 Although the
exact function of protocadherin-1 is unknown, it is thought to be involved in cell adhesion and
maintenance of the epithelial barrier. Due to its interactions with PP1α, primary expression in
terminally differentiated epithelial cells, and possible interaction with SMAD3 it may also be involved
in cell signalling and epithelial cell differentiation.113-115
Protocadherin-1 polymorphisms
In an initial study Koppelman et al (2009)114 identified PCDH1 as a novel BHR susceptibility
gene. They identified 22 polymorphisms in PCDH1, one of which (rs3797054, Ala750Ala) was
significantly associated to BHR in three Dutch populations and one US population. Interestingly, this
SNP does not result in an amino acid substitution. As it is located in the 3’ UTR of exon three it may
influence mRNA stability or splicing. A three base-pair insertion/deletion polymorphism (IVS3-116)
was also associated with BHR risk in two Dutch populations and two US populations. This
polymorphism is also located in the 3’ UTR of exon three. Both rs3797054 and IVS3-116 were also
significantly associated with asthma risk. The major allele of a polymorphism in the fifth cadherin
repeat (rs3822357, Ala514Thr) was also associated with BHR in a US and a UK population.114
Toncheva et al (2012)116 attempted to replicate these results. As PCDH1 is located on
chromosome 5q31, nearby a cytokine cluster previously linked to atopy, asthma and atopic
dermatitis, they performed linkage disequilibrium (LD) analysis to confirm that PCDH1 is itself
associated to BHR. There was no LD between PCDH1 and surrounding genes, indicating that it is
indeed a susceptibility gene for BHR. However, they were unable to find a significant association of
rs3797054, rs3822357 or IVS3-116 with BHR in their cross-sectional German population. They did
find a protective association of rs7719391 to asthma. Upon further analysis, they found no
polymorphic associations to atopic asthma and two SNPs (rs2974704 and rs11167761) that were
significantly associated to non-atopic asthma.116 Their failure to replicate the results of Koppelman
- 24 -
et al (2012)114 may be due to different determinants of BHR or due to the small, underpowered
study design.116
Although PCDH1 polymorphisms have been associated with BHR and asthma, the functional
role of these variants is unknown. Koning et al (2012)117 have also found an association of the IVS3116 variant of PCDH1 with AD in two Dutch populations. Future studies will be needed to determine
the function of the various polymorphic forms and splice variants in the bronchial epithelium and
the epidermis. The association of PCDH1 polymorphisms to susceptibility risk of asthma and atopic
dermatitis also needs to be confirmed in larger studies.
Filaggrin
Filaggrin is not expressed in the bronchial epithelium118 however, numerous studies have
found an association between FLG mutations and asthma when it co-presents with AD.32-33,39,42,44 The
common interpretation is that a decreased skin barrier leads to increased allergen sensitization
encouraging the atopic march towards asthma.32,39,42 Rogers et al (2007 and 2008)44,119 however,
disagree with this interpretation. Similar to Palmer et al (2006)32 they have found an association
between FLG mutations and asthma with AD and no association of FLG mutations in asthma without
AD. The lack of association between FLG polymorphisms and asthma alone has led them to
conclude that FLG mutations do not predispose towards asthma at all, but rather that FLG mutations
predispose to AD and AD predisposes to asthma as part of the atopic march.44,119
It is interesting to note that asthma patients with FLG polymorphisms have been found to
have more severe disease independent of AD status.42,120 Although FLG mutations may not directly
increase the risk of developing asthma, they seem to interact with other factors to increase disease
severity or encourage the atopic march.
Serine protease inhibitor, Kazal-type, 5
Asthma has previously been linked to the 5q31-33 chromosomal region where SPINK5 is
located.121 In addition, LEKTI is an inhibitor of tryptase, an important asthma mediator.57 Due to this,
SPINK5 has been investigated for a role in asthma development and some associations between
asthma and SPINK5 polymorphisms have been reported. Walley et al (2001)58 found a weak
association of the E420K polymorphism to asthma and Kabesch et al (2004)62 repeated this finding in
a large German population-based study. However, in more cases the association is not found.61-62,121
Recently, Jongepier et al (2005)121 found no association of SPINK5 polymorphisms with asthma in
two Dutch populations. They also found that SPINK5 is not expressed in the lungs. This, combined
with the lack of asthma over-expression in Netherton syndrome patients, indicates that SPINK5
polymorphisms likely do not increase the risk of developing asthma.121
- 25 -
Current Therapies and Future Perspective
Disruption of tissue homeostasis, due to genetic and environmental factors, is now seen as
fundamental in the initiation and perpetuation of allergic inflammation and disease. This paradigm
shift has led to changes in therapeutic approaches and the development of novel, tissue-targeted
treatments.
Atopic Dermatitis
Topical corticosteroids are the standard treatment for acute lesions in atopic dermatitis.
They interact with glucocorticoid receptors on immune cells, keratinocytes and fibroblasts to block
the transcription of pro-inflammatory genes.122 Although topical corticosteroids are quite effective
at reducing symptoms and disease severity they fail to address the underlying mechanism of disease
and there is some evidence that they further disrupt the skin barrier and increase the risk of
microbial infection.23 Topical emollients are also widely used to treat AD. They are creams or
ointments that create a superficial barrier of non-physiologic lipids, such as petrolatum, lanolin
mineral oil or silicon.122-123 Emollients have been quite successful in treating AD symptoms; however
there is some evidence that the non-physiologic lipids may obstruct the underlying barrier defects
rather than correct them.23
In healthy skin the lipid lamellae is composed of approximately 50% ceramides, 25%
cholesterol and 10-20% fatty acids. In AD skin these three lipids are significantly reduced. Emollient
therapies containing these physiologic lipids have recently become available. When topically applied,
physiologic lipids are thought to penetrate the SC and get processed into lamellar bodies for
secretion into the SC where they can restore the epidermal barrier.23,123There is some evidence that
on their own or in the incorrect ratio they may have a negative effect on barrier repair.123 Ceramide
dominant treatments, such as EpiCeram® which uses a 3:1:1 ratio of ceramides, cholesterol and fatty
acids, seems to provide optimal barrier repair.23,123 CeraVe,® another ceramide based cream which is
under development uses multilamellar vesicle emulsions to slowly release the lipids over a 24 hour
period. This is promising as a once-daily barrier-repair therapy for AD.123 In addition, gene repair
therapies may be available in the coming years. Ex-vivo lentiviral transduction of keratinocytes with
SPINK5 has been shown to restore LEKTI production in human NS epidermal cells grafted onto a
mouse model.124 As the available treatments for AD are very effective, not only at addressing the
symptoms but also at repairing the underlying barrier defects, this type of treatment will likely only
be used in cases of sever barrier breakdown, such as in NS.
Future research should focus on identifying biomarkers of early disease to detect at-risk
children. Prophylactic treatment of these children with barrier repairing creams could potentially
prevent allergen sensitization and development of atopic dermatitis. There have been a few studies
indicating that prophylactic emollient treatments in infants may reduce the incidence of atopic
dermatitis and enhance skin barrier.122
Asthma
Asthma is a very heterogeneous disease; therefore it is unlikely that a single treatment or
therapy with be completely effective. However, with a clearer definition of asthma subtypes,
targeted therapies could be quite successful.18,84 Many immune targeted therapies, with varying
levels of success are under development. Anti-IgE therapy, which was expected to be curative for
allergic diseases, has been disappointing. Omalizumab, an anti-IgE monoclonal antibody (mAb), has
shown some benefit in patients with moderate to severe asthma in that it can reduce systemic IgE
levels, FcєRI on mast cells and basophils and can reduce eosinophil levels in some patients. However,
it likely fails to reduce site-specific IgE levels and has low cost-effectiveness.4,18 Many cytokine
targeted therapies are also under development. In clinical trials some patients show improved
- 26 -
pulmonary function and reduced symptoms in response to anti-IL-12 mAbs. Multiple anti-IL-4 mAbs
are also undergoing clinical trials; however, their success has been much lower than anticipated. This
may be due to the ability of IL-13 to signal through the same receptor complex negating the effect of
IL-4 reduction. Anti-IL-13 mAbs are also under development. A phase 2 clinical trial shows some
promise in certain subgroups of asthma. Anti-IL-5 mAbs were expected to greatly reduce asthma
symptoms; although they reduce eosinophil levels to some extent, they did not result in any
significant lung function improvement. Blocking TSLP signalling with a soluble antagonist and small
molecule targeting of chemokine receptors including CX3CR1 on CD4+ T cells and CCR3 on
eosinophils have also shown some promise.2,18
Allergen specific immunotherapy (SIT) is the only curative treatment for asthma; however it
is controversial due to the risk of anaphylaxis. In randomized control trials SIT has been fairly
successful in reducing asthma symptoms and decreasing medication use, however there is a high risk
of local and systemic side effects and this treatment does not seem to improve overall lung
function.125 Currently the standard asthma treatment is inhaled corticosteroids (ICS) alone or
combined with long-acting bronchodilators.79 Corticosteroids are immunosuppressive agents that
target eosinophilic airway inflammation, while bronchodilators aim to reverse bronchoconstriction
and relieve breathing difficulties.4,125 In most patients this treatment is very successful in suppressing
asthma symptoms.125
Although the currently available asthma treatments are quite effective, they primarily target
the symptoms or immune mediated mechanisms of the disease.79 Given the critical role of the
epithelium in both the disease initiation and perpetuation, novel therapeutic design should focus on
epithelial barrier repair and the prevention of disease. Xiao et al (2011)107 have shown that topically
applied (inhaled) epidermal growth factor (EGF) can increase TJ numbers and improve the epithelial
barrier function in asthmatic patients without causing uncontrolled cell proliferation or goblet cell
hyperplasia. There is recent evidence that ICS may increase tight junction integrity and aid in barrier
repair as well as suppress excess cell proliferation, potentially decreasing airway remodelling.126-127
Therapeutically targeting the epithelial may prevent inflammation, rather than simply suppressing it.
Some studies of PRR modulation have shown promise. In mouse models TLR7/8 agonists significantly
reduce airway inflammation and prevent airway remodelling. TLR7/8 stimulation resulted in
decreased cytokine production, decreased inflammatory cell infiltration in the lungs, decreased
smooth muscle proliferation and decreased goblet cell hyperplasia and metaplasia.128-129 Targeting
the airway epithelium with a combination of TLR agonists and barrier repair factors may prevent
allergen entry and the initiation of an allergic inflammatory response.
Many asthma patients develop disease as part of the atopic march. These patients will have
presented with atopic dermatitis before developing asthma. Early identification of children at-risk of
developing asthma subsequent to AD could allow for administration of preventative treatments.
This could include topical epidermal barrier repair treatment to prevent systemic allergen
sensitization and prevention of respiratory viral infections to avoid compromising the airway
epithelium. Many of the genetic loci associated with asthma are distinct from the loci associated
with AD indicating that, while in some cases AD can increase the risk of asthma development in line
with the atopic march, asthma is also a distinct disease.29 As such, potentially preventative
interventions also need to be targeted at asthma directly. In most cases of childhood diagnosed
asthma, wheezing precedes disease development.84 Viral infections are often the cause of this
wheezing and can cause epithelial barrier damage increasing risk of asthma development.
Vaccinating young children against RSV or HRV, the two most common respiratory viruses associated
with asthma risk, may decrease asthma incidence. Anti-RSV antibodies are under development but
not yet available for clinical testing in children.84 Edlmayr et al (2009)130 developed a novel vaccine
for allergy and rhinovirus infection. They created a fusion protein of VP1, a major HRV surface
protein, and a synthetic Phl p 1 protein of a major grass pollen allergen. Vaccination with this fusion
protein resulted in a strong protective IgG antibody response against both the allergen and the
virus.130 This study indicates the potential of prophylactic allergic vaccination.
- 27 -
Conclusions
Allergic diseases arise from complex interactions between genetic predispositions and
environmental factors. The classical view of allergic diseases is based primarily on immune
dysregulation; however, this alone cannot fully explain their development. Over 40% of the western
population is atopic however, only 7-10% percent of these develop asthma and only a small
proportion develops atopic dermatitis (AD).73-74 The recent association of tissue restricted factors
with asthma and AD indicates that local tissue factors play a critical role in determining the location
of disease establishment.17,19 The primary tissue role is as a barrier against allergen entry and
sensitization. Disruption of the barrier by physical damage or allergen activity can result in the
production of Th2 cytokines and chemokines driving the allergic immune response.26,78 The skin and
the airway epithelium are the primary sites susceptible to allergen entry. As such, in healthy
individuals, they are highly structured barriers.
The skin is a multi-layer barrier which, when functional, protects against water loss and
invasion of unwanted or harmful environmental materials. The barrier consists of the stratum
corneum with its associated structural and regulatory components, the lipid lamellae, the acid
mantle and tight junctions.17,21 Intrinsic deficiencies at any of these levels predisposes towards
allergen sensitization and the development of atopic dermatitis. Genetic polymorphisms in FLG,
KLK7, SPINK5 and CSTA can result in a breakdown of the SC barrier.32,54,58,64 FLG polymorphisms can
also affect the pH mantle and lipid lamellae barriers and CLDN1 polymorphisms can disrupt the TJ
barrier.32,68 Although it can be difficult to unravel the exact contribution of these polymorphisms to
disease development, they all indicate that the epidermal barrier is an essential protective force
against allergic disease in the skin.
The airway epithelium is a single layer barrier that depends on the adhesive forces between
neighbouring cells to prevent allergen, microbial and chemical entry. Tight and adherens junctions
are the primary enforcers of the epithelial barrier. Cell adhesion genes, CLDN1, OCLN, ZO-1, CDH1,
PCDH1 and CTNNA1, have all been associated with increased risk of asthma highlighting their
essential role in epithelial barrier integrity and the protective nature of the barrier against allergen
sensitization and disease development.107-108,112 The association of genes functionally involved in
airway remodelling in asthma, such as ADAM33 and CLDN1, indicate that tissue related factors are
also involved in disease progression.69,91 A predisposition to epithelial barrier dysfunction and
aberrant barrier repair may lead to allergen sensitization and development of an overactive immune
response in asthma patients.
The current understanding of tissue barrier dysregulation at the core of allergic diseases has
opened the door to novel therapeutic options. The emerging treatments do not only focus on
immune suppression, but aim to repair the underlying tissue barrier deficiencies. Early identification
of susceptible individuals and prophylactic barrier repair therapies may reduce the incidence of
allergic diseases in the future.
Acknowledgements
I would like to thank Dr Knol for his supervision.
- 28 -
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
Kay, A.B. 2001. Allergy and Allergic Diseases: first of two parts. The New England Journal of Medicine 344(1):3037
Holgate, S.T. 2012. Innate and adaptive immune responses in asthma. Nature Medicine 18(5):673-683
Kay, A.B. 2000. Overview of ‘allergy and allergic diseases: with a view to the future’. British Medical Bulletin
56(4):843-864
Galli, S.J. and Tsai, M. 2012. IgE and mast cells in allergic disease. Nature Medicine 18(5):693-704
Islam, S.A. and Luster, A.D. 2012. T cell homing to epithelial barriers in allergic disease. Nature Medicine
18(5):705-715
Liu F. et al. 2011. IgE, mast cells, and eosinophils in atopic dermatitis. Clinical Reviews in Immunology 41:298-310
Spergel, J.M. and Paller, A.S. 2003. Atopic dermatitis and the atopic march. Journal of Allergy and Clinical
Immunology 112:S118-S127
Barnes, K.C. 2000. Evidence for common genetic elements in allergic disease. Journal of Allergy and Clinical
Immunology 106(5):S192-S200
Woodfolk, J.A. 2005. A new paradigm for immunoglobulin E in allergic diseases. Current Allergy and Asthma
Reports 5:227-232
Jutel, M. and Akdis, C.A. 2011. T-cell subset regulation in atopy. Current Allergy and Asthma Reports 11:139-145
Williams, C.M.M. et al. 2012. Cytokine pathways in allergic disease. Toxicologic Pathology 40:205-215
Ou, L.S. and Huang, J.L. 2007. Cellular aspects of atopic dermatitis. Clinical Reviews in Allergy and Immunology
33:191-198
Proud, D. and Leigh, R. 2011. Epithelial cells and airway diseases. Immunological Reviews 242:186-204
Kiyohara, C. et al. 2008. Genetic susceptibility to atopic dermatitis. Allergology International 57:39-56
Riis, J.L., et al. 2011. Kinetics and differential expression of the skin-related chemokines CCL27 and CCL17 in
psoriasis, atopic dermatitis and allergic contact dermatitis. Experimental Dermatology 20(10):789-794
Wenzel, S.E. 2012. Asthma phenotypes: the evolution from clinical to molecular approaches. Nature Medicine
18(5):716-725
Cork, M.J. et al. 2009. Epidermal barrier dysfunction in atopic dermatitis. Journal of Investigative Dermatology
129:1892-1908
Akdis, C.A. 2012. Therapies for allergic inflammation: refining strategies to induce tolerance. Nature Medicine
18(5):736-749
Holgate, S.T. 2011. The sentinel role of the airway epithelium in asthma pathogenesis. Immunological Reviews
242: 205-219
Werfel, T. 2009. The role of leukocytes, keratinocytes, and allergen-specific IgE in the development of atopic
dermatitis. Journal of Investigative Dermatology 129:1878-1891
Kubo, A. et al. 2012. Epidermal barrier dysfunction and cutaneous sensitization in atopic diseases. The Journal of
Clinical Investigation 122(2):440-447
Wolf, R. and Wolf, D. 2012. Abnormal epidermal barrier in the pathogenesis of atopic dermatitis. Clinics in
Dermatology 30:329-334
Elias, P.M. and Wakefield, J.S. 2011. Therapeutic implications of a barrier-based pathogenesis of atopic
dermatitis. Clinical Reviews in Allergy and Immunology 41:282-295
Boguniewicz, M. and Leung, D.Y.M. 2011. Atopic dermatitis: a disease of altered skin barrier and immune
dysregulation. Immunological Reviews 242:233-246
Kaplan, D.H. et al. 2012. Early immune events in the induction of allergic contact dermatitis. Nature Reviews in
Immunology 12:114-124
Takai, T. 2011. TSLP expression: cellular sources, triggers, and regulatory mechanisms. Allerology International
61:3-17
Uchi, H. et al. 2000. Cytokines and chemokines in the epidermis. Journal of Dermatological Science 24(Suppl
1.):S29-S38
Boguniewicz, M., and Leung, D.Y.M. 2001. Pathophysiologic mechanisms in atopic dermatitis. Seminars in
Cutaneous Medicine and Surgery 20(4):217-225
Bowcock, A.M. and Cookson, W.O.C.M. 2004. The genetics of psoriasis, psoriatic arthritis, and atopic dermatitis.
Human Molecular Genetics 13(1):R43-R55
Runswick, S., et al. 2007. Pollen proteolytic enzymes degrade tight junctions. Respirology 12:834-842
Incorvaia, C. et al. 2008. Allergy and the skin. Clinical and Experimental Immunology 153(Suppl. 1):27-29
Palmer, C.N.A. et al. 2006. Common loss-of-function variants of the epidermal barrier protein filaggrin are a
major predisposing factor for atopic dermatitis. Nature Genetics 38(4):441-446
Weidinger, S. et al. 2008. Filaggrin mutations, atopic eczema, hay fever, and asthma in children. Journal of
Allergy and Clinical Immunology 121:1203-1209
Brown, S.J. and McLean, W.H.I. 2012. One remarkable molecule: filaggrin. Journal of Investigative Dermatology
132:751-762
O’Regan, G.M. et al. 2008. Filaggrin in atopic dermatitis. Journal of Allergy and Clinical Immunology 122:698-693
- 29 -
36. Osawa, R. et al. 2011. Filaggrin gene defects and the risk of developing allergic disorders. Allergology
International 60:1-9
37. Kezic, S. et al. 2011. Levels of filaggrin degredation products are influenced by both filaggrin genotype and atopic
dermatitis severity. Allergy 66:934-940
38. Smith, F.J.D. et al. 2006. Loss-of-function mutations in the gene encoding filaggrin cause ichthyosis vulgaris.
Nature Genetics 38(3):337-342
39. Weidinger, S. et al. 2006. Loos-of-function variations within the filaggrin gene predispose for atopic dermatitis
with allergic sensitizations. Journal of Allergy and Clinical Immunology 118:214-219
40. Barnes, K.C. 2010. An update on the genetics of atopic dermatitis: scratching the surface in 2009. Journal of
Allergy and Clinical Immunology 125:16-29
41. Morar, N. et al. 2007. Filaggrin mutations in children with severe atopic dermatitis. Journal of Investigative
Dermatology 127:1667-1672
42. Irvine, A.D. et al. 2011. Filaggrin mutations associated with skin and allergic diseases. The New England Journal of
Medicine 365:1315-1327
43. Holloway, J.W. et al. 2010. Genetics of allergic disease. Journal of Allergy and Clinical Immunology 125:S81-S94
44. Rogers, A.J. et al. 2007. Filaggrin mutations confer susceptibility to atopic dermatitis but not to asthma. Journal
of Allergy and Clinical Immunology 120:1332-1337
45. Irvine, A.D, and Mclean, W.H. 2006. Breaking the (un)sound barrier: filaggrin is a major gene for atopic
dermatitis. Journal of Investigative Dermatology 126(6):1200-1202
46. Ginger, R.S. et al. 2005. Filaggrin repeat number polymorphism is associated with dry skin phenotype. Archives
of Dermatological Research 297:235-241
47. Hoste, E. et al. 2011. Caspase 14 is required for filaggrin degradation to natural moisturizing factors in the skin.
Journal of Investigative Dermatology 131:2233-2241
48. Meyer-Hoffert, U. 2009. Reddish, scaly, and itch: how proteases and their inhibitors contribute to inflammatory
skin diseases. Archivum Immunologiae et Therapia Experimentalis 57:345-354
49. Furio, L., and Hovnanian, A. 2011. When activity requires breaking up: LEKTI proteolytic activation cascade for
specific proteinase inhibition. Journal of Investigative Dermatology 131:2169-2173
50. Rawlings, A.V. and Voegeli, R. 2012. Stratum corneum proteases and dry skin conditions. Cell and Tissue Research
epub ahead of print 9 Oct 2012.
51. Fortugno, P., et al. 2012. The 420K LEKTI variant alters LEKTI proteolytic activation and results in protease
deregulation: implications for atopic dermatitis. Human Molecular Genetics 21(19):4187-4200
52. Zhao, L.P. et al. 2012. Association of SPINK5 gene polymorphisms with atopic dermatitis in Northeast China.
Journal of the European Academy of Dermatology and Venerology 26:572-577
53. Elias, P.M and Wakefield, J.S. 2011. Therapeutic implications of a barrier-based pathogenesis of atopic dermatitis.
Clinical Reviews in Allergy and Immunology 41:282-295
54. Vasilopoulos, Y., et al. 2004. Genetic association between an AACC insertion in the 3’ UTR of the stratum
corneum chymotryptic enzyme gene and atopic dermatitis. Journal of Investigative Dermatology 123:62-66
55. Weidinger, S., et al. 2008. Analysis of the individual and aggregate genetic contributions of previously identified
serine peptidase inhibitor Kazal type 5 (SPINK5), kallikrein-related peptidase 7 (KLK7), and filaggrin (FLG)
polymorphisms to eczema risk. Journal of Allergy and Clinical Immunology 122:560-568
56. Norgett, E.E. and Kelsell, D.P. 2002. SPINK5: both rare and common skin disease. TRENDS in Molecular Medicine
8(1):7
57. Moffat, M.F. 2004. SPINK5: a gene for atopic dermatitis and asthma. Clinical and Experimental Allergy 34:325-327
58. Walley, A.J. et al. 2001. Gene polymorphisms in Netherton and common atopic disease. Nature Genetics 29:175178
59. Ishida-Yamamoto, A., et al. 2005. LEKTI is localized in lamellar granules, separated from KLK5 and KLK7, and is
secreted in the extracellular spaces of the superficial stratum granulosum. The Journal of Investigative
Dermatology 124:360-366
60. Kato, A., et al. 2003. Association of SPINK5 gene polymorphisms with atopic dermatitis in the Japanese
population. British Journal of Dermatology 148:665-669
61. Nishio, Y., et al. 2003. Association between polymorphisms in the SPINK5 gene and atopic dermatitis in the
Japanese. Genes and Immunity 4:515-517
62. Kabesch, M., et al. 2004. Association between polymorphisms in serine protease inhibitor, kazal type 5 and
asthma phenotypes in a large German population sample. Clinical and Experimental Allergy 34:340-345
63. Di, W-L. et al. 2009. A heterozygous null mutation combined with the G1258A polymorphism of SPINK5 causes
impaired LEKTI function and abnormal expression of skin barrier proteins. British Journal of Dermatology
161:404-412
64. Vasiolopoulos, Y., et al. 2007. A nonsynonymous substitution of cystatin A, a cysteine protease inhibitor of house
dust mite protease, leads to decreased mRNA stability and shows a significant association with atopic dermatitis.
Allergy 62:514-519
65. Kato, T., et al. 2005. Cystatin A inhibits IL-8 production by keratinocytes stimulated with Der p1 and Der pf:
biochemical skin barrier against mite cysteine proteases. Journal of Allergy and Clinical Immunology 116:169-176
- 30 -
66. Ogawa, T., et al. 2008. Upregulation of the release of granulocyte-macrophage colony-stimulating factor from
keratinocytes stimulated with cysteine protease activity of recombinant major mite allergens, Der f 1 and Der p 1.
International Archives of Allergy and Immunology 146:27-35
67. Seguchi, T., et al. 1996. Decreased expression of filaggrin in atopic skin. Archives of Dermatological Research
288(8):442-446
68. De Benedetto, A., et al. 2011. Tight junction defects in patients with atopic dermatitis. Journal of Allergy and
Clinical Immunology 127:773-778
69. Fujita, H., et al. 2011. Claudin-1 expression in airway smooth muscle exacerbates airway remodelling in asthmatic
subjects. Journal of Allergy and Clinical Immunology 127:1612-1621
70. Furuse, M., et al. 2002. Claudin-based tight junctions are crucial for the mammalian epidermal barrier: a lesson
from claudin-1-deficient mice. Journal of Cell Biology 156(6):1099-1111
71. De Benedetto, A., et al. 2011. Reductions in claudin-1 may enhance susceptibility to herpes simplex virus 1
infections in atopic dermatitis. Journal of Allergy and Clinical Immunology 128(1):242-246
72. Nawijn, M.C., et al. 2011. E-cadherin: gatekeeper of airway mucosa and allergic sensitization. Trends in
Immunology 32(6):248-255
73. Holgate, S.T. 2007. Epithelium dysfunction in asthma. Journal of Allergy and Clinical Immunology 120(6):12331244
74. Holgate, S.T. 2008. The airway epithelium is central to the pathogenesis of asthma. Allergology International
57:1-10
75. Tam, A. et al. 2011. The airway epithelium: more than just a structural barrier. Therapeutic Advances in
Respiratory Disease 5(4):255-273
76. Rydell-Törmänen, K., et al. 2012. Smooth muscle in tissue remodeling and hyper-reactivity: airways and arteries.
Pulmonary Pharmacology In Press
77. Trautmann, A., et al. 2005. Apoptosis and loss of adhesion of bronchial epithelial cells in asthma. International
Archives of Allergy and Immunology 138:142-150
78. Bartemes, K.R. and Kita, H. 2012. Dynamic role of epithelium-derived cytokines in asthma. Clinical Immunology
143:222-235
79. Lambrecht, B.N. and Hammad, H. 2012. The airway epithelium in asthma. Nature Medicine 18(5):684-692
80. Mattial, P. et al. 2011. Allergy as an epithelial barrier disease. Clinical and Translational Allergy 1:5-12
81. Kay, A.B. 2001. Allergy and Allergic Diseases: second of two parts. The New England Journal of Medicine
344(2):109-113
82. Holgate, S.T. et al. 2009. The role of the airway epithelium and its interaction with environmental factors in
asthma pathogenesis. Proceedings of the American Thoracic Society 6:655-659
83. Sharma, N., et al. 2011. Role of ADAM33 gene and associated single nucleotide polymorphisms in asthma. Allergy
and Rhiniology 2:e63-e70
84. Holt, P.G. and Sly, P.D. 2012. Viral infections and atopy in asthmas pathogenesis: new rationales for asthma
prevention and treatment. Nature Medicine 18(5):726-735
85. Russell, S.L., et al. 2012. Early life antibiotic-driver changes in microbiota enhance susceptibility to allergic
asthma. EMBO Reports 13:440-447
86. Beck, J.M., et al. 2012. The microbiome of the lung. Translational Research 160:258-266
87. Busse, W.W. and Lemanske, R.F. 2001. Asthma. The New England Journal of Medicine 244(5):350-362
88. Jansen, D.F., et al. 1997. (A)Symptomatic bronchial hyper-responsiveness and asthma. Respiratory Medicine
91:121-134
89. Davies, D.E. 2009. The role of the epithelium in airway remodelling in asthma. Proceedings of the American
Thoracic Society 6:678-682
90. Lazaar, A.L. and Panettieri, R.A. 2003. Is airway remodelling clinically relevant in asthma? American Journal of
Medicine 115:652-659
91. Van Eerdewegh, P., et al. 2002. Association of the ADAM33 gene with asthma and bronchial
hyperresponsiveness. Nature 418:426-430
92. Wolfsburg, T.G., et al. 1995. ADAM, a novel family of membrane proteins containing a disintegrin and
metalloprotease domain: multipotential functions in cell-cell and cell-matrix interactions. The Journal of Cell
Biology 131(2):275-278
93. Chae, SC., et al. 2003. Identification of novel polymorphisms in the ADAM33 gene. Journal of Human Genetics
48:278-281
94. Brocker, C.N., et al. 2009. Evolutionary divergence and functions of the ADAM and ADAMTS gene families.
Human Genomics 4(1):43-55
95. Powell, R.M., et al. 2003. ADAM33: a novel therapeutic target for asthma. Expert Opinions on Therapeutic Targets
7(4):485-494
96. Koppelman, G.H. and Sayers, I. 2011. Evidence of a genetic contribution to lung function decline in asthma.
Journal of Allergy and Clinical Immunology 128:479-484
97. Howard, T.D., et al. 2003. Association of a disintegrin and metalloprotease 33 (ADAM33) gene with asthma in
ethnically diverse populations. Journal of Allergy and Clinical Immunology 112:717-722
- 31 -
98. Lind, D.L., et al. 2003. ADAM33 is not associated with asthma in Puerto Rican or Mexican populations. American
Journal of Respiratory and Critical Care Medicine 168:1312-1316
99. Raby, B.A., et al. 2004. ADAM33 polymorphisms and phenotype associations in childhood asthma. Journal of
Allergy and Clinical Immunology 113:1071-1078
100. Tripathi, P., et al. 2012. Haplotype association of ADAM33 (T+1, S+1 and V-3) gene variants in genetic
susceptibility to asthma in Indian population. Annals of Human Biology 39(6):479-483
101. Werner, M., et al. 2004. Asthma is associated with single-nucleotide polymorphisms in ADAM33. Clinical and
Experimental Allergy 34(1):26-31
102. Holgate, S.T., et al. 2006. ADAM33: a newly identified protease involved in airway remodelling. Pulmonary
Pharmacology and Therapeutics 19:3-11
103. Lopez-Guisa, J.M., et al. 2012. Airway epithelial cells from asthmatic children differentially express
proremodelling factors. Journal of Allergy and Clinical Immunology 129:990-997
104. Yang, Y., et al. 2008. Epigenetic mechanisms silence a disintegrin and metalloprotease 33 expression in bronchial
epithelial cells. Journal of Allergy and Clinical Immunology 121:1393-1399
105. Lee, J.Y., et al. 2006. A disintegrin and metalloproteinase 33 protein in patients with asthma: relevance to airflow
limitation. American Journal of Respiratory and Critical Care Medicine 173:729-735
106. Puxeddu, I., et al. 2008. The soluble form of a disintegrin and metalloprotease 33 promotes angiogenesis:
implications for airway remodelling in asthma. Journal of Allergy and Clinical Immunology 121:1400-1406
107. Xiao, C., et al. 2011. Defective epithelial barrier function in asthma. Journal of Allergy and Clinical Immunology
128:549-556
108. de Boer, W.I., et al. 2008. Altered expression of epithelial junctional proteins in atopic asthma: possible role in
inflammation. Canadian Journal of Physiology and Pharmacology 86:105-112
109. Ahdieh, M., et al. 2001. Lung epithelial barrier function and wound healing are decreased by IL-4 and IL-13 and
enhanced by IFN-γ. American Journal of Physiology-Cell Physiology 281:C2029-C2038
110. Heijink, I.H., et al. 2007. Down-regulation of E-cadherin in human bronchial epithelial cells leads to epidermal
growth factor receptor-dependent Th2 cell-promoting activity. The Journal of Immunology 178:7678-7685
111. Ierodiakonou, D., et al. 2011. E-cadherin gene polymorphisms in asthma patients using inhaled corticosteroids.
European Respiratory Journal 38:1044-1052
112. Hackett, T.L., et al. 2011. Intrinsic phenotypic differences of asthmatic epithelium nad its inflammatory responses
to respiratory syncytial virus and air pollution. American Journal of Respiratory Cell and Molecular Biology
45:1090-1100
113. Redies, C., et al. 2005. Δ-protocadherins: unique structures and functions. Cellular and Molecular Life Sciences
62:2840-2852
114. Koppelman, G.H., et al. 2009. Identification of PCDH1 as a novel susceptibility gene for bronchial
hyperresponsiveness. American Journal of Respiratory and Critical Care Medicine 180:929-935
115. Koning, H., et al. 2012. Characterization of protocadherin-1 expression in primary bronchial epithelial cells:
association with epithelial cell differentiation. The FASEB Journal 26:439-448
116. Toncheva, A.A., et al. 2012. Genetic variants in Protocadherin-1, bronchial hyperresponsiveness, and asthma
subphenotypes in German children. Pediatric Allergy and Immunology 23:636-641
117. Koning, H., et al. 2012. Protocadherin-1 polymorphisms are associated with eczema in two Dutch birth cohorts.
Pediatric Allergy and Immunology 23:270-277
118. Ying, S. et al. 2006. Lack of filaggrin expression in the human bronchial mucusa. Journal of Allergy and Clinical
Immunology 118(6):1386-1388
119. Letter to the editor. McLean, W.H.I. et al. 2008. Filaggrin variants confer susceptibility to asthma. Journal of
Allergy and Clinical Immunology 121(5):1294-1295
120. Palmer, C.N.A. et al. 2007. Filaggrin null mutations are associated with increased asthma severity in children and
young adults. Journal of Allergy and Clinical Immunology 120:64-68
121. Jongepier, H., et al. 2005. Polymorphisms in SPINK5 are not associated with asthma in a Dutch population.
Journal of Allergy and Clinical Immunology 115:486-492
122. Valdman-Grinshpoun, Y., et al. 2012. Barrier-restoring therapies in atopic dermatitis: current approaches and
future perspectives. Dermatology Research and Practice 2012: 6 pages
123. Saijć, D., et al. 2012. A look at epidermal barrier function in atopic dermatitis: physiologic lipid replacement and
the role of ceramides. Skin Therapy Letters 17(7):6-9
124. Di, W-L., et al. 2011. Ex-vivo gene therapy restores LEKTI activity and corrects the architecture of Netherton
syndrome-derived skin grafts. Molecular Therapy 19(2):408-416
125. Quirce, S., et al. 2012. Emerging drugs for asthma. Expert Opinion on Emerging Drugs 17(2):219-237
126. Sekiyama, A., et al. 2012. Glucocorticoids enhance airway epithelial barrier integrity. International Pharmacology
12:350-357
127. Wadsworth, S.J., et al. 2006. Glucocorticoids increase repair potential in a novel in vitro human airway epithelial
wounding model. Journal of Clinical Immunology 26(4):376-387
128. Camateros, P., et al. 2007. Chronic asthma-induced airway remodelling is prevented by toll-like receptor-7/8
ligand S28463. American Journal of Respiratory and Critical Care Medicine 125:1241-1249
- 32 -
129. Xirakia, C., et al. 2010. Toll-like receptor 7-triggerd immune response in the lung mediates acute and long-lasting
suppression of experimental asthma. American Journal of Respiratory and Critical Care Medicine 181:1207-1216
130. Edlmayr, J., et al. 2009. A combination vaccine for allergy and rhinovirus infections based on rhinovirus-derived
surface protein VP1 and a non-allergenic peptide of the major timothy grass pollen allergen Phl p 1. Journal of
Immunology 182:6298-6306
- 33 -
Download