You are on page 1of 9

184

CHA PTER 1 4

Genetics and Aetiology of Atopic Dermatitis


Elke Rodriguez & Stephan Weidinger
Department of Dermatology, Allergology and Venereology, University Hospital Schleswig‐Holstein, Campus Kiel, Kiel, Germany

Introduction, 184 Conclusions and future directions, 190


Genes implicated in the aetiology of eczema, 187
ATOPIC DERMATITIS

Abstract by approximately 20% of patients, and they are neither necessary


SECTION 3:

nor sufficient to cause the disease. Most of the other loci harbour
candidate genes with functions related to immune regulation, in
Atopic dermatitis or eczema represents a typical multifactorial dis-
particular innate signalling and T‐cell specification and activation,
ease in that there is an individual hereditary predisposition that is
but the causative genes or gene products as well as the underlying
triggered by environmental and lifestyle factors. The heritability
molecular mechanisms remain to be identified and characterized.
of atopic dermatitis, i.e. the genetically determined proportion of
Together, the established susceptibility loci explain about 20% of
the total variance observed in a population, is estimated to be 70–
the total estimated heritability. This ‘missing heritability’ may be
80%. The possibilities for deciphering inherited risk factors have
explained by gene interactions, rare gene variants not yet discov-
impressively increased during the last years. As a result, more than
ered and epigenetic changes, but may also relate to an overestima-
30 susceptibility loci have been successfully identified. Null vari-
tion of the heritable disease component and the phenotypic com-
ants in the gene encoding the key epidermal barrier protein filag-
plexity of the disease.
grin are the strongest single risk factor, but they are carried only

Key points • Two notable exceptions are loss‐of‐function variants in the gene
FLG, which cause the dry skin condition ichthyosis vulgaris due
to a deficiency of the epidermal structural protein filaggrin, and
• Atopic dermatitis has a very strong heritable component.
infrequent variants in the gene encoding GARP, which lower the
• Genetic risk loci for atopic dermatitis are being discovered
expression of this receptor for latent transforming growth factor
through a range of strategies from linkage studies to genome‐
(TGF)‐β on the surface of regulatory T cells.
wide association studies.
• Known risk loci explain about 20% of the total estimated
• Thus far, more than 30 susceptibility loci for atopic dermatitis
heritability of atopic dermatitis.
have been identified. For most of these regions the causative
genes or gene products remain to be identified and characterized.

been suggested that eczema is part of a syndrome of


Introduction ‘atopic diseases’, in which patients may develop food
Challenges in defining (atopic) dermatitis/ allergy, eczema, asthma and rhinitis in any order and in
eczema any combination over time [4]. Atopy itself is defined as
Eczema (atopic dermatitis, atopic eczema) is the most ‘personal or familial tendency to become sensitized and
common chronic skin disease in infants and children, produce IgE antibodies in response to low doses of
with prevalence rates of up to 30%. It is estimated that allergens, usually proteins’ and to ‘develop typical
approximately 60% of patients with childhood eczema symptoms such as asthma, rhinoconjunctivitis, or
undergo spontaneous remission in early adolescence, but eczema/dermatitis’ [5]. Based on the fact that eczema
up to 50% may have recurrences in adulthood, and the symptoms often precede those of asthma and because
disease can persist into or start in adulthood, making it eczema represents a well‐established risk factor for
one of the most common skin disorders across all ages [1]. asthma, it has been further hypothesized that a suscepti­
Eczema, asthma and rhinitis tend to cluster in the ble child commonly passes through a sequential/over­
same individuals and families [2], and are often accom­ lapping series of phenotypes from eczema and food
panied by elevated levels of total serum IgE antibodies allergy to asthma and subsequently allergic rhinitis
and aberrant IgE‐mediated responses to otherwise (‘atopic march”) [6–9]. While allergic sensitization
harmless environmental agents [3]. Therefore it has ­represents one possible and plausible mechanistic link

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 14 Genetics and Aetiology of Atopic Dermatitis 185

between asthma and eczema, it is notable that a signifi­ Evidence for a genetic basis for eczema
cant proportion of patients with eczema are not ‘atopic’, There is a longstanding recognition that atopic diseases
i.e. they have normal total serum IgE concentrations and cluster in families and are hereditary disorders [18–20].
no specific IgE responses [3,10]. Furthermore, epidemi­ Twin studies showed distinctly higher concordance rates
ological research indicates that sensitization might sim­ among monozygotic than dizygotic twin pairs (for
ply be a shared epiphenomenon and suggests that the eczema 0.23–0.86 vs. 0.15–0.50), and segregation analyses
association between asthma and eczema may occur suggested that genetic factors account for more than 70%
much earlier, i.e. early co‐occurrence of eczema and of the variance in the susceptibility to eczema [21–24].
wheeze progressing to asthma [10–12]. Thus, the role However, unlike monogenic disorders, which are caused
and temporal significance of elevated IgE in eczema is by mutations in a single gene, eczema and atopic disor­
as yet unclear, and the concept of an ‘allergic march’ ders are likely to be complex poly‐ or oligogenetic traits,
may represent an oversimplification of complex disease thought to be the result of a complicated network of
co‐associations. Other discovered mechanisms such as numerous susceptibility loci that exert additive or syner­

ATOPIC DERMATITIS
epidermally produced thymic stromal lymphopoietin gistic effects but may have only a small role when considered
(TSLP)‐mediated lung inflammation may provide in isolation [25]. In addition, interacting environmental

SECTION 3:
­alternative explanations for an early co‐association of factors are thought to precipitate the polygenic risk back­
eczema and asthma [13,14]. ground into disease manifestation [26]. The importance
Eczema has a wide spectrum of clinical presentations of environmental influences is emphasized by the wide
and it is unclear whether it is a single disorder with dif­ ranges in concordance rates, the incomplete concordance
ferent clinical manifestations or a group of syndromes among monozygotic twins and the changing and variable
with unique or overlapping pathophysiological path­ prevalences of atopic symptoms both between countries
ways that open out into a rather uniform clinical pres­ with similar ethnic groups and within countries [27,28].
entation. However, a rigorous definition of patient The dissection of complex traits is further hampered by
subgroups is highly desirable in order to facilitate epi­ phenocopy, incomplete penetrance and genetic heterogeneity
demiological, genetic and clinical investigations. In the [26]. In addition, a set of studies suggested that, particu­
absence of a gold standard or adequate laboratory tests larly in atopic disorders such as eczema, inheritance of
to diagnose eczema, numerous lists of diagnostic crite­ certain polymorphisms from the mother is more likely to
ria have been developed in order to establish a defini­ be associated with allergic diseases in the child than
tion. At present, the UK diagnostic criteria have been inheritance from the father [29]. This parent‐of‐origin
the most widely validated and appear to be applicable effect is supported by epidemiological studies that indi­
and repeatable across all ages and many ethnicities [15]. cate a crucial role of environmental exposures during
However, the ideal set of diagnostic criteria still has to pregnancy [30] and a more close relationship of the
be established [16]. infant’s disease risk to maternal than paternal disease sta­
The confusing terminology for atopic diseases, with tus [31–33]. Genomic imprinting is one important epige­
terms such as atopic eczema, atopic dermatitis, childhood netic mechanism that might explain such parent‐of‐origin
eczema, atopiform dermatitis and flexural dermatitis fre­ effects. Imprinting is a phenomenon in which disease‐
quently used synonymously in the literature, reflects the predisposing alleles only show their effects in a particular
complexity of the diseases and the as yet insufficient epigenetic context, for example methylation induced by
knowledge on their pathophysiology. It has previously the parental origin of the variant, leading to mono‐allelic
been suggested that ‘eczema’ be defined as the disease for­ expression in the somatic cells of the offspring. Differential
merly called ‘atopic eczema’ or ‘atopic dermatitis’, expression can occur in all cells, or in specific tissues or
whereas the term ‘atopic eczema’ be reserved for those developmental stages [34].
patients with eczema and evidence for IgE involvement
[5,17]. However, as pointed out in a review, this division Methods used in the genetic dissection
might not adequately reflect the natural history of this dis­ of eczema
ease [1], and it has to be considered that so far most, if not For elucidation of the genetic background of complex dis­
all, studies on the genetics of eczema were performed eases, genetic association studies and linkage analyses
prior to these suggestions. Also, most existing DNA collec­ play an important role. The DNA sequence of human
tions have been assembled using older definitions. beings is on average 99.9% identical. The remaining 0.1%
It is anticipated that difficulties in defining eczema and of DNA sequence differences between any two individu­
its phenotypes may be overcome or at least improved by als are mainly based on common single nucleotide poly­
studying the genetics, and that the results of molecular morphisms (SNPs), in which a single nucleotide within
studies will be of great nosological significance, enabling the DNA sequence is replaced with an alternative one,
a classification of atopic disorders based on the underly­ and where each variation is present to some appreciable
ing genetic effects rather than on hypothetical concepts degree within a population (e.g. >1%). SNPs have become
and clinical symptoms as is the case today. A similar a favourite genetic tool in the investigation of complex
development has already taken place in other medical diseases, although many are nonfunctional and therefore
fields such as in neurodegenerative diseases, many of result in neutral phenotypic outcomes. However, func­
which are now defined by their mutated genes (e.g. spi­ tional SNPs can predispose individuals to disease, or
nocerebellar ataxias). influence its severity, progression or individual response
186 Section 3 Atopic Dermatitis and Related Disorders

to medicine. At the molecular level, these SNPs can affect inadequate assessment of the trait of interest and
the human phenotype by interfering on both levels of the i­nappropriate controls, variable study designs, inap­
protein synthesis machinery: noncoding SNPs may dis­ propriate statistical modelling and failure to correct
rupt transcription factor binding sites, splice sites and for multiple comparisons, genetic and environmental
other regulatory elements on the transcriptional level, heterogeneity, publication bias (more positive reports
whereas coding SNPs can cause an amino acid change are submitted to and accepted by journals) and lack of
and alter the functional or structural properties of the independent replication [26,41].
translated protein. Since the turn of the millennium, association studies
Whole genome linkage analysis aims at the identifica­ have been revolutionized through newly developed
tion of chromosomal regions associated with a disease high‐throughput SNP genotyping platforms and
by identifying genetic markers, e.g. microsatellites or knowledge gained from the HapMap project, which
SNPs, co‐segregating with the disease within families. showed that the human genome is organized into
Susceptibility regions identified in such hypothesis‐free blocks of haplotypes with limited diversity within each
ATOPIC DERMATITIS

screens typically encompass several megabases of DNA of these blocks [42]. Association studies aim at the
sequence and might contain hundreds of genes. Thus, detection of proxy variants, which are in linkage dise­
SECTION 3:

intensive follow‐up analyses are needed to narrow down quilibrium (LD) with the causal variant. LD refers to
the region of interest and to determine single disease the nonindependent inheritance of alleles, e.g. SNPs, at
genes. While this traditional approach has been extremely two or more loci, which extend over relatively short
successful in the identification of monogenic disease distances and may contain either a single gene or a few
genes [35], it has only limited power to detect risk genes genes only [38]. As a consequence of this genomic
for complex traits due to epistasis, incomplete penetrance, architecture a limited set of SNPs can capture the vast
polygeneity or phenotypic heterogeneity. Very few link­ majority of common variations and ‘tag’ the haplotype
age approaches have led to the identification of causal pattern sufficiently.
disease variants in this setting. For eczema several link­ Since 2005, commercial SNP‐typing arrays have been
age studies have identified various putative disease loci available and allow the simultaneous investigation of
in the past [36]. However, with the exception of the filag­ up to 4 million SNPs. The markers used are selected to
grin (FLG) gene, which partly explains the linkage signal provide maximal coverage of all common variations via
observed on chromosome 1q21 [37], it has not been pos­ LD‐based tagging, or even spacing within the genome
sible to assign a disease gene to any of the linkage regions [43]. These hypothesis‐free genome‐wide association
identified so far. studies (GWAS) were successfully used to identify com­
Association studies are thought to provide a more mon disease‐associated variants (defined as those present
powerful method for detecting complex disease alleles, in more than 5% of the population). Another useful source
in particular alleles that only confer a modest disease for genetic association studies was provided by the
risk. These studies statistically analyse the correlation 1000 Genomes project (http://www.internationalgenome.
between a genetic marker, usually an SNP, and the dis­ org), which between 2008 and 2015 sequenced the
ease in unrelated individuals (case‐control and case‐ genomes of more than 2500 individuals from 26 different
cohort studies) or their transmission in families populations, thereby providing a comprehensive data set
(family‐based design) [38]. An association between the of common as well as low‐frequency variants.
genotype and the phenotype is assumed if the genetic Technical improvements in recent years have also led
marker and the disease occur together more often than to a substantial decrease in costs of next‐generation
expected by chance. Due to technical limitations, sequencing technologies. In contrast to genetic associa­
genetic association studies have been limited to the tion studies that analyse a limited number of prese­
study of candidate genes, i.e. genes located in a region lected single nucleotide variants distributed over the
showing linkage in previous screening studies and/or genome, DNA sequencing provides information on
because of their function and expression patterns each single base constituting an individual human DNA
(positional and/or functional candidate genes) [39]. sequence. Whereas whole genome sequencing still is
Based on the assumption that the primary defect in rather costly, whole exome sequencing approaches,
eczema is immunological and prior to the discovery of which exclusively target the coding DNA regions of the
FLG (see Filaggrin), for many years candidate genes human genome, already allow the identification of as
involved in antigen presentation and cell‐mediated/ yet unknown rare functional variants within larger
humoral immune response, and cell signalling/cellular numbers of samples at reasonable costs. This approach
movement have been investigated, as well as genes has already been very successfully applied within fami­
that had been associated with asthma and/or atopy. lies affected by rare monogenic diseases, and will
More than 100 associated risk genes for eczema were undoubtedly increase our knowledge of genetic risk
reported up to 2009 by candidate gene association factors for common complex diseases like eczema.
studies [40], but most of these studies lack stringent Due to genetic studies the aetiological concept of
replication and have to be interpreted with caution. eczema underwent a profound change within recent
Potential reasons for irreproducibility of results years from a primarily immune‐mediated disorder to one
include low power (as a result of small sample sizes), that is equally, or may even be primarily, based on a skin
inappropriate selection of candidate loci and markers, barrier malfunction.
Chapter 14 Genetics and Aetiology of Atopic Dermatitis 187

Genes implicated in the aetiology highly specific for eczema [48,54–56]. Further, for most of
of eczema these loci the causative gene or gene product as well as
the underlying molecular mechanisms remain to be
Association studies, in particular those using genome‐ identified and characterized. Some notable exceptions are
wide or targeted high‐throughput approaches, have presented here.
robustly identified more than 30 susceptibility regions for
eczema (Table 14.1) [44–52]. These loci together explain
approximately 20% of the estimated heritability. Most of Filaggrin
them harbour candidate genes that contribute to immune Filaggrin (filament aggregating protein; FLG) was first
mechanisms, in particular to innate immune signalling described in 1977 as an insoluble protein purified from rat
and T‐cell activation and specification (Table 14.1, epidermis that interacted with intermediate filaments in
Fig. 14.1), and appear to impact the susceptibility for a vitro [57]. It represents a structural protein with key func­
range of immune‐mediated diseases rather than being tions in the formation and maintenance of the cornified

ATOPIC DERMATITIS
SECTION 3:
Table 14.1 Susceptibility loci and most plausible candidate genes identified through genome‐wide association studies for atopic eczema

Associated Candidate gene Known/proposed function


locus

1q21.2 CIART Circadian transcriptional repressor


1q21.3 FLG Terminal epidermal differentiation; aggregation of keratin intermediate filaments
1q21.3 IL6R Subunit of the IL‐6 receptor; differentiation of multiple immune cells, especially B cells
2p13.3 CD207 C‐type lectin with mannose binding specificity; major receptor on Langerhans cells; antigen
uptake, processing and presentation
2p16.1 PUS10 Posttranscriptional modification of structural RNAs; involved in apoptosis
2p25.1 LINC00299 Long noncoding RNA; no known function
2q12.1 IL1RL1 Subunit of the IL‐33 receptor; Th cell function
IL18R1/IL18RAP Subunits of the IL‐18 receptor; regulation of inflammatory response via NF‐κB
2q24.3 XIRP2 Actin cytoskeleton and cell–cell junction organization; protection of actin filaments from
depolymerization
3p21.1 RFT1 Oligosaccharide transporter; protein N‐glycosylation
3p22.3 CCR4 C‐C chemokine receptor; leucocyte trafficking
3q13.2 CCDC80 Cell adhesion and matrix assembly
4q27 IL2/IL21 Differentiation, proliferation and activation of multiple immune cells (T cells, B cells,
macrophages, natural killer cells)
5p13.2 IL7R Subunit of the IL‐7 and TSLP receptor; proliferation of lymphoid progenitors; release of T‐cell‐
attracting chemokines; promotion of Th2‐cell response
5q22.1 TSLP Release of T‐cell‐attracting chemokines; promotion of Th2‐cell response; AMP activity in the oral
cavity and on the skin
5q31.1 IL13/IL4 Mainly produced by activated Th2 cells; B‐cell proliferation; IgE isotype switching
6p21.32 HLA‐DRB Antigen processing and presentation MHC class II
6p21.33 MICB Stress‐induced self‐antigen; activation of cytolytic response of NK cells, αβ T cells, and γδ T cells
7p22.2 CARD11 TCR‐mediated T cell activation; NF‐κB activation
8q21.13 ZBTB10 Transcription regulation
9p21.3 DMRTA1 Transcription regulation
10p15.1 IL15RA/IL2RA IL‐15 and IL‐2 receptor subunits; proliferation and stimulation of different lymphocytes
10q21.2 ZNF365 Mitotic cytokinesis; maintenance of genome stability
11p13‐12 PRR5L Regulation of protein kinase C phosphorylation; survival and organization of the cytoskeleton;
cell migration
11p15.4 NLRP10 Regulation of the innate immune system; pro‐inflammatory cytokine release; inhibition of
apoptosis; anti‐inflammatory activity
11q13.1 OVOL1 Transcription factor; hair formation and spermatogenesis
11q13.5 LRRC32 Regulation of Treg function and TGF‐β activation
11q24.3 ETS1 Transcription factor; direct control of cytokine and chemokine expression
14q13.2 PPP2R3C Regulation of protein phosphatases; activation‐induced cell death of B cells
16p13.13 CLEC16A C‐type lectin; regulation of mitophagy/autophagy and mitochondrial health
17q21.2 STAT3 Signal transducer and transcription activator; mediation of cellular responses to interleukins
17q21.32‐33 ZNF652 Transcription regulation
19p13.2 ACTL9 Cytoskeletal functions
ADAMTS10 Metalloprotease; assembly of extracellular matrix components
20q13.2 CYP24A1 Vitamin D metabolism
20q13.33 TNFRSF6B TNF receptor superfamily; protection against apoptosis

Sorted by chromosome; AMP, antimicrobial peptide; IL, interleukin; MHC, major histocompatibility complex; NF‐κB, nuclear factor ‘kappa‐light‐chain‐enhancer’
of activated B cells; NK cells, natural killer cells; RNA, ribonucleic acid; TCR, T‐cell receptor; TGF‐β, transforming growth factor β; Th cell, T‐helper cell; TNF,
tumour necrosis factor; TSLP, thymic stromal lymphopoietin.
188 Section 3 Atopic Dermatitis and Related Disorders

Epidermal barrier Environmental sensing


FLG, OVOL1, CARD11,
ACTL9/ADAMTS10 HLA-DRB, MICB
ATOPIC DERMATITIS
SECTION 3:

Tissue response
Immune regulation CCDC80, CYP24A1,
IL1RL1/IL18R1/IL18RAP LRRC32, NLRP10, PRR5L,
IL2/IL21, IL4/IL13, TNFRSF6B,
IL6R, CLEC16A ZNF365, ZNF652
Fig. 14.1 Functional overview for selected candidate
genes identified through genome‐wide association
studies. Source: Adapted from Weidinger and Novak
(2016) [53].

envelope within the outermost layer of the human reduced in the epidermis of individuals with this disease
epidermis, and is essential for the prevention of penetra­ [63,64]. However, in‐depth analysis of the protein‐encoding
tion of environmental agents such as microbes or allergens gene transpired to be technically difficult because of its
into the organism and also, on the other hand, for controlling repetitive structural nature, and the sequence of the entire
transepidermal water loss (TEWL) [58,59]. FLG gene was finally published in 2006, identifying two
Profilaggrin is expressed as a large inactive precursor loss‐of‐function variants (p.R501X and c.2282del4) to be
protein of 400 kDa in the stratum granulosum of the epi­ the cause of IV [65]. Subsequently the same group
dermis, where it represents the main constituent of the detected significant associations between these variants
keratohyalin F granules (Fig. 14.2). It is composed of and eczema [66].
10–12 tandem repeats, each encoding a functionally active Since that time more than 50 recurrent and family‐
FLG monomer. Upon cornification of keratinocytes, pro­ specific variants have been identified within the FLG
filaggrin is dephosphorylated and proteolytically cleaved gene in European and Asian populations [67]. These are
into its active subunits. These monomers lead to a dense present in up to 10% of the general European population,
bundling of keratin intermediate filaments, leading to and up to 40% of eczema patients are carriers of at least
collapse of the keratinocyte’s cytoskeleton during their one of the five most recurrent null variants (p.R501X,
differentiation into corneocytes, and finally to desquama­ c.2282del4, p.S3247X, p.R2447X and c.3702delG), which
tion of the stratum corneum. Concurrently, the amino account for about 95% of all known variants within FLG
(N)‐terminus of the protein is translocated into the cell [67]. Interestingly, frequency appears to increase from
nucleus, where it is believed to fulfill an additional but as southern to northern Europe, which might reflect eth­
yet unknown role in regulation of terminal differentia­ nic differences. In populations from UK and Ireland,
tion. Moreover, degradation products of FLG, free hygro­ FLG loss‐of‐function variants seem to be more prevalent
scopic amino acids and their derivatives, constitute a compared to continental Europe [65,68–72]. Of the more
large part of the pool of so‐called natural moisturizing than 20 described loss‐of‐function variants found in
factors (NMFs) within the skin [61]. NMFs play an impor­ the European population, the majority could not be found
tant role in regulation of skin hydration and skin pH, the in Asian individuals [65,73,74] and vice versa. Similarly,
latter of which is closely linked to protease activity and no FLG loss‐of‐function variants predominate in
antimicrobial defence [62]. The importance of FLG‐ African‐American children; however, uncommon
derived breakdown products for skin barrier function is FLG loss‐of‐function variants in African‐American chil­
supported by the remarkably short half‐life of FLG, dren have been identified and shown to be associated
which exists only for 6 hours before its full proteolysis with more persistent AD [75]. Each ethnic group seems to
into NMFs. have its own exclusive null variants showing the same
FLG was suspected of involvement in keratinization strong impact on the development of eczema. All reported
disorders such as ichthyosis vulgaris (IV) more than variants are frameshift or nonsense variants leading to a
25 years ago, and its expression was found to be clearly premature translation stop and hence truncation of the
Chapter 14 Genetics and Aetiology of Atopic Dermatitis 189

Putative filaggrin functions

Outer stratum corneum


Hydration through hygroscopic
amino acids UCA and PCA
Inner stratum corneum Possible contribution to acid mantle
through acid degradation products
Lipid bilayer
Filament binding, barrier integrity
Granular layer Profilaggrin in granular layer:
Pro-protein, nonfunctional

(a) (b)

ATOPIC DERMATITIS
SECTION 3:
PCA
H2O

PADs 1+3 Caspase 14


+PCA
TGMs Profilaggrin A domain
“NMF”
Profilaggrin B domain
Ca2+
Matriptase Profilaggrin tail domain
Dephosphorylation LEKTI
CAP1/Prss Filaggrin

Keratohyalin F granule
Nucleus
Free amino acid

(c)
Fig. 14.2 Filaggrin expression and putative functions in the skin barrier. (a) The precursor pro‐protein profilaggrin is strongly expressed within keratohyalin
granules, tightly limited to and accounting for the typical appearance of the granular layer. The stratum corneum stains strongly positive for filaggrin. (b)
Filaggrin has several proposed site‐specific functions under the influence of the epidermal terminal differentiation programme through the outer granular
layer (cleavage of profilaggrin to filaggrin), lipid bilayer of the inner stratum corneum (filament compaction, contribution to barrier integrity) and during
desquamation of the outer stratum corneum (production of amino acid degradation products that contribute to the hydration of these outer layers and
probably contribute to the ‘acid mantle’). (c) Current knowledge of molecular control of filaggrin homeostasis. Profilaggrin is dephosphorylated in
conditions of increasing calcium concentration and then proteolytically cleaved by the proteases matriptase (inhibited by the protease inhibitor LETKI) and
CAP1/Prss. Post proteolysis, the filaggrin tail domain locates to the nucleus as part of the terminal differentiation process. Free filaggrin protein is cross‐
linked to keratin filaments by transglutaminases (TGMs) and subsequently deiminated by peptidylarginine deiminases (PADs) 1 and 3. Further posttransla-
tional modification is undertaken by caspase 14 to produce the free amino acid hygroscopic degradation products urocanic acid (UCA) and pyrrolidone
carboxylic acid (PCA), collectively known as natural moisturizing factor (NMF), which contributes to stratum corneum hydration. Source: O’Regan et al.
2008 [60]. Reproduced with permission of Elsevier.

profilaggrin molecule. The two most common variants, Th2 cytokine cluster
p.R501X (a nonsense variant of the arginine codon 501 to One of the most consistent associations with eczema and
a stop codon) and c.2282del4 (a frameshift variant at posi­ a variety of other immune‐related disorders such as
tion 2282 due to a four base pair [bp] deletion), in the first Crohn’s disease, asthma and psoriasis has been observed
repeat of the gene impede any FLG synthesis from these for the T‐helper 2 (Th2) cytokine cluster on chromosome
alleles. As the 3′ gene sequence seems to be important for 5q31 [77,78], a locus that harbours the genes encoding
posttranslational processing into functional FLG subu­ interleukin (IL)‐3, IL‐4, IL‐5 and IL‐13. These cytokines
nits, even variants located in this region prevent or reduce are mainly produced by differentiated CD4+ Th2 cells
production of free FLG in the stratum corneum [62,69]. that are responsible for the antibody‐mediated immune
Scientific evidence for this gene as one of the strongest response against parasites, allergens and bacteria. Th2
risk factors for eczema is compelling due to numerous cytokines show a great functional overlap and are
replication studies and meta‐analyses, which revealed an involved in Th2 cell development and polarization, B‐cell
overall odds ratio of >3.0 for eczema [76]. Due to the dis­ proliferation and differentiation, IgE antibody produc­
covery of FLG, susceptibility genes for eczema are now tion, and activation and recruitment of certain immune
divided into two major functional groups: genes contrib­ cell subsets such as mast cells and eosinophils to inflam­
uting to epidermal or epithelial structures, and genes matory sites. Different potential risk SNPs have been
encoding immune regulatory proteins. identified throughout the whole region, and functionally
190 Section 3 Atopic Dermatitis and Related Disorders

disruptive variants within IL4 and IL13 genes are con­ by targeted next‐generation sequencing revealed a
vincingly associated with total IgE levels and atopic dis­ number of low‐frequency variants within the coding
eases including eczema (reviewed in [79]). Additionally, region of LRRC32 to be strongly associated with eczema.
various GWAS identified associated SNPs within RAD50, LRRC32 encodes the protein glycoprotein A repetitions
a gene situated within the cytokine cluster between predominant (GARP), a cell surface receptor mainly
IL4/IL13 and IL5, encoding a DNA repair protein that is expressed on activated regulatory T cells (Tregs), which
ubiquitously expressed and therefore lacks any direct bio­ binds and activates latent TGF‐β, thereby regulating the
logical connection to eczema. However, in mouse models bioavailability of this multifunctional cytokine [87]. All of
RAD50 has been shown to contain an evolutionarily con­ the identified variants lead to an amino acid exchange
served locus control region (LCR), which is supposed to within the primary structure of the protein, and structural
tightly control expression of the neighbouring Th2 modelling of GARP predicted a defective posttransla­
cytokines [80]. Hence, associated polymorphisms in tional modification due to the identified variants, finally
RAD50 influence regulation of Th2 cytokine gene tran­ leading to incorrect folding and constrained transporta­
ATOPIC DERMATITIS

scription rather than RAD50 gene function itself. tion of GARP to the outer cell membrane. Overexpression
Regulation through the LCR within RAD50 occurs via a experiments of mutated GARP indeed showed reduction
SECTION 3:

complex recruitment of transcription factors and other of the membrane‐bound protein, and a reduced conver­
regulatory molecules and includes epigenetic remodelling sion rate of CD4+/CD25− T cells into Tregs was observed
of the three‐dimensional structure of chromatin within when obtained from individuals carrying the identified
this region. An intrachromosomal loop allows direct variants [86]. Tregs are of high importance for suppres­
interaction of the transcription factor machinery bound to sion of effector T cells and other leucocytes and for con­
the LCR with the Th2 cytokine gene promoter regions in trol of inflammatory immune responses and autoimmunity
order to activate or repress their expression [81,82]. The [88], and experimental deactivation of GARP has been
LCR has also been shown to be rapidly demethylated in shown to lead to a reduction in the suppressor activity of
murine cell lines of stimulated Th2 cells [83], linking the these cells [89]. It is possible to speculate that the missing
epigenetic mechanism of DNA methylation with activa­ membrane expression of GARP on activated Tregs leads
tion of Th2 cytokine expression. Inactivation of the LCR to a decreased activation rate of TGF‐β and thereby affects
in mice leads to a reduction of Th2 cytokine and IgE levels downstream function of this important cytokine, finally
[84], and variants within the LCR impact its transcrip­ resulting in an overshooting inflammatory immune reac­
tional activity [85]. tion. Moreover, Tregs are suspected to be involved in a
Together, genetic as well as epigenetic elements seem to variety of other diseases, and association of the previ­
regulate expression of this important immune gene clus­ ously mentioned eczema marker has also been detected
ter, and variants might interfere with both mechanisms, in a GWAS on Crohn’s disease, implying that the 11q13.5
thereby promoting Th2‐dominated immune reactions locus and GARP are also important within the disease
characteristically for atopy‐related disorders like eczema. pathophysiology of further (auto)immune diseases.
Given the number of immune‐mediated diseases that
have been linked to the Th2 cytokine cluster, it seems very
Conclusions and future directions
likely that variants within this region are more relevant
for the atopic state sensu lato than for any individual Despite much progress over the past two decades, our
distinct atopic disease. understanding of the complex genetic susceptibility to
Identification of further causal variants that probably eczema is still incomplete. In particular, there is a lack of
exist within this important risk locus is hampered by the follow‐up studies in order to determine affected genes and
complex marker–marker correlation in this genomic downstream mechanisms. Thus far only the 1q21.3, 5q31
region and the gene–gene interactions through overlap­ and 11q13.5 loci have been functionally explored. The
ping functional pathways. important role of the skin barrier within eczema has been
clearly evidenced with the identification and validation of
GARP (glycoprotein A repetitions FLG variants as the strongest known genetic risk factors for
predominant)/LRRC32 (leucine‐rich the disease. This paradigm change has now also led to early
repeat‐containing protein 32) intervention studies of primary prevention of the disease by
Another consistently‐replicated eczema risk locus has improving the skin barrier function with daily emollient
been identified on chromosome 11q13.5, with the strong­ applications. The results of these randomized intervention
est association observed for an SNP located in an inter­ studies have been an approximate 50% reduction of eczema
genic region between the two annotated genes chromosome incidence in high‐risk newborns [90, 91], lending further
11 open reading frame 30 (C11orf30) and leucine rich repeat support for the key importance of a functionally intact skin
containing 32 (LRRC32). Assignment of the signal to one of barrier. Likewise, genetic findings that imply an important
its neighbouring genes could not be accomplished, as role of variation in genes affecting (auto‐)immune regula­
both are attractive candidates due to their known func­ tion, in particular innate signalling and T‐cell activation and
tions in epithelial immunity and differentiation and acti­ specification, have contributed to the development of drugs
vation of regulatory T cells, respectively, and the causative modifying immune pathways, some of which are already in
risk gene marked by the associated SNP has more recently advanced stages of development and showed encouraging
been identified [86]. Genetic fine mapping of the region results in clinical trials [92–94].
Chapter 14 Genetics and Aetiology of Atopic Dermatitis 191

In order to deepen our understanding of the genetic 19 Sneddon IB. The management of infantile eczema. Med Press 1951;
226:329–33.
architecture of eczema, there is a need for further large‐
20 Schaffer N. Atopic dermatitis in the older child. J Asthma Res
scale and detailed whole‐genome studies, follow‐up 1966;3:189–91.
studies for fine‐mapping and functional investigations, 21 Larsen FS, Holm NV, Henningsen K. Atopic dermatitis. A genetic‐
and holistic approaches that integrate phenotypic varia­ epidemiologic study in a population‐based twin sample. J Am Acad
Dermatol 1986;15:487–94.
bles and information from different molecular levels. In 22 Schultz Larsen F. Atopic dermatitis: a genetic‐epidemiologic study in
addition, it will be necessary to revisit traditional pheno­ a population‐based twin sample. J Am Acad Dermatol 1993;
type definitions based on familial components, heritable 28:719–23.
endophenotypes and genetic markers. To better define 23 Thomsen SF, Ulrik CS, Kyvik KO et al. Importance of genetic factors
in the etiology of atopic dermatitis: a twin study. Allergy Asthma Proc
pathophysiological mechanisms and to identify novel 2007;28:535–9.
therapeutic targets and biomarkers that predict therapeu­ 24 van Beijsterveldt CE, Boomsma DI. Genetics of parentally reported
tic response, there has been increasing interest in develop­ asthma, eczema and rhinitis in 5‐yr‐old twins. Eur Respir J 2007;
29:516–21.
ing computational analysis and predictive models of AD

ATOPIC DERMATITIS
25 Phillips PC. Epistasis—the essential role of gene interactions in the
[95]. The final frontier will certainly be to translate genetic structure and evolution of genetic systems. Nat Rev Genet
findings into an improved classification and the develop­ 2008;9:855–67.

SECTION 3:
ment of more targeted interventions. 26 Glazier AM, Nadeau JH, Aitman TJ. Finding genes that underlie com­
plex traits. Science 2002;298:2345–9.
27 Williams H, Robertson C, Stewart A et al. Worldwide variations in the
References prevalence of symptoms of atopic eczema in the International Study
1 Bieber T. Atopic dermatitis. N Engl J Med 2008;358:1483–94. of Asthma and Allergies in Childhood. J Allergy Clin Immunol
2 Wuthrich B. The natural history of atopic dermatitis. In: Ring J, 1999;103:125–38.
Przybilla B, Ruzicka T (eds) Handbook of Atopic Dermatitis, 2nd edn. 28 Asher MI, Montefort S, Bjorksten B et al. Worldwide time trends in the
Berlin: Springer Verlag, 2006:150–6. prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and
3 Novak N, Bieber T. Allergic and nonallergic forms of atopic diseases. eczema in childhood: ISAAC Phases One and Three repeat multi­
J Allergy Clin Immunol 2003;112:252–62. country cross‐sectional surveys. Lancet 2006;368:733–43.
4 Wuthrich B, Schmid‐Grendelmeier P. The atopic eczema/dermatitis 29 Moffatt MF, Cookson WO. The genetics of asthma. Maternal effects in
syndrome. Epidemiology, natural course, and immunology of the atopic disease. Clin Exp Allergy 1998;28(suppl 1):56–61.
IgE‐associated (“extrinsic”) and the nonallergic ("intrinsic") AEDS. 30 Warner JA, Jones CA, Jones AC, Warner JO. Prenatal origins of allergic
J Investig Allergol Clin Immunol 2003;13:1–5. disease. J Allergy Clin Immunol 2000;105:S493–8.
5 Johansson SG, Bieber T, Dahl R et al. Revised nomenclature for allergy 31 Dold S, Wjst M, von Mutius E et al. Genetic risk for asthma, allergic
for global use: Report of the Nomenclature Review Committee of the rhinitis, and atopic dermatitis. Arch Dis Child 1992;67:1018–22.
World Allergy Organization, October 2003. J Allergy Clin Immunol 32 Ruiz RG, Kemeny DM, Price JF. Higher risk of infantile atopic
2004;113:832–6. dermatitis from maternal atopy than from paternal atopy. Clin Exp
6 Bergmann RL, Edenharter G, Bergmann KE et al. Atopic dermatitis in Allergy 1992;22:762–6.
early infancy predicts allergic airway disease at 5 years. Clin Exp 33 Diepgen TL, Blettner M. Analysis of familial aggregation of atopic
Allergy 1998;28:965–70. eczema and other atopic diseases by ODDS RATIO regression models.
7 Gustafsson D, Sjoberg O, Foucard T. Development of allergies and J Invest Dermatol 1996;106:977–81.
asthma in infants and young children with atopic dermatitis—a pro­ 34 Reik W, Walter J. Genomic imprinting: parental influence on the
spective follow‐up to 7 years of age. Allergy 2000;55:240–5. genome. Nat Rev Genet 2001;2:21–32.
8 Hakansson K, Thomsen SF, Ulrik CS et al. Increase in the prevalence 35 McKusick VA. Mendelian Inheritance in Man and its online version,
of rhinitis among Danish children from 1986 to 2001. Pediatr Allergy OMIM. Am J Hum Genet 2007;80:588–604.
Immunol 2007;18:154–9. 36 Lee YA, Wahn U, Kehrt R et al. A major susceptibility locus for atopic
9 Spergel JM. From atopic dermatitis to asthma: the atopic march. Ann dermatitis maps to chromosome 3q21. Nat Genet 2000;26:470–3.
Allergy Asthma Immunol 2010;105:99–106. 37 Morar N, Edster P, Street T et al. Fine mapping of susceptibility genes
10 Illi S, von Mutius E, Lau S et al. The natural course of atopic dermatitis for atopic dermatitis in the epidermal differentiation complex on
from birth to age 7 years and the association with asthma. J Allergy chromosome 1q21. Br J Dermatol 2007;157:3.
Clin Immunol 2004;113:925–31. 38 Altshuler D, Daly MJ, Lander ES. Genetic mapping in human disease.
11 Williams H, Flohr C. How epidemiology has challenged 3 prevailing Science 2008;322:881–8.
concepts about atopic dermatitis. J Allergy Clin Immunol 2006; 39 Tabor HK, Risch NJ, Myers RM. Candidate‐gene approaches for stud­
118:209–13. ying complex genetic traits: practical considerations. Nat Rev Genet
12 Flohr C, Weiland SK, Weinmayr G et al. The role of atopic sensitiza­ 2002;3:391–7.
tion in flexural eczema: findings from the International Study of 40 Barnes KC. An update on the genetics of atopic dermatitis: scratching
Asthma and Allergies in Childhood Phase Two. J Allergy Clin the surface in 2009. J Allergy Clin Immunol 2010;125:16–29.e1–11.
Immunol 2008;121:141–7.e4. 41 Lohmueller KE, Pearce CL, Pike M et al. Meta‐analysis of genetic
13 He R, Oyoshi MK, Garibyan L et al. TSLP acts on infiltrating effector association studies supports a contribution of common variants to
T cells to drive allergic skin inflammation. Proc Natl Acad Sci U S A susceptibility to common disease. Nat Genet 2003;33:177–82.
2008;105:11875–80. 42 The International HapMap Consortium. The International HapMap
14 Demehri S, Morimoto M, Holtzman MJ, Kopan R. Skin‐derived TSLP Project. Nature 2003;426:789–96.
triggers progression from epidermal‐barrier defects to asthma. PLoS 43 Grant SF, Hakonarson H. Microarray technology and applications in
Biol 2009;7:e1000067. the arena of genome‐wide association. Clin Chem 2008;54:1116–24.
15 Williams HC, Burney PG, Hay RJ et al. The U.K. Working Party’s 44 Esparza‐Gordillo J, Weidinger S, Folster‐Holst R et al. A common
Diagnostic Criteria for Atopic Dermatitis. I. Derivation of a minimum variant on chromosome 11q13 is associated with atopic dermatitis.
set of discriminators for atopic dermatitis. Br J Dermatol 1994;131: Nat Genet 2009;41:596–601.
383–96. 45 Sun LD, Xiao FL, Li Y et al. Genome‐wide association study identifies
16 Brenninkmeijer EE, Schram ME, Leeflang MM et al. Diagnostic crite­ two new susceptibility loci for atopic dermatitis in the Chinese Han
ria for atopic dermatitis: a systematic review. Br J Dermatol 2008; population. Nat Genet 2011;43:690–4.
158:754–65. 46 Hirota T, Takahashi A, Kubo M et al. Genome‐wide association study
17 Johansson SG, Hourihane JO, Bousquet J et al. A revised nomencla­ identifies eight new susceptibility loci for atopic dermatitis in the
ture for allergy. An EAACI position statement from the EAACI Japanese population. Nat Genet 2012;44:1222–6.
nomenclature task force. Allergy 2001;56:813–24. 47 Paternoster L, Standl M, Chen CM et al. Meta‐analysis of genome‐
18 Cooke R, van der Veer A. Human sensitization. J Immunol wide association studies identifies three new risk loci for atopic
1916;1:201–5. dermatitis. Nat Genet 2012;44:187–92.
192 Section 3 Atopic Dermatitis and Related Disorders

48 Ellinghaus D, Baurecht H, Esparza‐Gordillo J et al. High‐density 73 Nomura T, Sandilands A, Akiyama M et al. Unique mutations in the
genotyping study identifies four new susceptibility loci for atopic filaggrin gene in Japanese patients with ichthyosis vulgaris and atopic
dermatitis. Nat Genet 2013;45:808–12. dermatitis. J Invest Dermatol 2007;119:434–40.
49 Esparza‐Gordillo J, Schaarschmidt H, Liang L et al. A functional IL‐6 74 Nomura T, Akiyama M, Sandilands A et al. Prevalent and rare muta­
receptor (IL6R) variant is a risk factor for persistent atopic dermatitis. tions in the gene encoding filaggrin in Japanese patients with ichthyo­
J Allergy Clin Immunol 2013;132:371–7. sis vulgaris and atopic dermatitis. J Invest Dermatol 2009;129:1302–5.
50 Weidinger S, Willis‐Owen SA, Kamatani Y et al. A genome‐wide 75 Margolis DJ, Mitra N, Gochnauer H et al. Uncommon filaggrin vari­
association study of atopic dermatitis identifies loci with overlapping ants are associated with persistent atopic dermatitis in African
effects on asthma and psoriasis. Hum Mol Genet 2013;22:4841–56. Americans. J Invest Dermatol 2018;138:1501–6.
51 Paternoster L, Standl M, Waage J et al. Multi‐ancestry genome‐wide 76 Rodriguez E, Baurecht H, Herberich E et al. Meta‐analysis of filag-
association study of 21,000 cases and 95,000 controls identifies new grin polymorphisms in eczema and asthma: robust risk factors in
risk loci for atopic dermatitis. Nat Genet 2015;47:1449–56. atopic disease. J Allergy Clin Immunol 2009;123:1361–70.e7.
52 Schaarschmidt H, Ellinghaus D, Rodriguez E et al. A genome‐wide 77 Rioux JD, Daly MJ, Silverberg MS et al. Genetic variation in the 5q31
association study reveals 2 new susceptibility loci for atopic dermati­ cytokine gene cluster confers susceptibility to Crohn disease. Nat
tis. J Allergy Clin Immunol 2015;136:802–6. Genet 2001;29:223–8.
53 Weidinger S, Novak N. Atopic dermatitis. Lancet 2016;387:1109–22. 78 Li Y, Chang M, Schrodi SJ et al. The 5q31 variants associated with
ATOPIC DERMATITIS

54 Ellinghaus D, Jostins L, Spain SL et al. Analysis of five chronic inflam­ psoriasis and Crohn’s disease are distinct. Hum Mol Genet
matory diseases identifies 27 new associations and highlights disease‐ 2008;17:2978–85.
specific patterns at shared loci. Nat Genet 2016;48:510–18. 79 Vercelli D. Discovering susceptibility genes for asthma and
SECTION 3:

55 Ferreira MAR, Vonk JM, Baurecht H et al. Eleven loci with new repro­ allergy. Nat Rev Immunol 2008;8:169–82.
ducible genetic associations with allergic disease risk. J Allergy Clin 80 Lee GR, Fields PE, Griffin TJ, Flavell RA. Regulation of the Th2
Immunol 2019;143(2):691–9. cytokine locus by a locus control region. Immunity 2003;19:
56 Devos M, Mogilenko DA, Fleury S et al. Keratinocyte expression of 145–53.
A20/TNFAIP3 controls skin inflammation associated with atopic der­ 81 Spilianakis CG, Flavell RA. Long‐range intrachromosomal interac­
matitis and psoriasis. J Invest Dermatol 2019;139:135–45. tions in the T helper type 2 cytokine locus. Nat Immunol 2004;
57 Dale BA. Purification and characterization of a basic protein from the 5:1017–27.
stratum corneum of mammalian epidermis. Biochim Biophys Acta 82 Lee GR, Spilianakis CG, Flavell RA. Hypersensitive site 7 of the TH2
1977;491:193–204. locus control region is essential for expressing TH2 cytokine genes
58 Candi E, Schmidt R, Melino G. The cornified envelope: a model of cell and for long‐range intrachromosomal interactions. Nat Immunol
death in the skin. Nat Rev Mol Cell Biol 2005;6:328–40. 2005;6:42–8.
59 Proksch E, Brandner JM, Jensen JM. The skin: an indispensable bar­ 83 Kim ST, Fields PE, Flavell RA. Demethylation of a specific hypersensi­
rier. Exp Dermatol 2008;17:1063–72. tive site in the Th2 locus control region. Proc Natl Acad Sci U S A
60 O’Regan GM, Sandilands A, McLean WH, Irvine AD. Filaggrin in 2007;104:17052–7.
atopic dermatitis. J Allergy Clin Immunol 2008;122:689–93. 84 Koh BH, Hwang SS, Kim JY et al. Th2 LCR is essential for regulation
61 Rawlings AV, Harding CR. Moisturization and skin barrier function. of Th2 cytokine genes and for pathogenesis of allergic asthma. Proc
Dermatol Ther 2004;17(suppl 1):43–8. Natl Acad Sci U S A 2010;107:10614–19.
62 Sandilands A, Sutherland C, Irvine AD, McLean WH. Filaggrin in the 85 Kretschmer A, Moller G, Lee H et al. A common atopy‐associated
frontline: role in skin barrier function and disease. J Cell Sci variant in the Th2 cytokine locus control region impacts transcrip­
2009;122:1285–94. tional regulation and alters SMAD3 and SP1 binding. Allergy 2014;
63 Fleckman P, Holbrook KA, Dale BA, Sybert VP. Keratinocytes cul­ 69:632–42.
tured from subjects with ichthyosis vulgaris are phenotypically 86 Manz J, Rodriguez E, ElSharawy A et al. Targeted resequencing and
abnormal. J Invest Dermatol 1987;88:640–5. functional testing identifies low‐frequency missense variants in
64 Sybert VP, Dale BA, Holbrook KA. Ichthyosis vulgaris: identification the gene encoding GARP as significant contributors to atopic der-
of a defect in synthesis of filaggrin correlated with an absence of kera­ matitis risk. J Invest Dermatol 2016;136:2380–6.
tohyaline granules. J Invest Dermatol 1985;84:191–4. 87 Wang R, Zhu J, Dong X et al. GARP regulates the bioavailability and
65 Smith FJ, Irvine AD, Terron‐Kwiatkowski A et al. Loss‐of‐function activation of TGFbeta. Mol Biol Cell 2012;23:1129–39.
mutations in the gene encoding filaggrin cause ichthyosis vulgaris. 88 Zhang H, Kong H, Zeng X et al. Subsets of regulatory T cells and their
Nat Genet 2006;38:337–42. roles in allergy. J Transl Med 2014;12:125.
66 Palmer CN, Irvine AD, Terron‐Kwiatkowski A et al. Common loss‐ 89 Fridrich S, Hahn SA, Linzmaier M et al. How soluble GARP enhances
of‐function variants of the epidermal barrier protein filaggrin are a major TGFbeta activation. PLoS One 2016;11:e0153290.
predisposing factor for atopic dermatitis. Nat Genet 2006;38:441–6. 90 Horimukai K, Morita K, Narita M et al. Application of moisturizer to
67 Irvine AD, McLean WH, Leung DY. Filaggrin mutations associated neonates prevents development of atopic dermatitis. J Allergy Clin
with skin and allergic diseases. N Engl J Med 2011;365:1315–27. Immunol 2014;134:824–30.e6.
68 Barker JN, Palmer CN, Zhao Y et al. Null mutations in the filaggrin 91 Simpson EL, Chalmers JR, Hanifin JM et al. Emollient enhancement of
gene (FLG) determine major susceptibility to early‐onset atopic the skin barrier from birth offers effective atopic dermatitis preven­
dermatitis that persists into adulthood. J Invest Dermatol 2007; tion. J Allergy Clin Immunol 2014;134:818–23.
127:564–7. 92 Beck LA, Thaci D, Hamilton JD et al. Dupilumab treatment in adults
69 Sandilands A, Terron‐Kwiatkowski A, Hull PR et al. Com­ with moderate‐to‐severe atopic dermatitis. N Engl J Med 2014;
prehensive analysis of the gene encoding filaggrin uncovers prevalent 371:130–9.
and rare mutations in ichthyosis vulgaris and atopic eczema. Nat Genet 93 Simpson EL, Bieber T, Guttman‐Yassky E et al. Two Phase 3 trials of
2007;39:650–4. dupilumab versus placebo in atopic dermatitis. N Engl J Med
70 Brown SJ, Relton CL, Liao H et al. Filaggrin null mutations and child­ 2016;375:2335–48.
hood atopic eczema: a population‐based case‐control study. J Allergy 94 Thaci D, Simpson EL, Beck LA et al. Efficacy and safety of dupilumab
Clin Immunol 2008;121:940–46.e3. in adults with moderate‐to‐severe atopic dermatitis inadequately
71 Rice NE, Patel BD, Lang IA et al. Filaggrin gene mutations are associ­ controlled by topical treatments: a randomised, placebo‐controlled,
ated with asthma and eczema in later life. J Allergy Clin Immunol dose‐ranging phase 2b trial. Lancet 2016;387:40–52.
2008;122:834–6. 95 Eyerich K, Brown SJ, Perez White BE et al. Human and computational
72 Weidinger S, O’Sullivan M, Illig T et al. Filaggrin mutations, atopic models of atopic dermatitis: a review and perspectives by an expert
eczema, hay fever, and asthma in children. J Allergy Clin Immunol panel of the International Eczema Council. J Allergy Clin Immunol
2008;121:1203–9. 2019;143(1):36–45.

You might also like