You are on page 1of 16

Journal of Dermatological Science 102 (2021) 142–157

Contents lists available at ScienceDirect

Journal of Dermatological Science


journal homepage: www.jdsjournal.com

Invited Review Article

Atopic dermatitis: Role of the skin barrier, environment, microbiome,


and therapeutic agents
Thomas Lugera,* , Masayuki Amagaib,c , Brigitte Drenod, Marie-Ange Dagnelied ,
Wilson Liaoe , Kenji Kabashimaf , Tamara Schikowskig , Ehrhardt Prokschh , Peter M. Eliasi,
Michel Simonj, Eric Simpsonk , Erin Grinichk , Matthias Schmuthl
a
Department of Dermatology, University of Münster, Münster, Germany
b
Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
c
Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
d
Dermatology Department, Nantes University, CHU Nantes, CIC 1413, CRCINA, Nantes, France
e
Department of Dermatology, University of California, San Francisco, CA, United States
f
Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
g
IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
h
Department of Dermatology, University of Kiel, Kiel, Germany
i
San Francisco VA Medical Center, University of California, San Francisco, CA, United States
j
UDEAR, Inserm, University of Toulouse, U1056, Toulouse, France
k
Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
l
Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria

A R T I C L E I N F O A B S T R A C T

Article history: Atopic dermatitis (AD) is a chronic, inflammatory skin disorder characterized by eczematous and pruritic
Received 18 January 2021 skin lesions. In recent decades, the prevalence of AD has increased worldwide, most notably in
Received in revised form 28 April 2021 developing countries. The enormous progress in our understanding of the complex composition and
Accepted 29 April 2021
functions of the epidermal barrier allows for a deeper appreciation of the active role that the skin barrier
plays in the initiation and maintenance of skin inflammation. The epidermis forms a physical, chemical,
Keywords: immunological, neuro-sensory, and microbial barrier between the internal and external environment.
Skin barrier
Not only lesional, but also non-lesional areas of AD skin display many morphological, biochemical and
Atopic dermatitis
Filaggrin
functional differences compared with healthy skin. Supporting this notion, genetic defects affecting
Inflammation structural proteins of the skin barrier, including filaggrin, contribute to an increased risk of AD. There is
Microbiota evidence to suggest that natural environmental allergens and man-made pollutants are associated with
Treatment an increased likelihood of developing AD. A compromised epidermal barrier predisposes the skin to
increased permeability of these compounds. Numerous topical and systemic therapies for AD are
currently available or in development; while anti-inflammatory therapy is central to the treatment of AD,
some existing and novel therapies also appear to exert beneficial effects on skin barrier function. Further
research on the skin barrier, particularly addressing epidermal differentiation and inflammation, lipid
metabolism, and the role of bacterial communities for skin barrier function, will likely expand our
understanding of the complex etiology of AD and lead to identification of novel targets and the
development of new therapies.
© 2021 Japanese Society for Investigative Dermatology. Published by Elsevier B.V. All rights reserved.

1. Introduction prevalence of AD is estimated at 15–20 % in children and 1–3 %


in adults worldwide [3], with an increase observed in recent
Atopic dermatitis (AD) is a common, inflammatory, pruritic decades [3]. Both the prevalence of AD and recent trends in
skin disorder with a complex etiology, involving interactions prevalence vary between countries, most notably between high-
between the external environment and the skin [1,2]. The and low-income countries. This is particularly apparent for older

* Corresponding author at: Dermatology Clinic, University of Münster, Von-Esmarch-Straβe 58, 48149, Münster, Germany.
E-mail address: luger@uni-muenster.de (T. Luger).

https://doi.org/10.1016/j.jdermsci.2021.04.007
0923-1811/ © 2021 Japanese Society for Investigative Dermatology. Published by Elsevier B.V. All rights reserved.
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

children (aged 13–14 years) for whom prevalence has increased [27,30,31]. The SG is the outermost layer of the epidermis that
primarily in developing countries but has decreased mainly in contains living cells, abundant in cytoplasmic filaggrin-containing
developed countries [4]. Early clinical signs of AD include skin keratohyalin granules [14,27,32]; formation of these liquid-like
dryness, roughness and pruritus, particularly in sensitive skin keratohyalin condensates has recently been shown to be driven by
areas [1,5]. AD has a major impact on the quality of life of patients, liquid-liquid phase separation of filaggrin proteins [32]. The cells in
their parents and caregivers, influencing lifestyle (e.g., restricting the SG are responsible for the synthesis and modification of
clothing, ability to participate in sports, sleep) as well as mental proteins involved in cornification [14,27]. At the SG/SC transition,
health [6]. keratinocytes undergo cell death to form corneocytes; large, flat
Profound alterations in skin barrier function, altered epidermal cells with a tetrakaidecahedron shape embedded in a lipid matrix,
morphology and stratum corneum (SC) lipid composition are well devoid of nucleus and any cytoplasmic organelles, and tightly
characterized in AD along with T helper (Th)2-predominant linked by corneodesmosomes (cell-cell junctions) [14,33]. Recent
inflammation [7,8]. Notably, the identification of loss-of-function evidence suggests that as cells approach the skin surface, a pH shift
mutations in the structural epidermal barrier filaggrin (FLG) gene drives dissipation of the liquid-like keratohyalin granules leading
has shifted understanding of the pathophysiology of AD from a to corneocyte formation [32].
purely immunological disorder towards a disorder substantially The SC comprises three layers with distinct barrier properties,
driven by impaired skin barrier function [1,9]. The filaggrin which likely correspond to the metabolic processing of filaggrin
precursor, profilaggrin, is a complex, highly phosphorylated [34]. The lower and middle layers function as outside-in barriers,
polypeptide that is cleaved during cornification to release whilst the upper layer acts as a “sponge”, in which solutes (such as
functional filaggrin [10,11]. Filaggrin binds to the keratin cytoskel- ions) flow in, with some being retained [34]. In the SC, corneocytes
eton, facilitating the formation of corneocytes, which form the provide physical protection for the underlying epidermis [14,27].
outer surface of the epidermis [11]. In the upper SC, filaggrin is then Corneocytes are surrounded by a resistant structure called a
degraded to free amino acids and subsequent derivatives, cornified cell envelope, a thick (10 nm) layer of crosslinked
including urocanic acid and pyrrolidone-5-carboxylic acid, which proteins that are covalently bound to ceramide lipids that replace
are part of the natural moisturizing factors [10–14]. the plasma membrane [14,35]. These envelopes are essential for
Defects in skin barrier genes have been shown to play a key role effective skin barrier function [35]. Together with extracellular
in development of allergic diseases [15,16]. For example, mutations lipids derived from lamellar bodies and arranged in multiple
in FLG confer a significantly increased risk of developing AD lamellar bilayers, corneocytes create a barrier to prevent water loss
alongside asthma, allergic rhinitis, food allergy, and hay fever and penetration of exogenous molecules from the external
[8,16]. In a study in Flg-null mice (Flg / ), newborn mice exhibited environment [27,35].
dry, scaly skin, but had normal SC hydration and transepidermal A large proportion of SC lipids are synthesized in keratinocytes,
water loss (TEWL) [17]. Premature shedding of SC layers and but some are also derived from extracutaneous sources [36]. The
increased desquamation were observed under mechanical stress, composition and architecture of SC extracellular lipids are critical
and antigens were able to penetrate the SC more efficiently [17]. for normal permeability and barrier function [37]. Several groups
These findings support the role of impaired barrier function as a have confirmed a particularly high percentage of ceramide species
key component of AD development. (50 % dry weight of SC lipids) in the SC, including ultra-long acyl-
Defects in other skin barrier component genes, including serine ceramide subclasses, together with free fatty acids and cholesterol,
peptidase inhibitor Kazal type 5 (SPINK5) mutations [18] and whereas relatively few phospholipids are present [36,37]. An outer
claudin-1 (CLDN1) impairment are associated with asthma, food coating of omega-hydroxy-ceramides is present on the protein-
allergy, and allergic contact dermatitis [19–22]. Inflamed skin and enriched corneocytes in the SC, providing a link between these
skin barrier dysfunction allow penetration of allergens and protein cells and the lipid lamellae [38]. In addition to forming a
antigens from the external environment, which can further trigger hydrophobic permeability barrier, some of the ceramides and free
the immune response and may lead to the development of fatty acids of the SC also exhibit antimicrobial properties which
immunoglobulin (Ig)E-mediated allergies [23]. In this review, we contribute to the barrier to pathogens [36]. In AD, SC lipid
will explore the structure and function of the skin barrier, composition and architecture is disturbed [39].
including its dysfunction in AD, contributing factors and potential In addition, in the living epidermal layers, tight junctions (cell-
approaches to treatment. to-cell junctions) form a barrier for body water and solutes at the
SG level, and have been shown to play an additional role in both
2. Epidermal changes in atopic dermatitis epidermal barrier function and cornification [40]. Organization of
epidermal tight junction barriers is a dynamic process that
The skin consists of many layers and forms a physical, involves intimate interactions between Langerhans cells and the
biochemical, immunological, microbial, and neuro-sensory barrier SG2 layer of keratinocytes [41]. When tight junction barriers are
between the internal and external environment (Fig. 1) [8,10,24]. intact, Langerhans cells can extend dendrites through them to
Most of the protective function is provided by the epidermis, a uptake external antigens that have infiltrated the SC barrier,
multi-layered epithelium comprising the SC, stratum granulosum suggesting tight junctions in skin serve as a liquid-liquid interface
(SG), stratum spinosum (SS) and the stratum basale (SB) [25,26]. barrier [41].
Keratinocytes are the predominant cell type in the epidermis, The SC provides a physical barrier against harmful substances
and undergo differentiation in an oriented process from the SB, the from the environment [29]. If pathogens enter the body due to a
deepest epidermal layer, toward the external surface of the skin disrupted barrier, the immune response is important to protect the
[25]. The SB is mainly composed of proliferating keratinocytes that living epidermal layers and the entire body [41]. In addition to
are attached to the dermis via complex multi-protein structures constituting an integral structural element of the epidermis,
[27]. The SS overlies the SB, comprising a variety of cells that differ keratinocytes are involved in both innate and adaptive immunity
in shape, structure and subcellular properties [28,29]. Within the [29]. Along with neutrophils, keratinocytes produce antimicrobial
SS, keratinocytes start producing lamellar bodies - membrane- peptides (AMPs) and act as a first line of defence against external
bound organelles and tubuloreticular networks containing a pathogens [29]. For example, keratinocytes have been shown to
number of lipids, glycosidases and acid hydrolases that function produce pro-inflammatory cytokines such as interleukin (IL)-1β
to deliver precursors of SC lipids into the intercellular space and IL-18 via the inflammasome signaling pathway, and to interact

143
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

Fig. 1. The components of the epidermal barrier form a microbial, physical, chemical, immunological and neuro-sensory barrier.
Microbial products contribute to a normal epidermal barrier in several ways. Microbes produce AMPs, as well as free fatty acids, and stimulate the innate immune system
[120,124]. The physical barrier consists of corneocytes, desmosomes, claudins and lipids [8,10,22]. The chemical barrier is composed of molecules that contribute to hydration
(e.g., NMFs) and the prevention of infections (e.g., AMPs) [10]. The immunological barrier involves components of the innate immune system (e.g., AMPs produced by
keratinocytes) and the adaptive immune system (e.g., LCs and lymphocytes) [8,10,254]. Components of the neuro-sensory system function as danger sensors and include
keratinocytes and ion channels expressed on cutaneous nerves [29,255]. Upon activation, sensory nerve endings release neuropeptides such as substance P, which mediates
itch, pain and inflammation [255–257].
AMP, antimicrobial peptide; DC, dendritic cell; ECM, extracellular matrix; Eo, eosinophil; IL, interleukin; ILC, innate lymphoid cell; LC, Langerhans cell; M, macrophage; N,
neutrophil; NMF, natural moisturizing factor; TLR, toll-like receptor.

with T cells, inducing T cell activation or antigen-specific tolerance total lipids have been observed in the SC of patients with AD, which
[29]. In addition to T cells and keratinocytes, Langerhans cells are may explain the impaired water-barrier function of the skin [37].
present in the epidermis, where they play an important role in Even more important is a reduction in the chain length of the fatty
adaptive immunity [29]. acids in the ceramides, a reduction in the chain length of the free
Patients with AD exhibit injured skin that is inflamed, as fatty acids, as well as changes in the amounts of saturated and
demonstrated by erythema and pruritus [42]. Acute skin lesions unsaturated fatty acids, which correlate with SC lipid bilayer
are characterized by intensely pruritic papules over erythematous structure [55]. Langerhans cells, mast cells and inflammatory
skin; chronic skin lesions are distinguished by lichenification and dendritic epidermal cells have also been observed in AD skin, with
fibrotic papules [42,43]. Impairment of skin barrier function, as the latter two cell types appearing to migrate from the dermis into
assessed by increased TEWL values and increased percutaneous the epidermis [29,56–58]. The dendrites of Langerhans cells, but
penetration, is a characteristic of AD patients’ skin in both non- not inflammatory dendritic epidermal cells, extend through the
lesional and lesional areas [22]. Non-lesional areas of skin may be tight junction barriers, probably to capture external antigens [58].
dry and scaling and display subclinical inflammation [13,44–48]. These differences in cell behavior may play a role in AD
Lesional areas present marked intercellular edema (spongiosis), pathophysiology [58].
prominent hyper- and sometimes parakeratosis and acanthosis An important aspect of the skin barrier is pH; the pH of normal
[42,49]. In terms of structural changes, abnormal keratohyalin non-neonatal skin surface ranges from 4 to 6, but in AD it is often
granules have been observed in the SG of AD skin, together with increased towards neutral and basic levels [59,60]. Lesional skin
thinning and sometimes, absence of this cell layer [50]. In addition, has the highest pH in patients with AD, and unaffected skin has a
atomic force microscopy studies have shown an increased number higher pH than in control subjects [61]. Less acidic areas of skin
of villus-like protrusions on the corneocyte surface in AD skin [51] surface are more susceptible to AD expression; the higher pH of the
and corneocytes are reduced in size [52]. Furthermore, the cheeks in infants, and of intertriginous and flexural skin in adults,
cornified envelope is defective with highly decreased compaction corresponds to the increased likelihood of AD lesions in these areas
of corneocytes and reduced intercellular lipids [53]. Abnormalities [59]. The formation of the SC involves several pH-dependent
are observed in lamellar body secretion in the epidermis of AD skin enzymes, particularly for the synthesis and arrangement of lipids
[54], with lamellar bodies retained in the corneocyte matrix and within the layer [60]. Increased pH disturbs barrier formation,
alterations observed in extracellular lamellae in the SC [54]. A reduces the antimicrobial capability of the skin, and may cause
significant reduction in the proportion of very long chain patients with AD to be more susceptible to infections by
ceramides in the lipid composition as well as a lower amount of Staphylococcus and other neutral-pH-favoring pathogenic bacteria

144
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

on the skin [59]. Desquamation of the SC also occurs more rapidly loci [75]. So far, the greatest genetic significance has been
at neutral pH than at acidic pH, leading to decreased integrity of demonstrated for mutations in the FLG gene, which lead to
the skin barrier [59]. defective skin barrier function [16]. The skin barrier may be most
Data indicate that multiple proteins involved in epidermal important in AD initiation and is being targeted for AD primary
development and barrier function are altered in AD skin (Fig. 2) prevention with promising early results [76]. However, to date
[62]. Gene and protein expression of cornified cell envelope these results have not been replicated in larger studies [77].
components including loricrin, involucrin, filaggrin, filaggrin-2, To date, defects have been noted in genes encoding four
hornerin and others are significantly reduced in AD, and this may important components of the epidermal barrier: corneocytes (FLG,
be correlated with the severity of disease [45,53,62–65]. On the filaggrin-2 [FLG2], OVOL1, Small Proline Rich Protein 3 [SPRR3]),
other hand, keratin K6/K16 are upregulated [45,53,66]. AD is also lipid-rich matrix (Mattrin [TMEM79]), tight junctions (CLDN1), and
associated with reduced expression of corneodesmosomal pro- protease inhibitors (SPINK5) (Table 1 [12,16–18,22,78–88]) (Fig. 2).
teins, most notably corneodesmosin [67–69]. Defects in FLG and FLG2 genes are associated with impairment of
the epidermal barrier and more persistent symptoms of AD
3. Genetics of AD [16,89]. OVOL1 knockout reduces the production of filaggrin and
the integrity of the skin barrier, each of which is associated with a
As previously mentioned, defects in skin barrier genes can greater risk of AD [8,15,16,82,85–87,90]. Further, a mutation in
contribute to the development of AD (Fig. 2). The heritability of AD SPRR3 has been associated with a 20 % increased risk of AD [80].
is approximately 75 % and the concordance rate is higher in Presence of the missense mutation rs6684514 in the TMEM79 gene
monozygotic twins compared with dizygotic twins (72 % vs 23 %), impacts the lipid matrix of the skin barrier and is associated with
indicating that genetic factors play a major role in the development AD [83,84]. In addition, defects in genes involved in tight junctions
of AD [70]. Family genetic studies identified several possible AD and epidermal remodeling, including CLDN1 and SPINK5, respec-
regions (e.g., ATOD1: 3q21); however, these studies were not tively, have been associated with AD [18,22,78,88,91]. The
definitive in their results [71–73]. Subsequent genetic studies expression and activity of proteases, such as Kallikrein-related
recognized that AD is a disease likely involving common variants, peptidase 7 (KLK7) (formally known as stratum corneum
thus, several Genome Wide Association Studies (GWAS) were chymotryptic enzyme), are also important determinants of barrier
pursued [74]. A study investigating over 360,000 cases of eczema, thickness and integrity [52]. A significant association between a
asthma and hay fever versus healthy controls identified 99 AD risk mutation in the gene encoding KLK7 and AD was found in a study

Fig. 2. Genetic and environmental factors lead to skin barrier breakdown in AD. [Reprinted and adapted from J Invest Dermatol, Vol 129(8), Cork MJ et al., Epidermal barrier
dysfunction in atopic dermatitis, pp. 1892-908, © 2009, with permission from Elsevier] [52].
AD is a heterogeneous condition in which a variety of genetic and environmental causes lead to skin barrier breakdown [1]. Filaggrin gene variants may lead to decreased
natural moisturizing factor, which reduces stratum corneum hydration and increases pH [16]. Increases in pH enhance protease (KLK5, KLK7 etc.) activity and inhibit lipid-
generating enzymes [52]. Together with defects in the genes encoding proteases and protease inhibitors (e.g., SPINK5), these changes increase the breakdown of
corneodesmosomes, deregulate desquamation and impair lipid lamellae formation [52,78]. Genetic changes in cornified envelope (e.g., FLG variants and SPRR3), lipid matrix
(e.g., TMEM79) and tight junction (CLDN1) components are thought to impair the structural integrity of the cornified envelope and lipid lamellae [16,22,80,83,84]. Tight
junction defects and increased pH impair antimicrobial activity, increasing the probability of S. aureus infections, which then worsen skin barrier breakdown [42,253].
Environmental factors such as soap, detergents and exogenous proteases further enhance protease activity, interacting with genetic defects to break down the skin barrier [1].
When the skin barrier is impaired, the penetration of irritants and allergens into the skin increases, triggering skin inflammation and raising protease activity [42,52].
*includes filaggrin.
AD, atopic dermatitis; CLDN1, Claudin 1; FLG, filaggrin; KLK5, Kallikrein-related peptidase 5; KLK7, Kallikrein-related peptidase 7; S. aureus, Staphylococcus aureus; SPINK5,
Serine Peptidase Inhibitor Kazal Type 5; SPRR3, Small Proline Rich Protein 3; TCS, topical corticosteroids; TMEM79, Transmembrane Protein 79.

145
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

of 103 patients with AD and 261 matched controls [52]. In addition, neuro-immune cross-talk may play a role in airborne protein-
mice overexpressing human KLK7 develop skin changes similar to mediated skin inflammation [94].
those observed in chronic AD [52]. Other extrinsic environmental risk factors for AD development
include temperature, humidity, ultraviolet radiation, air pollutants,
4. Influence of environment on AD tobacco smoke exposure, water hardness, rural versus urban living,
diet, breastfeeding, probiotics, and prebiotics [95]. These risk
Natural allergens such as house dust mites, cockroaches, pet factors are thought to contribute to AD by acting as pruritogens and
dander and multiple pollens have all been reported to induce AD in irritants, upregulating inflammatory processes and/or worsening
a subpopulation of patients, through cutaneous exposure [92]. skin barrier function [95].
Such airborne proteins penetrate the epidermis and worsen the Several epidemiological studies have assessed the association
severity of AD [92]. The prevalence of AD due to airborne proteins between air pollution particulates (nitric oxides [NOx], sulfur
is hard to estimate as it is difficult to prove which airborne factors dioxide [SO2], ozone [O3], general traffic exhaust emissions) and
are involved. Patients with AD relating to airborne proteins the epidemiology of AD [96]. Unfortunately, available data cannot
typically have lesions on air-exposed skin areas, and a history of support a meaningful meta-analysis to investigate this association
exacerbation after exposure to airborne proteins [92]. In addition, [96].
their disease is normally severe and resistant to conventional A recent systematic review of 57 environmental epidemiologic
therapies, and often co-occurs with asthma [92]. In a 5-year studies was performed to determine if an association exists
retrospective study, patients who had a positive pin prick test for between air pollution and AD [97]. In 31 studies, the association
inhalation allergens, including dust mites, grass pollen and weed between air pollution and the prevalence of AD was assessed. A
pollen, were more likely to have asthma and allergic rhinitis [93]. significant positive association between AD and traffic exhaust-
The authors concluded that the risk of developing asthma and related emissions, especially from truck traffic, was found in 8 out
allergic rhinitis is very high in patients with AD [93]. The of 11 cross-sectional studies with small-scale measurement or
mechanisms via which airborne proteins induce AD are only modeling of exposure [97]. Further, in 5 out of 5 studies, a
partly understood [94]. One recent study used a mouse model of significant positive association was found between AD and air
AD to show that house dust mites directly activate clusters of pollution based on self-assessed traffic intensity, in particular that
MRGPRB2+ mast cells and TRPV1+Tac1+ nociceptors (nociceptive of trucks [97]. An association between AD and measured
sensory neurons) to drive type 2 skin inflammation [94]. Therefore, background pollution levels was detected in only 2 out of 15

Table 1
Genetic defects of the skin barrier associated with atopic dermatitis.

Skin barrier Mutations


component
Corneocyte  Filaggrin (FLG)
o Mutations strongly associated with AD in specific ethnicities [16,86]
o Deficiency leads to:
& Failure in skin barrier function [16]
& Decreased UCA and PCA (natural moisturizing factors) in the SC [12,79]
& Flg / mice have dry, scaly skin; premature shedding of SC layers and increased desquamation under mechanical stress; increased antigen
penetration of the SC [17]
 Filaggrin-2 (FLG2)
o S2377X (stop-gain mutation) and H1249R (missense mutation): >1.5 fold risk of AD persistence [81,89]
 Ovo Like Transcriptional Repressor 1 (OVOL1); transcription factor in suprabasal keratinocytes that promotes FLG expression [87]
o GWAS variant rs10791824 increases risk of developing AD by 12 % in Europeans [82]
 Small Proline Rich Protein 3 (SPRR3); cross-bridging protein in the cornified cell envelope [80]
o Variant rs28989168 with extra repeats of 8 amino acid sequence, increases the risk of developing AD by 20 % [80]

Lipid matrix  Mattrin (TMEM79); discovered through genetic analysis of Flaky Tail mouse [83]
o Missense mutation rs6684514 associated with human AD and defective skin barrier [84]
o Mouse models of TMEM79 negative mice displayed [83,84]:
& Increased IgE levels*
& Spontaneous dermatitis
& Atopy
& Increased cutaneous sensitization*
& Impaired lamellar body protein secretion
& Impaired SC formation

Tight junctions  Claudin-1 (CLDN1); suprabasal tight junction protein [22]


o Expression decreased in non-lesional AD skin [22]
o Genetically associated with AD in Europeans, African-Americans and Ethiopians [22,91]
o Deficiency associated with increased susceptibility to HSV-1 [253]
o Variant may potentiate effect of mold exposure on AD risk [88]

Epidermal  Serine Peptidase Inhibitor, Kazal Type 5 (SPINK5); epidermal protease activity inhibitor
remodeling o Autosomal recessive mutations in Netherton Syndrome and associated with AD and allergy [18,78]
o Genetically associated with AD in multiple studies in Japan, China and Taiwan [18,78]
o Mutations result in [78]:
& Decreased expression of filaggrin
& Increased expression of pro-Th2 cytokine TSLP

AD, atopic dermatitis; Flg / , Flg-null; GWAS, genome-wide association study; HSV-1, herpes simplex virus type 1; PCA, pyrrolidone-5-carboxylic acid; SC, stratum corneum;
t-UCA, trans-urocanic acid; TSLP, thymic stromal lymphopoietin.
*
when compared with wild-type mice.

146
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

studies. Significant associations were found between maternal air expression is thought to play a role in controlling bacterial
pollution exposure during pregnancy and AD in recent birth cohort colonization and infections associated with skin barrier disruption
studies in South Korea, China, and Taiwan [98–102]; despite the in AD [110,111]. However, the level of upregulation observed in the
greater levels of fine particulate matter (PM10) in China, this skin of patients with AD was considerably lower than in patients
significant association with AD was found only for NO2, supporting with psoriasis (in whom skin infections are less common),
the relationship between traffic pollution and AD [97,99–101]. indicating an inadequate increase in epidermal AMPs to prevent
However, in corresponding studies in Europe and the USA, such bacterial colonization [110,112,114].
associations were unclear or not investigated [97]. In addition, in 7
out of 10 predominantly East Asian studies (in South Korea and 6. Role of the microbiome in AD
China), symptom intensity in individuals with existing AD was
observed to increase with greater concentrations of particulate The skin microbiome represents the totality of microbes and
(PM10, PM 2.5) and gaseous (NO2, volatile organic compounds, O3, associated genes resident of the human skin. These communities
SO2) air pollutants [97]; 4 of these studies indicated that vary between each of the three layers of the skin (epidermis,
meteorological factors such as low relative humidity are also dermis, hypodermis) due to differences in physicochemical
associated with worsening AD [103–106]. environments, such as moisture, pH and oxygen concentration
While there is no evidence to indicate that AD is affected by [115,116]. The skin microbiome and its interaction with human
‘large scale’ varying exposure to air pollutants such as PM10 or SO2 immunity is a vital consideration in understanding skin disease
[97], there are considerable data to suggest that ‘small scale’ pathophysiology [117–121]. Recent advances in genetic sequencing
varying exposure to traffic exhaust pollution (particularly truck technologies and computational tools have allowed sequencing of
traffic) can influence the prevalence of AD [97]. In addition, traffic a large proportion of the microbiome, which will further our
exhaust emissions have been shown to increase the incidence (as knowledge of interactions between host and bacterial communi-
assessed through maternal exposure during pregnancy) and the ties [122].
symptoms of AD [97]. Further research is required to identify Cutaneous microorganism communities closely interact with
which particulates contribute to these observed effects, and the host innate immune effectors including macrophages, natural
potential mechanisms through which air pollution-induced health killer cells, and mast cells [123–125]. Of interest, commensal
effects are mediated. For this reason, there is virtually nothing microorganisms are also capable of modulating gene expression in
known about potential mechanisms that might mediate air the skin and have been shown to be involved in the upregulation of
pollution-induced health effects relevant for AD. As air pollu- innate immune response genes and genes involved in cytokine
tion-induced health effects require the penetration of pollutants activity [124]. The skin's innate immune system consists of
into viable skin, skin barrier integrity is most likely involved. In resident or skin-recruiting effectors [126,127]. Crosstalk between
support of this, a recent study found that topical exposure of skin commensals and immune effectors is possible due to the
mouse skin to diesel exhaust particles increased the expression of expression of PAMP (pathogen-associated molecular pattern)-
the neurotrophic factor artemin in keratinocytes, through a specific receptors (PRRs) [128]. PRRs are classified into four major
mechanism involving the activation of the aryl hydrocarbon families [128,129]: Toll-like receptors (TLRs), C-type lectin
receptor [107]. In addition, artemin was observed to induce the receptors (CLRs), nucleotide-binding oligomerization domain
growth of transient receptor potential cation channel subfamily V (NOD)-like receptors (NLRs) and RIG-like receptors (RLRs). PRRs
member 1 (TRPV1)-positive nerve fibers in the skin and to cause allow cells involved in innate immunity to monitor and sense
pruritus [107]. Furthermore, overexpression of the xenobiotic commensal and pathogenic microorganisms and are generally
receptor pregnane X receptor (PXR) impairs the epidermal barrier specific to certain classes of microbial pathogens [128,129]. With
and triggers a Th2 immune response [108,109]. these close interactions between skin microorganisms and
immune effectors, a balance is crucial to maintain skin homeosta-
5. Role of antimicrobial protein expression in AD sis and prevent unnecessary inflammatory responses. Imbalance
within the skin microbiome is a potent trigger for skin inflamma-
Patients with AD frequently suffer from skin infections caused tion and is described in several inflammatory dermatoses, in
by bacteria and viruses [110,111]. Staphylococcus aureus (S. aureus) particular in AD [42,116,130–132].
is the most common bacterial infection afflicting patients with AD, Dermatological conditions such as psoriasis and AD have been
90 % of whom are colonized with S. aureus versus 5–30 % of healthy associated with loss of diversity in the skin microbiome
individuals [111]. Impaired AMP expression in the skin of patients [42,116,130–134], with AD being characterized by the dispropor-
with AD is suggested to contribute to the increased susceptibility tionate colonization of S. aureus [42,130,131]. In addition,
to bacterial infections [111]. Corynebacterium mastitidis, S. aureus and Corynebacteruim bovis
Human beta-defensin (hBD)-2, hBD-3, and human cathelicidin (C. bovis) have been found to emerge sequentially during AD onset
peptide (LL-37) are AMPs found in the epidermis; the target of LL- [135]. Antibiotics that specifically target these bacteria almost
37 is unknown, however hBD-2 is known to target Gram-negative completely reverse the associated dysbiosis and skin inflammation
bacteria, and hBD-3 is known to target S. aureus [110–112]. In [135]. S. aureus was found to drive AD formation, whilst C. bovis
comparisons between the epidermis of patients with AD and induced Th2 cell responses [135]. The role of different dysbiotic
patients with psoriasis, a markedly reduced expression of all three flora during AD onset suggests that the skin microbiome may be a
AMPs was observed supporting the hypothesis of impaired AMP potential target for future therapeutic interventions [135].
expression in AD, probably as a consequence of the Th2 cytokine Indeed, probiotics and prebiotics are being explored as
effect [112,113]. innovative treatments for dermatological diseases such as AD
Furthermore, an analysis of the skin of healthy individuals and [136–140]. Interestingly, nutritional supplementation with pre-
patients with psoriasis or AD found that hBD-2, hBD-3, RNase-7 (a biotics and probiotics has been shown to improve symptoms,
broad spectrum AMP), and psoriasin (a strong inhibitor of quality of life and clinical severity of AD in some studies, although
Escherichia coli growth, also known as S100A7) showed increased this could not be verified in others [137,141].
expression in the skin of patients with AD compared with the The impact of topical treatment on the skin microbiome, and
epidermis of healthy individuals, but a weaker induction than that consequent influence on AD, has also been explored in multiple
seen in patients with psoriasis [114]. This upregulation of AMP studies [142–149]. A study showed that long-term emollient use in

147
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

infants at high-risk of developing AD reduced skin pH and study period, the emollient plus resulted in 56 % and 60 %
increased the proportion of Staphylococcus salivarius in the skin reductions in mean xerosis and pruritus severity, respectively
[146]. Emollient-mediated microbiome changes may consequently [147]. These improvements in xerosis and pruritus were significant
play a role in restoring skin dysbioses and preventing AD, although regardless of skin condition (P < 0.0001) [147]. In another recent
additional studies are required to confirm this [146]. The topical study, an ointment containing Lactobacillus reuteri DSM 17,938 was
application of specific strains of bacteria has also been investigated safe and led to significant improvements in SCORing AD (SCORAD)
for the treatment of AD. One study highlighted the importance of index and local SCORAD after 4 and 8 weeks of treatment in adults
supplemented emollients in the management of AD; in this with AD, compared with baseline values (P < 0.001) [148]. Finally, a
randomized comparative trial, 60 patients with moderate AD were study conducted by the National Institutes of Health found that
treated with an emollient supplemented with a biomass of specific strains of Roseomonas mucosa applied to lesional skin
Vitreoscilla filiformis, or a different emollient without biomass provided clinical benefit to both children and adults with
supplementation, twice daily for 1 month [142]. Patients who established AD [149]. Additional studies of microbiome trans-
received the supplemented emollient experienced greater plants are currently underway in AD, including those that build on
improvements in AD scores than those treated with the previous research, demonstrating that reintroduction of coagu-
comparator emollient [142]. The authors concluded that the lase-negative Staphylococcus strains on the skin of patients with AD
supplemented emollient was able to normalize skin microbiota (following isolation from healthy subjects and patients with AD)
and reduce the number and severity of AD flares compared with reduced colonization by S. aureus [149,150].
emollient alone [142]. A recent open-label and real-world 7-day Possible crosstalk between the skin microbiome and other
study evaluated the use of an ‘emollient plus’ containing an human body microbiomes is an emerging concept that suggests a
Aquaphilus dolomiae extract in 5,910 patients with a variety of skin gut-skin axis [151–153]. The mechanisms by which the gut
conditions (3,511 [65.2 %] patients had AD) [147]. Over the 7-day microbiome influences skin homeostasis have not yet been fully

Fig. 3. The pathophysiology and treatment of AD. [Adapted from Kim J, et al. Allergy Asthma Proc. 2019] [258].
Inherited genetic defects and epigenetic changes in skin and immune components are thought to contribute to the immune dysregulation and skin barrier dysfunction that
underlies the symptoms observed in AD [18,72,158]. Immune abnormalities alter epidermal lipid composition and this together with tight junction defects allows microbes
and allergens to infiltrate the skin, leading to further skin inflammation and skin barrier impairment [42,253,258]. Gut microbial dysbiosis may also modulate systemic
immunity and promote pro-inflammatory responses [151–154]. Therapy for AD may be preventative or reactive [2]. Maintenance therapy with emollients is the main
underlying strategy in AD treatment, and is used to repair the skin barrier and prevent AD relapse [2,158]. Recent research has highlighted the possible utility of ceramide-
containing emollients and probiotics in the management of AD and the prevention of relapse [158,161,164,165]. Reactive treatment of AD typically involves the use of topical
anti-inflammatory agents (e.g., TCI and TCS) to correct immune dysregulation, emollients to hydrate and repair the skin and topical antibiotics in cases of bacterial
colonization [2,175–177,186,187]. In severe AD, systemic anti-inflammatory agents, biologics and light therapy may be used to target immune dysregulation [189,217–219].
Several new biologic agents and small molecule drugs are in development for the treatment of AD [249–252].
AD, atopic dermatitis; TCI, topical calcineurin inhibitor; TCS, topical corticosteroids.

148
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

explored; however, according to recent studies, this could involve 8. Topical treatment for AD
the modulatory effect of gut bacteria commensals on systemic
immunity [154]. For example, specific gut microorganisms and Topical treatments are used in the management of AD to
metabolites (retinoic acid, polysaccharide A) from Bacteroides suppress inflammation, relieve symptoms such as itching, restore
fragilis and Faecalibacterium prausnitzii are able to promote the skin barrier function and to prevent relapse of AD. Due to the
accumulation of regulatory T cells and Th cells, which facilitate reduced risk of adverse events when compared with systemic
anti-inflammatory responses and pro-inflammatory responses, treatments, topical treatments are well suited for patients with
respectively [155]. Further research is needed to investigate the mild-to-moderate AD [175]. Liberal use of emollients is an
gut-skin axis and its impact on skin disease physiopathology. important part of AD treatment, with long-term studies demon-
Importantly, understanding these pathways will lead to novel strating that use of emollients may be effective in reducing the risk
therapies based on targeting one organ (i.e., the gut) to improve the of AD development in infants, and in reducing the need for TCS in
health of another (i.e., the skin). established disease [76,146,158].
TCS have formed the cornerstone of AD treatment for more
7. Prevention of AD than 50 years, and are recommended for the treatment of AD
globally [2,175–177]. There are many different compounds and
Treatment options for AD are illustrated in Fig. 3. The formulations of TCS, which can vary greatly in potency
management of AD focusses first on taking steps to prevent AD [2,176,178]. TCS are recommended for the short-term treatment
and allow skin barrier recovery. Reducing contact with allergens of AD flares as, although highly effective at reducing inflammation
and irritants, with the use of specific barrier creams or ointments, and pruritus, persistent use increases the risk of local side effects
can give the skin barrier time to recover and prevent inflammation on the skin [2,178]. More potent formulations are associated with
[92,156]. Repair of the skin barrier can take longer than clinical greater risk of adverse events [175,178], which can include skin
improvement, particularly when corticosteroids are used to reduce atrophy, skin barrier impairment, increased risk of skin infections
the clinical signs of AD, such as reddening of the skin [157]. In these and tachyphylaxis [175,178–181]. The risk of systemic side effects
instances, inflammatory cells may still be present and as such, with the use of TCS is low and linked to the potency of the
allergens must be avoided for a prolonged period to allow skin formulation, extent of skin inflammation and the age of the
barrier repair [157]. General skin care, generous use of emollients patient [175]. However, corticophobia (patient fear of TCS-related
containing lipid mixtures and water binding compounds [158], and side effects) is a substantial challenge that has arisen with the use
proactive treatment with topical corticosteroids (TCS) or topical of TCS [175].
calcineurin inhibitors (TCIs) [159,160], can help to prevent AD TCIs selectively inhibit nuclear factor of activated T cells (NFAT)
relapse. to decrease the activity of a range of transcription factors that
There are several preventative measures under investigation control cell division and reduce the expression of pro-inflamma-
that specifically target the skin barrier. These include topical tory cytokines, including cytokine Th1 and Th2 [182,183]. TCIs also
formulations of cannabinoid receptor modulators, proteins, inhibit mast cell and neutrophil activation, release of inflammatory
adjusted pH, flavonoids, plasminogen activator type 2 receptor mediators and directly influence nerve fiber function by binding to
(PAR2) inhibitors, and lipids [161–163]. Cer-dominant, triple- the TRPV1 [2,175,176,182,184]. Binding to TRPV1 results in an initial
physiologic lipid, barrier repair therapy for AD (Cer:cholesterol: burning sensation with TCI use that typically improves with the
free fatty acids at a 3:1:1 M ratio), has been reported to target both resolution of lesions [175,182,185].
the reduction in lipids and the decline in Cer observed in AD, AD management guidelines recommend the use of TCIs,
therefore targeting two abnormalities central to skin barrier particularly pimecrolimus, for the treatment of mild-to-moderate
dysfunction. This formulation, when provided at an acidic pH, has AD in sensitive skin areas [2,176,186–189]. Tacrolimus ointment is
shown efficacy in humans with AD [161,164]. A more recent study approved for the treatment of moderate-to-severe AD: tacrolimus
investigated the use of a cream and lotion containing ceramides 1, ointment 0.03 % for the treatment of patients aged 2–15 years and
3 and 6-II, triglycerides and cholesterol [165]. Unlike earlier tacrolimus ointment 0.1 % for patients aged 16 years [176].
studies, these agents were delivered via a multi-vesicular emulsion Pimecrolimus 1 % cream is approved for mild-to-moderate AD in
(MVE) controlled-release system [165]. When compared with adults and children (aged 2 years), and is also approved in
three reference emollients in a double-blind, intra-subject, several countries for use in children aged 3 months [190]. A
vehicle-controlled, single open-application test, the test cream meta-analysis of the long-term treatment of children with AD
and lotion were the only products to impart a clinically meaningful found both pimecrolimus and tacrolimus to have similar efficacy
improvement in skin hydration and dryness that was sustained for in treating the clinical signs of AD [191]. Pimecrolimus and
24 h following a single application [165]. This may reduce the tacrolimus have also demonstrated clinical benefits as mainte-
burden of managing AD with typical emollients, which typically nance treatments for AD. When compared with vehicle treat-
require application 3–4 times per day [165]. ment, tacrolimus treatment significantly reduced the number of
Experimental barrier disruption and subsequent monitoring of disease exacerbations requiring substantial therapeutic interven-
barrier recovery has been used to uncover disease mechanisms and tion (median difference, 2; P < 0.001) [159] and intermittent
pathology that is not detected by the assessment of basal function treatment with pimecrolimus significantly reduced the median
[166]. Using barrier recovery studies, signals and metabolic time to first AD flare (P < 0.001) [192]. When compared with
responses to barrier abnormality have been identified, including conventional TCS, treatment of patients with AD with tacrolimus
a delay in barrier recovery when cholesterol synthesis is inhibited 0.1 % ointment for one year reduced the severity of skin atrophy,
[167,168]. Barrier recovery studies have also been used to compare which was associated with an improved skin barrier and increased
the efficacy of putative preventive and therapeutic strategies. In collagen synthesis [179,193]. In addition, in accordance with
addition, the topical use of corticosteroids [169] and the systemic European guidelines, pimecrolimus is recommended for the
use of cyclosporine (CsA) [170] and cytokine-receptor antibodies treatment of facial lesions and children [2]. Contrary to historical
(e.g., anti-IL-4 receptor antibody dupilumab) [171,172], have been reports, there is no evidence for an association between the use of
shown to indirectly enhance the skin barrier to a certain degree. In TCIs and the occurrence of lymphomas or skin tumors [194]. While
contrast, allergic reaction, malnutrition and essential fatty acid transient application site burning is common following TCI
deficiency can delay skin barrier recovery [173,174]. application, treatment with pimecrolimus has demonstrated less

149
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

severe and shorter duration application site reactions, including AD in 3D in vitro skin models [214]. However, neither agonist nor
burning sensations, compared with tacrolimus in patients with AD antagonist changed skin barrier function [214]. In human skin,
[195]. Importantly, pimecrolimus does not impair the skin barrier VTP-38543, a LXR agonist, has been shown to significantly increase
and has been shown to restore the epidermal barrier in damaged the expression of loricin and filaggrin within the epidermal skin
skin via the regulation of genes which are essential for normal skin barrier compared with vehicle treatment in patients with mild-to-
barrier function [157]. moderate AD (P = 0.02) [216]. VTP-38543 also suppressed cellular
Novel topical treatments for AD have begun to emerge. infiltrates and down-regulated expression of innate immunity
Phosphodiesterase 4 (PDE4) inhibitors, such as crisaborole and markers, including IL-6 [216]. Although the skin barrier was
difamilast, inhibit PDE4 to reduce the release of pro-inflammato- restored, no improvement in clinical scores for AD were observed
ry cytokines [196,197]. Crisaborole is approved in the United [216].
States (US) for the treatment of mild-to-moderate AD in patients
aged 2 years [198,199]. It has been reported to induce skin 9. Systemic treatment for AD
barrier-related RNA transcripts and to decrease TEWL within 1–2
weeks of topical application [200]. Crisaborole 2 % ointment There are several available guidelines and reviews on the
significantly reduced the severity of pruritus compared with efficacy, safety and clinical use of various systemic therapies for the
vehicle treatment in a pooled analysis of clinical data [196], while treatment of AD [189,217,218]. These guidelines provide recom-
being well tolerated, with the most commonly reported mendations in terms of which treatments are best suited for the
treatment-related adverse events being application site pain severity of AD, but most do not provide guidance on when to
and pruritus [196]. Similarly, difamilast 1 % cream has rapid and advance therapy from topical treatments. The International
sustained effects on pruritus and is currently being investigated Eczema Committee (IEC) Expert Panel provides an algorithm to
in the treatment of AD [197]. guide decision making on when to advance to systemic therapy
Janus kinase (JAK) has been implicated in AD, with inhibition of [219]. The algorithm considers factors such as the severity of AD,
JAK being associated with sustained anti-pruritic effects [201]. level of patient education provided, any alternative diagnoses
Tofacitinib is a potent pan-JAK antagonist developed in both (infections or allergic contact dermatitis), amount of time on TCS
topical and oral formulations [202–204]. In a small clinical trial in treatment, quality of life of the patient and use of phototherapy
six patients with moderate-to-severe AD refractory to standard [219].
treatment, a decrease in SCORAD with tofacitinib was observed There are many systemic therapies that have demonstrated
(36.5 % at week 8 to 12.2 % at week 29 [P < 0.05]) [205]. Ruxolitinib clinical efficacy in the treatment of AD [189]; however, a limited
is a potent JAK1/JAK2 inhibitor, which provides a high degree of number of these therapies have been shown to improve barrier
myelosuppression [206–208]. In a recent Phase 2 trial in patients integrity via the study of biomarkers [220]. Biomarkers provide an
with AD, treatment with ruxolitinib cream significantly improved objective measure of response to therapy and are particularly
the Eczema Area and Severity Index (EASI) score versus vehicle important in diseases that have a complex etiology, such as AD
cream at week 4 (71.6 % improvement vs 15.5 %, P < 0.001) [206]. [220].
Ruxolitinib cream also provided significant improvement in the Narrow-band ultraviolet B (UVB) (311–312 nm) phototherapy
Investigators Global Assessment (IGA) score and pruritus com- is utilized in the treatment of a wide range of conditions including
pared with vehicle at week 4, with only mild or moderate adverse AD, vitiligo, generalized pruritus and psoriasis [221–224]. UVB
events reported [206]. A further pan-JAK inhibitor under treatment has multiple modes of action including apoptosis of
investigation for the treatment of AD is delgocitinib [209]. In a infiltrating T cells [222], immunosuppression [221,225], inhibi-
Phase 2 randomized-controlled trial in adults with moderate-to- tion of pro-inflammatory cytokines [226], antimicrobial proper-
severe AD, EASI scores were significantly improved at 4 weeks ties against S. aureus [227], and upregulation of the cutaneous
following treatment with all doses of delgocitinib compared with vitamin D system [228]. Suberythemal doses of UVB radiation
vehicle ointment (P < 0.001) [209]. All reported adverse events have been shown to increase mRNA levels for key epidermal
were mild or moderate in severity, and included nasopharyngitis enzymes for the synthesis of cholesterol, fatty acids, and
and application-site skin infections [209]. There is preliminary sphingolipids in mice [228]; increases in epidermal involucrin
evidence from in vitro studies and from animal testing that JAK- and filaggrin expression were also observed, indicating suber-
inhibition is beneficial for the skin barrier [210]. ythemal doses of UVB may aid corneocyte formation and result in
Tapinarof is a novel topical aryl hydrocarbon receptor (AhR) overall enhancement of barrier function and homeostasis through
agonist that has recently been investigated in patients with AD several mechanisms [228]. Treatment of AD with UVB photother-
[211,212]. In this study, the rate of treatment success (minimum 2- apy has been shown to have many positive clinical effects;
grade IGA score improvement) with tapinarof cream at week 12 however, due to the risk of possible adverse events, phototherapy
ranged from 34 to 53 % (dose dependent) and was significantly should be used only as a second-level treatment in adults and
higher than that with vehicle treatment (24–28 %). Further, children [2].
treatment success was maintained 4 weeks following tapinarof CsA is a strong immunosuppressant that has been used for the
therapy and treatment-emergent adverse events were mild-to- treatment of many conditions, and is approved for the manage-
moderate in intensity [211]. ment of AD in Europe [189]. CsA binds to cyclophilin, creating a
Liver X receptors (LXRs) are involved in the maintenance of the CsA-cyclophilin complex to reversibly suppress T cells [229,230].
epidermal skin barrier and inflammatory responses, forming a The CsA-cyclophilin complex inhibits the activity of calcineurin
promising target for the treatment of AD [213–215]. In a 3D- phosphatase, which is responsible for translocation and down-
cultured human skin model, the LXR antagonist GSK2033 reduced stream activation of NFAT transcription factors (TFs) [229,230].
the quantity and increased the mean chain length of monounsat- Consequently, the transcription of inflammatory cytokine genes,
urated ceramides and free fatty acids compared with control [214]. IL-2 and IL-4, is blocked and the production of interferon gamma is
Conversely, when compared with control, the LXR agonist inhibited [229–231]. CsA can also bind to steroid-receptor-
T0901317 led to a lipid profile with increased quantities of associated-heat-shock protein 56, preventing the production of
monounsaturated ceramides and free fatty acids followed by a pro-inflammatory cytokines IL-1, IL-3, IL-4, IL-5, IL-6, IL-8, and
reduction in the mean chain length of these lipids more similar to tumor necrosis factor-α [232]. Following administration, CsA
the lipid composition in diseased skin like Netherton syndrome or demonstrates rapid efficacy in patients with AD; however, disease

150
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

reoccurrence may occur within 4 weeks of treatment discontinu- the impaired skin barrier in AD, opening avenues for the
ation [233]. In one study, treatment of AD with CsA has been shown development of additional barrier-related therapies.
to reverse regenerative hyperplasia, indicated by reductions in
epidermal thickness and proliferation biomarkers K16 and Ki67 10. Conclusion
[234]. In patients with AD, serious adverse events are uncommon
as the doses used are generally lower than those in trials for organ Advances in our understanding of the structural components of
transplant [230,235–237]. CsA treatment is contraindicated in the epidermal skin barrier, epidermal lipids and epidermal
patients with renal impairment and in patients with significant differentiation, and in particular identification of FLG loss-of-
uncontrolled hypertension, as well as in those who experience a function mutations, has highlighted the importance of the skin
hypersensitivity reaction to CsA [238]. barrier in AD. Although skin barrier defects have been shown to
Vitamin D deficiency is common in patients with AD and in the play a major role in the pathogenesis of AD, with genetic
general population [239]. The majority of vitamin D is obtained alterations in epidermal barrier genes reported in AD and novel
from UVB exposure to the skin, although some is ingested as part of treatment targets identified, further research is required to fully
a healthy diet [240]. Vitamin D is involved in maintaining the understand the complex etiology of the disease. Together with a
body’s calcium levels, but also in immune modulation [240]. For genetic component, exposure to natural allergens, climate factors,
example, vitamin D aids regulation of AMPs in keratinocytes and possibly air pollutants have been shown to play a role in the
within the epidermis [241]. Reduced levels of AMPs are observed in development of AD, although additional data are required to
patients with AD, which could contribute to the greater number of determine the association between these particulates and AD, and
infections seen in these patients [241,242]. Vitamin D supplemen- underlying mechanisms. In addition, identification of the gut-skin
tation increases the level of AMPs, including cathelicidin and beta 2 axis and recent advances in genetic sequencing technologies will
defensin, in keratinocytes, neutrophils and monocytes [240,243]. likely lead to the improvement of treatment options for AD that
Vitamin D supplementation in patients with AD has shown mixed target the skin and gut microbiome. With the cornerstone of AD
clinical efficacy in terms of clinical improvement and immune treatment consisting of TCS, TCIs and emollients for many years,
response to treatment [189,242]. novel topical and systemic treatments are emerging which target
Dupilumab is a monoclonal antibody developed specifically for the skin barrier. Future research exploring the association between
the management of AD. Dupilumab is approved for the treatment the skin barrier and AD is likely to increase our understanding of
of adults with moderate-to-severe AD, and has recently been the multifaceted etiology of the disorder, opening doors to new and
approved by the US Food and Drug Administration for use in effective treatments.
adolescents (aged 12–17 years) with moderate-to-severe AD [244].
Dupilumab is a direct antagonist of the alpha subunit of both type 1
Funding
and type 2 IL-4 receptors [245]. By targeting the IL-4 receptor α
subunit, dupilumab blocks signaling of both IL-4 and IL-13 [246]
The writing/editorial support was funded by Meda Pharma S.p.
type 2 cytokines involved in pathogenesis of AD. Available data
A., a Viatris company.
demonstrate dupilumab to improve disease severity and reduce
epidermal hyperplasia and inflammation in AD [171,247,248].
Dupilumab has also demonstrated efficacy in improving skin Author’s declaration
barrier integrity [171,172]. In a transcriptomic analysis of skin
biopsy specimens from patients with AD, dupilumab has been The Authors made substantial contributions to conception and
shown to suppress levels of K16 (a marker for hyperplasia) and design, acquisition of data, or analysis and interpretation of data.
increase expression of terminal differentiation proteins filaggrin The Authors drafted the article or reviewed it critically for
and loricrin [171]. In another study, treatment with dupilumab was important intellectual content.
associated with significantly higher levels of filaggrin and loricrin The Authors gave final approval of the version to be published.
mRNA in lesional skin at 16 weeks versus baseline [172]. In the
same study, expression of lipid metabolism, tight junction and Declaration of Competing Interest
membrane channel genes (ELOVL3, FAR2, CLDN8, CLDN23, and
AQP9) was significantly greater in dupilumab-treated patients T. Luger has participated as Principal Investigator in clinical
compared with a placebo group at 16 weeks; levels of epidermal trials, advisory boards and has given lectures, sponsored by
proliferation measures (MKi67, K16, IL24, and IL26) were also Novartis, Lilly, La Roche Posay, Pfizer, Janssen, and Sanofi. He has
significantly lower in patients treated with dupilumab compared received consultancy/speaker honoraria from Novartis, Abbvie,
with placebo [172]. Galderma, La Roche Posay, Meda Pharma S.p.A. (a Viatris company),
Clinical trials with biologics targeting IL-13 (tralokinumab and Janssen, and Sanofi, and has acted as a scientific Advisory Board
lebrikizumab) and IL-31 (nemolizumab) are currently underway. member for Abbvie, Celgene, La Roche Posay, Janssen, Pfizer,
The data are promising, particularly concerning the improvement Menlo, Meda Pharma S.p.A. (a Viatris company), Galderma,
of pruritus [249–251]. In addition, there are several small Symrise, and Lilly. He has received research grants from: Celgene,
molecules including JAK inhibitors, H4 antagonists, PDE4 inhib- Janssen-Cilag, Leo, Meda Pharma S.p.A. (a Viatris company), and
itors and AhR antagonists currently being investigated [252]. Pfizer.
Biomarker studies to gauge treatment response, including with M. Amagai has received research grants from Maruho, Kose, and
phototherapy, CsA, dupilumab and vitamin D, are helping develop JSR.
understanding of the pathophysiology of AD. Research measuring B. Dreno has no relevant conflict of interest to declare.
the effects of systemic treatment on other biophysical parameters M.A. Dagnelie has no relevant conflict of interest to declare.
of barrier permeability such as pH, TEWL, hydration, and lipid W. Liao has received research grant funding from Abbvie,
organization is needed to expand etiological understanding. Amgen, Janssen, Leo Pharma, Novartis, Pfizer, Regeneron, Sanofi,
Further research is also needed regarding the true clinical benefit and TRex Bio.
of barrier restoration and whether systemic treatments that K. Kabashima has received consulting fees or advisory board
improve the skin barrier translate into clinical advantages. Finally, honoraria from Japan tobacco Inc., Chugai Pharmaceutical,
emerging data may allow targeting of specific molecular aspects of Maruho, and Pola Pharma, and has received research grants from

151
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

LEO Pharma, Japan tobacco Inc., P&G Japan, Eli Lilly Japan, Tanabe [12] S. Kezic, G.M. O’Regan, N. Yau, A. Sandilands, H. Chen, L.E. Campbell, K.
Mitsubishi, Ono Pharmaceutical, Kyowa Hakko Kirin, Pola Pharma, Kroboth, R. Watson, M. Rowland, W.H. McLean, A.D. Irvine, Levels of filaggrin
degradation products are influenced by both filaggrin genotype and atopic
AbbVie, Sanofi, and Kyorin Pharmaceutical. dermatitis severity, Allergy 66 (2011) 934–940.
T. Schikowski: No conflict of interest to declare. [13] G.M. O’Regan, A. Sandilands, W.H. McLean, A.D. Irvine, Filaggrin in atopic
E. Proksch has received funding from Bayer Consumer Care and dermatitis, J. Allergy Clin. Immunol. 124 (2009) R2–6.
[14] L. Eckhart, S. Lippens, E. Tschachler, W. Declercq, Cell death by cornification,
Dr. August Wolff, Arzneimittel. Biochim. Biophys. Acta 1833 (2013) 3471–3480.
P.M. Elias: No conflict of interest to declare. [15] M.M. Kelleher, A. Dunn-Galvin, C. Gray, D.M. Murray, M. Kiely, L. Kenny, W.H.I.
M. Simon has received funding from Pierre Fabre Dermo- McLean, A.D. Irvine, J.O. Hourihane, Skin barrier impairment at birth predicts
food allergy at 2 years of age, J. Allergy Clin. Immunol. 137 (2016)1111-1116.
Cosmétique, L’Oréal and BASF Beauty Care Solutions France. He is e8.
co-author of patent #WO2012140095A1. [16] A.D. Irvine, W.H. McLean, D.Y. Leung, Filaggrin mutations associated with
E. Simpson reports grants and personal fees from AbbVie, Eli skin and allergic diseases, N. Engl. J. Med. 365 (2011) 1315–1327.
[17] H. Kawasaki, K. Nagao, A. Kubo, T. Hata, A. Shimizu, H. Mizuno, T. Yamada, M.
Lilly, Incyte, Leo Pharmaceutical, Pfizer and Regeneron. He reports
Amagai, Altered stratum corneum barrier and enhanced percutaneous
grants and fees from Novartis. He reports grants from Kyowa Hakko immune responses in filaggrin-null mice, J. Allergy Clin. Immunol. 129 (2012)
Kirin and Merck and he reports personal fees from Dermira, Forte 1538–1546 e6.
Bio Rx, Menlo Therapeutics, Ortho Dermatologics and Sanofi- [18] Y. Liang, C. Chang, Q. Lu, The genetics and epigenetics of atopic dermatitis-
filaggrin and other polymorphisms, Clin. Rev. Allergy Immunol. 51 (2016)
Genzyme. 315–328.
E.Grinich: No conflict of interest to declare. [19] E. Birben, C. Sackesen, N. Turgutog lu, O. Kalayci, The role of SPINK5 in asthma
M. Schmuth has received research grants from ExpanScience, related physiological events in the airway epithelium, Respir. Med. 106 (2012)
349–355.
has participated as Principal Investigator in trials sponsored by [20] S.E. Ashley, H.T. Tan, P. Vuillermin, S.C. Dharmage, M.L.K. Tang, J. Koplin, L.C.
Roche, Amgen, MSD, GSK, Eli Lilly, Abbvie, Scenesse, Orfagen and Gurrin, A. Lowe, C. Lodge, A.L. Ponsonby, J. Molloy, P. Martin, M.C. Matheson,
has received travel funds from Nogra Pharma. He has not accepted R. Saffery, K.J. Allen, J.A. Ellis, D. Martino, The skin barrier function gene
SPINK5 is associated with challenge-proven IgE-mediated food allergy in
any honoraria from industry for advisory boards or speakers’ infants, Allergy 72 (2017) 1356–1364.
bureau participation and does not own pharma stocks, equity or [21] G. Egawa, K. Kabashima, Barrier dysfunction in the skin allergy, Allergol. Int.
patent licenses. 67 (2018) 3–11.
[22] A. De Benedetto, N.M. Rafaels, L.Y. McGirt, A.I. Ivanov, S.N. Georas, C. Cheadle,
A.E. Berger, K. Zhang, S. Vidyasagar, T. Yoshida, M. Boguniewicz, T. Hata, L.C.
Acknowledgements Schneider, J.M. Hanifin, R.L. Gallo, N. Novak, S. Weidinger, T.H. Beaty, D.Y.
Leung, K.C. Barnes, L.A. Beck, Tight junction defects in patients with atopic
dermatitis, J. Allergy Clin. Immunol. 127 (2011) 773–786 e1-7.
Medical writing support for the development of this review
[23] J.K. Gittler, J.G. Krueger, E. Guttman-Yassky, Atopic dermatitis results in
article, under the direction of the authors, was provided by Jane intrinsic barrier and immune abnormalities: implications for contact
Murphy, Samuel McCracken and Jack Lassados of Ashfield dermatitis, J. Allergy Clin. Immunol. 131 (2013) 300–313.
MedComms, an Ashfield Health company, and funded by Meda [24] P.M. Elias, Skin barrier function, Curr. Allergy Asthma Rep. 8 (2008) 299–305.
[25] R.R. Wickett, M.O. Visscher, Structure and function of the epidermal barrier,
Pharma S.p.A., a Viatris company. Am. J. Infect. Control 34 (2006) S98–S110.
[26] C.L. Simpson, D.M. Patel, K.J. Green, Deconstructing the skin:
References cytoarchitectural determinants of epidermal morphogenesis, Nat. Rev.
Mol. Cell Biol. 12 (2011) 565–580.
[27] E.A. Mauldin, J. Peters-Kennedy, Chapter 6 - integumentary system, sixth
[1] S. Weidinger, N. Novak, Atopic dermatitis, Lancet 387 (2016) 1109–1122. edition, in: M.G. Maxie (Ed.), Jubb, Kennedy & Palmer’S Pathology of
[2] A. Wollenberg, S. Barbarot, T. Bieber, S. Christen-Zaech, M. Deleuran, A. Fink- Domestic Animals, Vol. 1, W.B. Saunders, 2016, pp. 509–736.e1.
Wagner, U. Gieler, G. Girolomoni, S. Lau, A. Muraro, M. Czarnecka-Operacz, T. [28] D.W. Scott, W.H. Miller, Chapter 1 - structure and function of the skin, in: D.W.
Schafer, P. Schmid-Grendelmeier, D. Simon, Z. Szalai, J.C. Szepietowski, A. Scott, W.H. Miller (Eds.), Equine Dermatology, W.B. Saunders, Saint Louis,
Taieb, A. Torrelo, T. Werfel, J. Ring, European Dermatology Forum (EDF), the 2003, pp. 1–58.
European Academy of Dermatology Venereology (EADV), the European [29] F.O. Nestle, P. Di Meglio, J.-Z. Qin, B.J. Nickoloff, Skin immune sentinels in
Academy of Allergy Clinical Immunology (EAACI), the European Task Force on health and disease, Nat. Rev. Immunol. 9 (2009) 679–691.
Atopic Dermatitis (ETFAD), European Federation of Allergy and Airways [30] G.K. Menon, R. Ghadially, M.L. Williams, P.M. Elias, Lamellar bodies as
Diseases Patients’ Associations (EFA), the European Society for Dermatology delivery systems of hydrolytic enzymes: implications for normal and
and Psychiatry (ESDaP), the European Society of Pediatric Dermatology abnormal desquamation, Br. J. Dermatol. 126 (1992) 337–345.
(EPD), Global Allergy and Asthma European Network (GA2LEN), the [31] K.R. Feingold, Lamellar bodies: the key to cutaneous barrier function, J.
European Union of Medical Specialists (UEMS), Consensus-based European Invest. Dermatol. 132 (2012) 1951–1953.
guidelines for treatment of atopic eczema (atopic dermatitis) in adults and [32] F.G. Quiroz, V.F. Fiore, J. Levorse, L. Polak, E. Wong, H.A. Pasolli, E. Fuchs,
children: part I, J. Eur. Acad. Dermatol. Venereol. 32 (2018) 657–682. Liquid-liquid phase separation drives skin barrier formation, Science 367
[3] S. Nutten, Atopic dermatitis: global epidemiology and risk factors, Ann. Nutr. (2020).
Metab. 66 (2015) 8–16. [33] M. Yokouchi, T. Atsugi, M.V. Logtestijn, R.J. Tanaka, M. Kajimura, M. Suematsu,
[4] H. Williams, A. Stewart, E. von Mutius, W. Cookson, H.R. Anderson, Is eczema M. Furuse, M. Amagai, A. Kubo, Epidermal cell turnover across tight junctions
really on the increase worldwide? J. Allergy Clin. Immunol. 121 (2008) 947– based on Kelvin’s tetrakaidecahedron cell shape, Elife 5 (2016)e19593.
954 e15. [34] A. Kubo, I. Ishizaki, A. Kubo, H. Kawasaki, K. Nagao, Y. Ohashi, M. Amagai, The
[5] W. Watson, S. Kapur, Atopic dermatitis, Allergy Asthma Clin. Immunol. 7 stratum corneum comprises three layers with distinct metal-ion barrier
(2011) S4. properties, Sci. Rep. 3 (2013) 1731.
[6] S. Lewis-Jones, Quality of life and childhood atopic dermatitis: the misery of [35] A. Kalinin, L.N. Marekov, P.M. Steinert, Assembly of the epidermal cornified
living with childhood eczema, Int. J. Clin. Pract. 60 (2006) 984–992. cell envelope, J. Cell. Sci. 114 (2001) 3069–3070.
[7] S. Weidinger, L.A. Beck, T. Bieber, K. Kabashima, A.D. Irvine, Atopic dermatitis, [36] K.R. Feingold, P.M. Elias, Role of lipids in the formation and maintenance of
Nat. Rev. Dis. Primers 4 (2018) 1. the cutaneous permeability barrier, Biochim. Biophys. Acta 1841 (2014) 280–
[8] T. Tsakok, R. Woolf, C.H. Smith, S. Weidinger, C. Flohr, Atopic dermatitis: the 294.
skin barrier and beyond, Br. J. Dermatol. 180 (2019) 464–474. [37] J. Van Smeden, M. Janssens, G.S. Gooris, J.A. Bouwstra, The important role of
[9] C.N. Palmer, A.D. Irvine, A. Terron-Kwiatkowski, Y. Zhao, H. Liao, S.P. Lee, D.R. stratum corneum lipids for the cutaneous barrier function, Biochim. Biophys.
Goudie, A. Sandilands, L.E. Campbell, F.J. Smith, G.M. O’Regan, R.M. Watson, J. Acta 1841 (2014) 295–313.
E. Cecil, S.J. Bale, J.G. Compton, J.J. DiGiovanna, P. Fleckman, S. Lewis-Jones, G. [38] D. Crumrine, D. Khnykin, P. Krieg, M.Q. Man, A. Celli, T.M. Mauro, J.S.
Arseculeratne, A. Sergeant, C.S. Munro, B. El Houate, K. McElreavey, L.B. Wakefield, G. Menon, E. Mauldin, J.H. Miner, M.H. Lin, A.R. Brash, E. Sprecher,
Halkjaer, H. Bisgaard, S. Mukhopadhyay, W.H. McLean, Common loss-of- F.P.W. Radner, K. Choate, D. Roop, Y. Uchida, R. Gruber, M. Schmuth, P.M. Elias,
function variants of the epidermal barrier protein filaggrin are a major Mutations in recessive congenital ichthyoses illuminate the origin and
predisposing factor for atopic dermatitis, Nat. Genet. 38 (2006) 441–446. functions of the corneocyte lipid envelope, J. Invest. Dermatol. 139 (2019)
[10] M. Le Lamer, L. Pellerin, M. Reynier, L. Cau, V. Pendaries, C. Leprince, M.C. 760–768.
Mechin, G. Serre, C. Paul, M. Simon, Defects of corneocyte structural proteins [39] M. Janssens, J. van Smeden, G.J. Puppels, A.P. Lavrijsen, P.J. Caspers, J.A.
and epidermal barrier in atopic dermatitis, Biol. Chem. 396 (2015) 1163–1179. Bouwstra, Lipid to protein ratio plays an important role in the skin barrier
[11] A. Sandilands, C. Sutherland, A.D. Irvine, W.H.I. McLean, Filaggrin in the function in patients with atopic eczema, Br. J. Dermatol. 170 (2014) 1248–
frontline: role in skin barrier function and disease, J. Cell. Sci. 122 (2009) 1255.
1285–1294.

152
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

[40] K. Morita, Y. Miyachi, M. Furuse, Tight junctions in epidermis: from barrier to Franke, J.E. Gudjonsson, S. Weidinger, Atopic dermatitis is an IL-13-Dominant
keratinization, Eur. J. Dermatol. 21 (2011) 12–17. disease with greater molecular heterogeneity compared to psoriasis, J. Invest.
[41] A. Kubo, K. Nagao, M. Yokouchi, H. Sasaki, M. Amagai, External antigen uptake Dermatol. 139 (2019) 1480–1489.
by Langerhans cells with reorganization of epidermal tight junction barriers, [66] J.M. Jensen, R. Folster-Holst, A. Baranowsky, M. Schunck, S. Winoto-Morbach,
J. Exp. Med. 206 (2009) 2937–2946. C. Neumann, S. Schutze, E. Proksch, Impaired sphingomyelinase activity and
[42] T. Bieber, Atopic dermatitis, Ann. Dermatol. 22 (2010) 125–137. epidermal differentiation in atopic dermatitis, J. Invest. Dermatol. 122 (2004)
[43] V.N. Sehgal, A. Khurana, V. Mendiratta, D. Saxena, G. Srivastava, A.K. 1423–1431.
Aggarwal, K. Chatterjee, Atopic dermatitis: clinical connotations, especially a [67] U.H. Lee, B.E. Kim, D.J. Kim, Y.G. Cho, Y.M. Ye, D.Y. Leung, Atopic dermatitis is
focus on concomitant atopic undertones in immunocompromised/ associated with reduced corneodesmosin expression: role of cytokine
susceptible genetic and metabolic disorders, Indian J. Dermatol. 61 (2016) modulation and effects on viral penetration, Br. J. Dermatol. 176 (2017) 537–
241–250. 540.
[44] T.C. Scharschmidt, M.Q. Man, Y. Hatano, D. Crumrine, R. Gunathilake, J.P. [68] S. Igawa, M. Kishibe, M. Honma, M. Murakami, Y. Mizuno, Y. Suga, M.
Sundberg, K.A. Silva, T.M. Mauro, M. Hupe, S. Cho, Y. Wu, A. Celli, M. Schmuth, Seishima, Y. Ohguchi, M. Akiyama, K. Hirose, A. Ishida-Yamamoto, H. Iizuka,
K.R. Feingold, P.M. Elias, Filaggrin deficiency confers a paracellular barrier Aberrant distribution patterns of corneodesmosomal components of tape-
abnormality that reduces inflammatory thresholds to irritants and haptens, J. stripped corneocytes in atopic dermatitis and related skin conditions
Allergy Clin. Immunol. 124 (2009) 496–506 e1-6. (ichthyosis vulgaris, Netherton syndrome and peeling skin syndrome type B),
[45] M. Suarez-Farinas, S.J. Tintle, A. Shemer, A. Chiricozzi, K. Nograles, I. J. Dermatol. Sci. 72 (2013) 54–60.
Cardinale, S. Duan, A.M. Bowcock, J.G. Krueger, E. Guttman-Yassky, [69] C.J. Broccardo, S. Mahaffey, J. Schwarz, L. Wruck, G. David, P.M. Schlievert, N.A.
Nonlesional atopic dermatitis skin is characterized by broad terminal Reisdorph, D.Y. Leung, Comparative proteomic profiling of patients with
differentiation defects and variable immune abnormalities, J. Allergy Clin. atopic dermatitis based on history of eczema herpeticum infection and
Immunol. 127 (2011) 954–964 e1-4. Staphylococcus aureus colonization, J. Allergy Clin. Immunol. 127 (2011) 186–
[46] P.M. Elias, Primary role of barrier dysfunction in the pathogenesis of atopic 193 E11.
dermatitis, Exp. Dermatol. 27 (2018) 847–851. [70] C. Elmose, S.F. Thomsen, Twin studies of atopic dermatitis: interpretations
[47] D.Y. Leung, M. Boguniewicz, M.D. Howell, I. Nomura, Q.A. Hamid, New and applications in the filaggrin era, J. Allergy (Cairo) 2015 (2015)902359.
insights into atopic dermatitis, J. Clin. Invest. 113 (2004) 651–657. [71] M. Bradley, C. Söderhäll, H. Luthman, C.-F. Wahlgren, I. Kockum, M.
[48] Q. Hamid, M. Boguniewicz, D.Y. Leung, Differential in situ cytokine gene Nordenskjöld, Susceptibility loci for atopic dermatitis on chromosomes 3,
expression in acute versus chronic atopic dermatitis, J. Clin. Invest. 94 (1994) 13, 15, 17 and 18 in a Swedish population, Hum. Mol. Genet. 11 (2002) 1539–
870–876. 1548.
[49] U. Darsow, U. Raap, S. Stander, Atopic dermatitis, in: E. Carstens, T. Akiyama [72] L. Bin, D.Y.M. Leung, Genetic and epigenetic studies of atopic dermatitis,
(Eds.), Itch: Mechanisms and Treatment, CRC Press/Taylor & Francis, Boca Allergy Asthma Clin. Immunol. 12 (2016) 52.
Raton (FL), 2014. [73] Y.A. Lee, U. Wahn, R. Kehrt, L. Tarani, L. Businco, D. Gustafsson, F. Andersson, A.
[50] M. Fartasch, T.L. Diepgen, O.P. Hornstein, Are hyperlinear palms and dry skin P. Oranje, A. Wolkertstorfer, A. v Berg, U. Hoffmann, W. Kuster, T. Wienker, F.
signs of a concomitant autosomal ichthyosis vulgaris in atopic dermatitis? Ruschendorf, A. Reis, A major susceptibility locus for atopic dermatitis maps
Acta Derm. Venereol. Suppl. (Stockh) 144 (1989) 143–145. to chromosome 3q21, Nat. Genet. 26 (2000) 470–473.
[51] C. Riethmuller, M.A. McAleer, S.A. Koppes, R. Abdayem, J. Franz, M. Haftek, [74] GWAS Catalog, The European Bioinformatics Institute (EMBL-EBI), the
L.E. Campbell, S.F. MacCallum, W.H.I. McLean, A.D. Irvine, S. Kezic, NHGRI-EBI Catalog of Published Genome-wide Association Studies. Atopic
Filaggrin breakdown products determine corneocyte conformation in Dermatitis, (2019) . (Accessed September 2020). https://www.ebi.ac.uk/
patients with atopic dermatitis, J. Allergy Clin. Immunol. 136 (2015) gwas/search?query=atopic%20dermatitis.
1573–1580 e2. [75] M.A. Ferreira, J.M. Vonk, H. Baurecht, I. Marenholz, C. Tian, J.D. Hoffman, Q.
[52] M.J. Cork, S.G. Danby, Y. Vasilopoulos, J. Hadgraft, M.E. Lane, M. Moustafa, R.H. Helmer, A. Tillander, V. Ullemar, J. van Dongen, Y. Lu, F. Ruschendorf, J.
Guy, A.L. Macgowan, R. Tazi-Ahnini, S.J. Ward, Epidermal barrier dysfunction Esparza-Gordillo, C.W. Medway, E. Mountjoy, K. Burrows, O. Hummel, S.
in atopic dermatitis, J. Invest. Dermatol. 129 (2009) 1892–1908. Grosche, B.M. Brumpton, J.S. Witte, J.J. Hottenga, G. Willemsen, J. Zheng, E.
[53] E. Guttman-Yassky, M. Suarez-Farinas, A. Chiricozzi, K.E. Nograles, A. Shemer, Rodriguez, M. Hotze, A. Franke, J.A. Revez, J. Beesley, M.C. Matheson, S.C.
J. Fuentes-Duculan, I. Cardinale, P. Lin, R. Bergman, A.M. Bowcock, J.G. Dharmage, L.M. Bain, L.G. Fritsche, M.E. Gabrielsen, B. Balliu, J.B. Nielsen, W.
Krueger, Broad defects in epidermal cornification in atopic dermatitis Zhou, K. Hveem, A. Langhammer, O.L. Holmen, M. Loset, G.R. Abecasis, C.J.
identified through genomic analysis, J. Allergy Clin. Immunol. 124 (2009) Willer, A. Arnold, G. Homuth, C.O. Schmidt, P.J. Thompson, N.G. Martin, D.L.
1235–1244 e58. Duffy, N. Novak, H. Schulz, S. Karrasch, C. Gieger, K. Strauch, R.B. Melles, D.A.
[54] P.M. Elias, J.S. Wakefield, Mechanisms of abnormal lamellar body secretion Hinds, N. Hubner, S. Weidinger, P.K.E. Magnusson, R. Jansen, E. Jorgenson, Y.A.
and the dysfunctional skin barrier in patients with atopic dermatitis, J. Lee, D.I. Boomsma, C. Almqvist, R. Karlsson, G.H. Koppelman, L. Paternoster,
Allergy Clin. Immunol. 134 (2014) 781–791 e1. Shared genetic origin of asthma, hay fever and eczema elucidates allergic
[55] M. Danso, W. Boiten, V. van Drongelen, K. Gmelig Meijling, G. Gooris, A. El disease biology, Nat. Genet. 49 (2017) 1752–1757.
Ghalbzouri, S. Absalah, R. Vreeken, S. Kezic, J. van Smeden, S. Lavrijsen, J. [76] E.L. Simpson, J.R. Chalmers, J.M. Hanifin, K.S. Thomas, M.J. Cork, W.H. McLean,
Bouwstra, Altered expression of epidermal lipid bio-synthesis enzymes in S.J. Brown, Z. Chen, Y. Chen, H.C. Williams, Emollient enhancement of the skin
atopic dermatitis skin is accompanied by changes in stratum corneum lipid barrier from birth offers effective atopic dermatitis prevention, J. Allergy Clin.
composition, J. Dermatol. Sci. 88 (2017) 57–66. Immunol. 134 (2014) 818–823.
[56] S. Imayama, Y. Shibata, Y. Hori, Epidermal mast cells in atopic dermatitis, [77] J.R. Chalmers, R.H. Haines, L.E. Bradshaw, A.A. Montgomery, K.S. Thomas, S.J.
Lancet 346 (1995) 1559. Brown, M.J. Ridd, S. Lawton, E.L. Simpson, M.J. Cork, T.H. Sach, C. Flohr, E.J.
[57] D.A. Groneberg, C. Bester, A. Grutzkau, F. Serowka, A. Fischer, B.M. Henz, P. Mitchell, R. Swinden, S. Tarr, S. Davies-Jones, N. Jay, M.M. Kelleher, M.R.
Welker, Mast cells and vasculature in atopic dermatitis – potential stimulus Perkin, R.J. Boyle, H.C. Williams, Daily emollient during infancy for
of neoangiogenesis, Allergy 60 (2005) 90–97. prevention of eczema: the BEEP randomised controlled trial, Lancet 395
[58] K. Yoshida, A. Kubo, H. Fujita, M. Yokouchi, K. Ishii, H. Kawasaki, T. Nomura, H. (2020) 962–972.
Shimizu, K. Kouyama, T. Ebihara, K. Nagao, M. Amagai, Distinct behavior of [78] P. Fortugno, L. Furio, M. Teson, M. Berretti, M. El Hachem, G. Zambruno, A.
human Langerhans cells and inflammatory dendritic epidermal cells at tight Hovnanian, M. D’Alessio, The 420K LEKTI variant alters LEKTI proteolytic
junctions in patients with atopic dermatitis, J. Allergy Clin. Immunol. 134 activation and results in protease deregulation: implications for atopic
(2014) 856–864. dermatitis, Hum. Mol. Genet. 21 (2012) 4187–4200.
[59] D.J. Panther, S.E. Jacob, The importance of acidification in atopic eczema: an [79] S. Kezic, A. Kammeyer, F. Calkoen, J.W. Fluhr, J.D. Bos, Natural moisturizing
underexplored avenue for treatment, J. Clin. Med. 4 (2015) 970–978. factor components in the stratum corneum as biomarkers of filaggrin
[60] M.H. Schmid-Wendtner, H.C. Korting, The pH of the skin surface and its genotype: evaluation of minimally invasive methods, Br. J. Dermatol. 161
impact on the barrier function, Skin Pharmacol. Physiol. 19 (2006) 296–302. (2009) 1098–1104.
[61] S. Seidenari, G. Giusti, Objective assessment of the skin of children affected by [80] I. Marenholz, V.A. Rivera, J. Esparza-Gordillo, A. Bauerfeind, M.A. Lee-Kirsch,
atopic dermatitis: a study of pH, capacitance and TEWL in eczematous and A. Ciechanowicz, M. Kurek, T. Piskackova, M. Macek, Y.A. Lee, Association
clinically uninvolved skin, Acta Derm. Venereol. 75 (1995) 429–433. screening in the Epidermal Differentiation Complex (EDC) identifies an
[62] D. Ghosh, L. Ding, U. Sivaprasad, E. Geh, J. Biagini Myers, J.A. Bernstein, G.K. SPRR3 repeat number variant as a risk factor for eczema, J. Invest. Dermatol.
Khurana Hershey, T.B. Mersha, multiple transcriptome data analysis reveals 131 (2011) 1644–1649.
biologically relevant atopic dermatitis signature genes and pathways, PLoS [81] D.J. Margolis, J. Gupta, A.J. Apter, O. Hoffstad, M. Papadopoulos, T.R. Rebbeck,
One 10 (2015)e0144316. B. Wubbenhorst, N. Mitra, Exome sequencing of filaggrin and related genes in
[63] B.E. Kim, D.Y.M. Leung, M. Boguniewicz, M.D. Howell, Loricrin and involucrin African-American children with atopic dermatitis, J. Invest. Dermatol. 134
expression is down-regulated by Th2 cytokines through STAT-6, Clin. (2014) 2272–2274.
Immunol. 126 (2008) 332–337. [82] L. Paternoster, M. Standl, J. Waage, H. Baurecht, M. Hotze, D.P. Strachan, J.A.
[64] L. Pellerin, J. Henry, C.Y. Hsu, S. Balica, C. Jean-Decoster, M.C. Méchin, B. Curtin, K. Bonnelykke, C. Tian, A. Takahashi, J. Esparza-Gordillo, A.C. Alves, J.P.
Hansmann, E. Rodriguez, S. Weindinger, A.M. Schmitt, G. Serre, C. Paul, M. Thyssen, H.T. den Dekker, M.A. Ferreira, E. Altmaier, P.M. Sleiman, F.L. Xiao, J.
Simon, Defects of filaggrin-like proteins in both lesional and nonlesional R. Gonzalez, I. Marenholz, B. Kalb, M.P. Yanes, C.J. Xu, L. Carstensen, M.M.
atopic skin, J. Allergy Clin. Immunol. 131 (2013) 1094–1102. Groen-Blokhuis, C. Venturini, C.E. Pennell, S.J. Barton, A.M. Levin, I. Curjuric,
[65] L.C. Tsoi, E. Rodriguez, F. Degenhardt, H. Baurecht, U. Wehkamp, N. Volks, S. M. Bustamante, E. Kreiner-Moller, G.A. Lockett, J. Bacelis, S. Bunyavanich, R.A.
Szymczak, W.R. Swindell, M.K. Sarkar, K. Raja, S. Shao, M. Patrick, Y. Gao, R. Myers, A. Matanovic, A. Kumar, J.Y. Tung, T. Hirota, M. Kubo, W.L. McArdle, A.J.
Uppala, B.E. Perez White, S. Getsios, P.W. Harms, E. Maverakis, J.T. Elder, A. Henderson, J.P. Kemp, J. Zheng, G.D. Smith, F. Ruschendorf, A. Bauerfeind, M.

153
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

A. Lee-Kirsch, A. Arnold, G. Homuth, C.O. Schmidt, E. Mangold, S. Cichon, T. [101] W. Liu, J. Cai, C. Huang, Y. Hu, Q. Fu, Z. Zou, C. Sun, L. Shen, X. Wang, J. Pan, Y.
Keil, E. Rodriguez, A. Peters, A. Franke, W. Lieb, N. Novak, R. Folster-Holst, M. Huang, J. Chang, Z. Zhao, Y. Sun, J. Sundell, Associations of gestational and
Horikoshi, J. Pekkanen, S. Sebert, L.L. Husemoen, N. Grarup, J.C. de Jongste, F. early life exposures to ambient air pollution with childhood atopic eczema in
Rivadeneira, A. Hofman, V.W. Jaddoe, S.G. Pasmans, N.J. Elbert, A.G. Shanghai, China, Sci. Total Environ. 572 (2016) 34–42.
Uitterlinden, G.B. Marks, P.J. Thompson, M.C. Matheson, C.F. Robertson, J.S. [102] C.C. Huang, H.J. Wen, P.C. Chen, T.L. Chiang, S.J. Lin, Y.L. Guo, Prenatal air
Ried, J. Li, X.B. Zuo, X.D. Zheng, X.Y. Yin, L.D. Sun, M.A. McAleer, G.M. O’Regan, pollutant exposure and occurrence of atopic dermatitis, Br. J. Dermatol. 173
C.M. Fahy, L.E. Campbell, M. Macek, M. Kurek, D. Hu, C. Eng, D.S. Postma, B. (2015) 981–988.
Feenstra, F. Geller, J.J. Hottenga, C.M. Middeldorp, P. Hysi, V. Bataille, T. [103] Y.M. Kim, J. Kim, Y. Han, B.H. Jeon, H.K. Cheong, K. Ahn, Short-term effects of
Spector, C.M. Tiesler, E. Thiering, B. Pahukasahasram, J.J. Yang, M. Imboden, S. weather and air pollution on atopic dermatitis symptoms in children: a panel
Huntsman, N. Vilor-Tejedor, C.L. Relton, R. Myhre, W. Nystad, A. Custovic, S.T. study in Korea, PLoS One 12 (2017)e0175229.
Weiss, D.A. Meyers, C. Soderhall, E. Melen, C. Ober, B.A. Raby, A. Simpson, B. [104] A. Li, L. Fan, L. Xie, Y. Ren, L. Li, Associations between air pollution, climate
Jacobsson, J.W. Holloway, H. Bisgaard, J. Sunyer, N.M.P. Hensch, L.K. Williams, factors and outpatient visits for eczema in West China Hospital, Chengdu,
K.M. Godfrey, C.A. Wang, D.I. Boomsma, M. Melbye, G.H. Koppelman, D. Jarvis, south-western China: a time series analysis, J. Eur. Acad. Dermatol. Venereol.
W.I. McLean, A.D. Irvine, X.J. Zhang, H. Hakonarson, C. Gieger, E.G. Burchard, 32 (2018) 486–494.
N.G. Martin, L. Duijts, A. Linneberg, M.R. Jarvelin, M.M. Noethen, S. Lau, N. [105] Y.M. Kim, J. Kim, K. Jung, S. Eo, K. Ahn, The effects of particulate matter on
Hubner, Y.A. Lee, M. Tamari, D.A. Hinds, D. Glass, S.J. Brown, J. Heinrich, D.M. atopic dermatitis symptoms are influenced by weather type: application of
Evans, S. Weidinger, Multi-ancestry genome-wide association study of spatial synoptic classification (SSC), Int. J. Hyg. Environ. Health 221 (2018)
21,000 cases and 95,000 controls identifies new risk loci for atopic 823–829.
dermatitis, Nat. Genet. 47 (2015) 1449–1456. [106] Q. Li, Y. Yang, R. Chen, H. Kan, W. Song, J. Tan, F. Xu, J. Xu, Ambient air
[83] T. Sasaki, A. Shiohama, A. Kubo, H. Kawasaki, A. Ishida-Yamamoto, T. Yamada, pollution, meteorological factors and outpatient visits for eczema in
T. Hachiya, A. Shimizu, H. Okano, J. Kudoh, M. Amagai, A homozygous Shanghai, China: a time-series analysis, Int. J. Environ. Res. Public Health 13
nonsense mutation in the gene for Tmem79, a component for the lamellar (2016) 1106.
granule secretory system, produces spontaneous eczema in an experimental [107] T. Hidaka, E. Ogawa, E.H. Kobayashi, T. Suzuki, R. Funayama, T. Nagashima, T.
model of atopic dermatitis, J. Allergy Clin. Immunol. 132 (2013) 1111–1120 e4. Fujimura, S. Aiba, K. Nakayama, R. Okuyama, M. Yamamoto, The aryl
[84] S.P. Saunders, C.S.M. Goh, S.J. Brown, C.N.A. Palmer, R.M. Porter, C. Cole, L.E. hydrocarbon receptor AhR links atopic dermatitis and air pollution via
Campbell, M. Gierlinski, G.J. Barton, G. Schneider, A. Balmain, A.R. Prescott, S. induction of the neurotrophic factor artemin, Nat. Immunol. 18 (2017) 64–73.
Weidinger, H. Baurecht, M. Kabesch, C. Gieger, Y.-A. Lee, R. Tavendale, S. [108] A. Elentner, D. Ortner, B. Clausen, F.J. Gonzalez, P.M. Fernandez-Salguero, M.
Mukhopadhyay, S.W. Turner, V.B. Madhok, F.M. Sullivan, C. Relton, J. Burn, S. Schmuth, S. Dubrac, Skin response to a carcinogen involves the xenobiotic
Meggitt, C.H. Smith, M.A. Allen, J.N.W.N. Barker, N.J. Reynolds, H.J. Cordell, A. receptor pregnane X receptor, Exp. Dermatol. 24 (2015) 835–840.
D. Irvine, W.H.I. McLean, A. Sandilands, P.G. Fallon, Tmem79/Matt is the [109] A. Elentner, M. Schmuth, N. Yannoutsos, T.O. Eichmann, R. Gruber, F.P.W.
matted mouse gene and is a predisposing gene for atopic dermatitis in Radner, M. Hermann, B. Del Frari, S. Dubrac, Epidermal Overexpression of
human subjects, J. Allergy Clin. Immunol. 132 (2013) 1121–1129. Xenobiotic Receptor PXR Impairs the Epidermal Barrier and Triggers Th2
[85] A. Teng, M. Nair, J. Wells, J.A. Segre, X. Dai, Strain-dependent perinatal Immune Response, J. Invest. Dermatol. 138 (2018) 109–120.
lethality of Ovol1-deficient mice and identification of Ovol2 as a downstream [110] J.M. Jensen, K. Ahrens, J. Meingassner, A. Scherer, M. Brautigam, A. Stutz, T.
target of Ovol1 in skin epidermis, Biochim. Biophys. Acta 1772 (2007) 89–95. Schwarz, R. Folster-Holst, J. Harder, R. Glaser, E. Proksch, Differential
[86] A. Torrelo, Atopic dermatitis in different skin types. What is to know? J. Eur. suppression of epidermal antimicrobial protein expression in atopic
Acad. Dermatol. Venereol. 28 (Suppl 3) (2014) 2–4. dermatitis and in EFAD mice by pimecrolimus compared to corticosteroids,
[87] G. Tsuji, T. Ito, T. Chiba, C. Mitoma, T. Nakahara, H. Uchi, M. Furue, The role of Exp. Dermatol. 20 (2011) 783–788.
the OVOL1-OVOL2 axis in normal and diseased human skin, J. Dermatol. Sci. [111] J.M. Schroder, Antimicrobial peptides in healthy skin and atopic dermatitis,
90 (2018) 227–231. Allergol. Int. 60 (2011) 17–24.
[88] H.S. Yu, M.J. Kang, J.W. Kwon, S.Y. Lee, E. Lee, S.I. Yang, Y.H. Jung, K. Hong, Y.J. [112] P.Y. Ong, T. Ohtake, C. Brandt, I. Strickland, M. Boguniewicz, T. Ganz, R.L. Gallo,
Kim, S.H. Lee, H.J. Kim, H.Y. Kim, J.H. Seo, B.J. Kim, H.B. Kim, S.J. Hong, Claudin- D.Y.M. Leung, Endogenous antimicrobial peptides and skin infections in
1 polymorphism modifies the effect of mold exposure on the development of atopic dermatitis, N. Engl. J. Med. 347 (2002) 1151–1160.
atopic dermatitis and production of IgE, J. Allergy Clin. Immunol. 135 (2015) [113] I. Nomura, E. Goleva, M.D. Howell, Q.A. Hamid, P.Y. Ong, C.F. Hall, M.A. Darst,
827–830 e5. B. Gao, M. Boguniewicz, J.B. Travers, D.Y. Leung, Cytokine milieu of atopic
[89] D.J. Margolis, J. Gupta, A.J. Apter, T. Ganguly, O. Hoffstad, M. Papadopoulos, T. dermatitis, as compared to psoriasis, skin prevents induction of innate
R. Rebbeck, N. Mitra, Filaggrin-2 variation is associated with more persistent immune response genes, J. Immunol. 171 (2003) 3262–3269.
atopic dermatitis in African American subjects, J. Allergy Clin. Immunol. 133 [114] J. Harder, S. Dressel, M. Wittersheim, J. Cordes, U. Meyer-Hoffert, U. Mrowietz,
(2014) 784–789. R. Fölster-Holst, E. Proksch, J.M. Schröder, T. Schwarz, R. Gläser, Enhanced
[90] B. Lee, A. Villarreal-Ponce, M. Fallahi, J. Ovadia, P. Sun, Q.C. Yu, S. Ito, S. Sinha, expression and secretion of antimicrobial peptides in atopic dermatitis and
Q. Nie, X. Dai, Transcriptional mechanisms link epithelial plasticity to after superficial skin injury, J. Invest. Dermatol. 130 (2010) 1355–1364.
adhesion and differentiation of epidermal progenitor cells, Dev. Cell 29 [115] A.L. Byrd, Y. Belkaid, J.A. Segre, The human skin microbiome, Nat. Rev.
(2014) 47–58. Microbiol. 16 (2018) 143–155.
[91] S. Asad, M.C. Winge, C.F. Wahlgren, K.D. Bilcha, M. Nordenskjöld, F. Taylan, M. [116] E.A. Grice, J.A. Segre, The skin microbiome, Nat. Rev. Microbiol. 9 (2011) 244–
Bradley, The tight junction gene Claudin-1 is associated with atopic 253.
dermatitis among Ethiopians, J. Eur. Acad. Dermatol. Venereol. 30 (2016) [117] K. Findley, E.A. Grice, The skin microbiome: a focus on pathogens and their
1939–1941. association with skin disease, PLoS Pathog. 10 (2014) e1004436-e1004436.
[92] S.G. Hostetler, B. Kaffenberger, T. Hostetler, M.J. Zirwas, The role of airborne [118] R.L. Gallo, T. Nakatsuji, Microbial symbiosis with the innate immune defense
proteins in atopic dermatitis, J. Clin. Aesthet. Dermatol. 3 (2010) 22–31. system of the skin, J. Invest. Dermatol. 131 (2011) 1974–1980.
[93] G. Stajminger, S. Marinovic-Kulisic, J. Lipozencic, Z. Pastar, Most common [119] Y. Yamazaki, Y. Nakamura, G. Nunez, Role of the microbiota in skin immunity
inhalant allergens in atopic dermatitis, atopic dermatitis/allergic rhinitis, and and atopic dermatitis, Allergol. Int. 66 (2017) 539–544.
atopic dermatitis/bronchial asthma patients: a five-year retrospective study, [120] Y.E. Chen, M.A. Fischbach, Y. Belkaid, Skin microbiota-host interactions,
Acta Dermatovenerol. Croat. 15 (2007) 130–134. Nature 553 (2018) 427–436.
[94] N. Serhan, L. Basso, R. Sibilano, C. Petitfils, J. Meixiong, C. Bonnart, L.L. Reber, T. [121] M. Egert, R. Simmering, C.U. Riedel, The association of the skin microbiota
Marichal, P. Starkl, N. Cenac, X. Dong, M. Tsai, S.J. Galli, N. Gaudenzio, House with health, immunity, and disease, Clin. Pharmacol. Ther. 102 (2017) 62–69.
dust mites activate nociceptor–mast cell clusters to drive type 2 skin [122] M.A. Malla, A. Dubey, A. Kumar, S. Yadav, A. Hashem, E.F. Abd_Allah, Exploring
inflammation, Nat. Immunol. 20 (2019) 1435–1443. the human microbiome: the potential future role of next-generation
[95] R. Kantor, J.I. Silverberg, Environmental risk factors and their role in the sequencing in disease diagnosis and treatment, Front. Immunol. 9 (2019)
management of atopic dermatitis, Expert Rev. Clin. Immunol. 13 (2017) 15– 2868.
26. [123] L.V. Hooper, D.R. Littman, A.J. Macpherson, Interactions between the
[96] L.T.N. Ngoc, D. Park, Y. Lee, Y.C. Lee, Systematic review and meta-analysis of microbiota and the immune system, Science 336 (2012) 1268–1273.
human skin diseases due to particulate matter, Int. J. Environ. Res. Public [124] J.S. Meisel, G. Sfyroera, C. Bartow-McKenney, C. Gimblet, J. Bugayev, J.
Health 14 (2017) 1458. Horwinski, B. Kim, J.R. Brestoff, A.S. Tyldsley, Q. Zheng, B.P. Hodkinson, D.
[97] U. Kramer, H. Behrendt, [Air pollution and atopic eczema : systematic review Artis, E.A. Grice, Commensal microbiota modulate gene expression in the
of findings from environmental epidemiological studies], Hautarzt 70 (2019) skin, Microbiome 6 (2018) 20.
169–184. [125] S. Igawa, A. Di Nardo, Skin microbiome and mast cells, Transl. Res. 184 (2017)
[98] J.-Y. Lee, D.K. Lamichhane, M. Lee, S. Ye, J.-H. Kwon, M.-S. Park, H.-C. Kim, J.-H. 68–76.
Leem, Y.-C. Hong, Y. Kim, M. Ha, E. Ha, Preventive effect of residential green [126] S. Eyerich, K. Eyerich, C. Traidl-Hoffmann, T. Biedermann, Cutaneous barriers
space on infantile atopic dermatitis associated with prenatal air pollution and skin immunity: differentiating a connected network, Trends Immunol.
exposure, Int. J. Environ. Res. Public Health 15 (2018) 102. 39 (2018) 315–327.
[99] C. Lu, L. Deng, C. Ou, H. Yuan, X. Chen, Q. Deng, Preconceptional and perinatal [127] A. SanMiguel, E.A. Grice, Interactions between host factors and the skin
exposure to traffic-related air pollution and eczema in preschool children, J. microbiome, Cell. Mol. Life Sci. 72 (2015) 1499–1515.
Dermatol. Sci. 85 (2017) 85–95. [128] H.D. de Koning, D. Rodijk-Olthuis, I.M. van Vlijmen-Willems, L.A. Joosten, M.
[100] Q. Deng, C. Lu, Y. Li, J. Sundell, N. Dan, Exposure to outdoor air pollution G. Netea, J. Schalkwijk, P.L. Zeeuwen, A comprehensive analysis of pattern
during trimesters of pregnancy and childhood asthma, allergic rhinitis, and recognition receptors in normal and inflamed human epidermis:
eczema, Environ. Res. 150 (2016) 119–127.

154
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

upregulation of dectin-1 in psoriasis, J. Invest. Dermatol. 130 (2010) 2611– [157] J.M. Jensen, S. Pfeiffer, M. Witt, M. Brautigam, C. Neumann, M. Weichenthal, T.
2620. Schwarz, R. Folster-Holst, E. Proksch, Different effects of pimecrolimus and
[129] T.H. Mogensen, Pathogen recognition and inflammatory signaling in innate betamethasone on the skin barrier in patients with atopic dermatitis, J.
immune defenses, Clin. Microbiol. Rev. 22 (2009) 240–273. Allergy Clin. Immunol. 124 (2009) R19–28.
[130] T. Bieber, Atopic dermatitis, N. Engl. J. Med. 358 (2008) 1483–1494. [158] M. Catherine Mack Correa, J. Nebus, Management of patients with atopic
[131] W. Francuzik, K. Franke, R.R. Schumann, G. Heine, M. Worm, dermatitis: the role of emollient therapy, Dermatol. Res. Pract. 2012 (2012)
Propionibacterium acnes abundance correlates inversely with 836931.
Staphylococcus aureus: data from atopic dermatitis skin microbiome, Acta [159] A. Wollenberg, S. Reitamo, G. Girolomoni, M. Lahfa, T. Ruzicka, E. Healy, A.
Derm. Venereol. 98 (2018) 490–495. Giannetti, T. Bieber, J. Vyas, M. Deleuran, Proactive treatment of atopic
[132] E.A. Grice, The skin microbiome: potential for novel diagnostic and dermatitis in adults with 0.1% tacrolimus ointment, Allergy 63 (2008) 742–
therapeutic approaches to cutaneous disease, Semin. Cutan. Med. Surg. 33 750.
(2014) 98–103. [160] M. Ruer-Mulard, W. Aberer, A. Gunstone, O.M. Kekki, J.L. Lopez Estebaranz, A.
[133] H. Xu, H. Li, Acne, the skin microbiome, and antibiotic treatment, Am. J. Clin. Vertruyen, A. Guettner, T. Hultsch, Twice-daily versus once-daily applications
Dermatol. 20 (2019) 335–344. of pimecrolimus cream 1% for the prevention of disease relapse in pediatric
[134] W.-M. Wang, H.-Z. Jin, Skin microbiome: an actor in the pathogenesis of patients with atopic dermatitis, Pediatr. Dermatol. 26 (2009) 551–558.
psoriasis, Chin. Med. J. 131 (2018) 95–98. [161] P.M. Elias, Lipid abnormalities and lipid-based repair strategies in atopic
[135] T. Kobayashi, M. Glatz, K. Horiuchi, H. Kawasaki, H. Akiyama, D.H. Kaplan, H.H. dermatitis, Biochim. Biophys. Acta 1841 (2014) 323–330.
Kong, M. Amagai, K. Nagao, Dysbiosis and Staphylococcus aureus [162] E. Proksch, M. Soeberdt, C. Neumann, A. Kilic, C. Abels, Modulators of the
colonization drives inflammation in atopic dermatitis, Immunity 42 (2015) endocannabinoid system influence skin barrier repair, epidermal
756–766. proliferation, differentiation and inflammation in a mouse model, Exp.
[136] M.A. Dagnelie, S. Corvec, M. Saint-Jean, V. Bourdes, J.M. Nguyen, A. Dermatol. 28 (2019) 1058–1065.
Khammari, B. Dreno, Decrease in diversity of Propionibacterium acnes [163] B.G. Jo, N.J. Park, J. Jegal, S. Choi, S.W. Lee, H. Jin, S.N. Kim, M.H. Yang, A new
Phylotypes in patients with severe acne on the back, Acta Derm. Venereol. 98 flavonoid from Stellera chamaejasme L., stechamone, alleviated 2,4-
(2018) 262–267. dinitrochlorobenzene-induced atopic dermatitis-like skin lesions in a
[137] M. Notay, N. Foolad, A.R. Vaughn, R.K. Sivamani, Probiotics, prebiotics, and murine model, Int. Immunopharmacol. 59 (2018) 113–119.
synbiotics for the treatment and prevention of adult dermatological diseases, [164] L.H. Kircik, J.Q. Del Rosso, D. Aversa, Evaluating clinical use of a ceramide-
Am. J. Clin. Dermatol. 18 (2017) 721–732. dominant, physiologic lipid-based topical emulsion for atopic dermatitis, J.
[138] Y. Yu, J. Champer, H. Garban, J. Kim, Typing of Propionibacterium acnes: a Clin. Aesthet. Dermatol. 4 (2011) 34–40.
review of methods and comparative analysis, Br. J. Dermatol. 172 (2015) [165] S.G. Danby, P.V. Andrew, K. Brown, J. Chittock, L.J. Kay, M.J. Cork, An
1204–1209. investigation of the skin barrier restoring effects of a cream and lotion
[139] F.H. Al-Ghazzewi, R.F. Tester, Impact of prebiotics and probiotics on skin containing ceramides in a multi-vesicular emulsion in people with dry,
health, Benef. Microbes 5 (2014) 99–107. eczema-prone, skin: the RESTORE study phase 1, Dermatol. Ther. (Heidelb.)
[140] J. Zeichner, S. Seite, From probiotic to prebiotic using thermal spring water, J. 10 (2020).
Drugs Dermatol. 17 (2018) 657–662. [166] M. Schmuth, K.R. Feingold, P.M. Elias, Stress test of the skin: the cutaneous
[141] R. Huang, H. Ning, M. Shen, J. Li, J. Zhang, X. Chen, Probiotics for the treatment permeability barrier treadmill, Exp. Dermatol. 29 (2020) 112–113.
of atopic dermatitis in children: a systematic review and meta-analysis of [167] K.R. Feingold, M.Q. Man, G.K. Menon, S.S. Cho, B.E. Brown, P.M. Elias,
randomized controlled trials, Front. Cell. Infect. Microbiol. 7 (2017) 392. Cholesterol synthesis is required for cutaneous barrier function in mice, J.
[142] S. Seite, H. Zelenkova, R. Martin, Clinical efficacy of emollients in atopic Clin. Invest. 86 (1990) 1738–1745.
dermatitis patients - relationship with the skin microbiota modification, Clin. [168] K.R. Feingold, M.Q. Man, E. Proksch, G.K. Menon, B.E. Brown, P.M. Elias, The
Cosmet. Investig. Dermatol. 10 (2017) 25–33. lovastatin-treated rodent: a new model of barrier disruption and epidermal
[143] S. Seite, G.E. Flores, J.B. Henley, R. Martin, H. Zelenkova, L. Aguilar, N. Fierer, hyperplasia, J. Invest. Dermatol. 96 (1991) 201–209.
Microbiome of affected and unaffected skin of patients with atopic [169] J.M. Jensen, M. Weppner, S. Dähnhardt-Pfeiffer, C. Neumann, M. Bräutigam, T.
dermatitis before and after emollient treatment, J. Drugs Dermatol. 13 (2014) Schwarz, R. Fölster-Holst, E. Proksch, Effects of pimecrolimus compared with
1365–1372. triamcinolone acetonide cream on skin barrier structure in atopic dermatitis:
[144] A.V. Gannesen, V. Borrel, L. Lefeuvre, A.I. Netrusov, V.K. Plakunov, M.G.J. a randomized, double-blind, right-left arm trial, Acta Derm. Venereol. 93
Feuilloley, Effect of two cosmetic compounds on the growth, biofilm (2013) 515–519.
formation activity, and surface properties of acneic strains of Cutibacterium [170] C. Bußmann, N. Novak, Systemic therapy of atopic dermatitis, Allergol. Sel. 1
acnes and Staphylococcus aureus, Microbiologyopen 8 (2019)e00659. (2017) 1–8.
[145] B. Dreno, R. Martin, D. Moyal, J.B. Henley, A. Khammari, S. Seite, Skin [171] J.D. Hamilton, M. Suarez-Farinas, N. Dhingra, I. Cardinale, X. Li, A. Kostic, J.E.
microbiome and acne vulgaris: staphylococcus, a new actor in acne, Exp. Ming, A.R. Radin, J.G. Krueger, N. Graham, G.D. Yancopoulos, G. Pirozzi, E.
Dermatol. 26 (2017) 798–803. Guttman-Yassky, Dupilumab improves the molecular signature in skin of
[146] M. Glatz, J.H. Jo, E.A. Kennedy, E.C. Polley, J.A. Segre, E.L. Simpson, H.H. Kong, patients with moderate-to-severe atopic dermatitis, J. Allergy Clin. Immunol.
Emollient use alters skin barrier and microbes in infants at risk for 134 (2014) 1293–1300.
developing atopic dermatitis, PLoS One 13 (2018)e0192443. [172] E. Guttman-Yassky, R. Bissonnette, B. Ungar, M. Suárez-Fariñas, M. Ardeleanu,
[147] M. Deleuran, V. Georgescu, C. Jean-Decoster, An emollient containing H. Esaki, M. Suprun, Y. Estrada, H. Xu, X. Peng, J.I. Silverberg, A. Menter, J.G.
aquaphilus dolomiae extract is effective in the management of Xerosis and Krueger, R. Zhang, U. Chaudhry, B. Swanson, N.M.H. Graham, G. Pirozzi, G.D.
pruritus: an international, real-world study, Dermatol. Ther. (Heidelb.) 10 Yancopoulos, J.D.D. Hamilton, Dupilumab progressively improves systemic
(2020) 1013–1029. and cutaneous abnormalities in patients with atopic dermatitis, J. Allergy
[148] É. Butler, C. Lundqvist, J. Axelsson, Lactobacillus reuteri DSM 17938 as a novel Clin. Immunol. 143 (2019) 155–172.
topical cosmetic ingredient: a proof of concept clinical study in adults with [173] J.A. Bouwstra, M. Ponec, The skin barrier in healthy and diseased state,
atopic dermatitis, Microorganisms 8 (2020) 1026. Biochim. Biophys. Acta Biomembr. 1758 (2006) 2080–2095.
[149] I.A. Myles, N.J. Earland, E.D. Anderson, I.N. Moore, M.D. Kieh, K.W. Williams, A. [174] P.M. Elias, M. Schmuth, Abnormal skin barrier in the etiopathogenesis of
Saleem, N.M. Fontecilla, P.A. Welch, D.A. Darnell, L.A. Barnhart, A.A. Sun, G. atopic dermatitis, Curr. Allergy Asthma Rep. 9 (2009) 265–272.
Uzel, S.K. Datta, First-in-human topical microbiome transplantation with [175] U. Nygaard, M. Deleuran, C. Vestergaard, Emerging treatment options in
Roseomonas mucosa for atopic dermatitis, JCI Insight 3 (2018)e120608. atopic dermatitis: topical therapies, Dermatology 233 (2017) 333–343.
[150] T. Nakatsuji, T.H. Chen, S. Narala, K.A. Chun, A.M. Two, T. Yun, F. Shafiq, P.F. [176] L.F. Eichenfield, W.L. Tom, T.G. Berger, A. Krol, A.S. Paller, K. Schwarzenberger,
Kotol, A. Bouslimani, A.V. Melnik, H. Latif, J.N. Kim, A. Lockhart, K. Artis, G. J.N. Bergman, S.L. Chamlin, D.E. Cohen, K.D. Cooper, K.M. Cordoro, D.M. Davis,
David, P. Taylor, J. Streib, P.C. Dorrestein, A. Grier, S.R. Gill, K. Zengler, T.R. Hata, S.R. Feldman, J.M. Hanifin, D.J. Margolis, R.A. Silverman, E.L. Simpson, H.C.
D.Y. Leung, R.L. Gallo, Antimicrobials from human skin commensal bacteria Williams, C.A. Elmets, J. Block, C.G. Harrod, W. Smith Begolka, R. Sidbury,
protect against Staphylococcus aureus and are deficient in atopic dermatitis, Guidelines of care for the management of atopic dermatitis: section 2.
Sci. Transl. Med. 9 (2017) eaah4680. Management and treatment of atopic dermatitis with topical therapies, J.
[151] S.-Y. Lee, E. Lee, Y.M. Park, S.-J. Hong, Microbiome in the gut-skin axis in atopic Am. Acad. Dermatol. 71 (2014) 116–132.
dermatitis, Allergy Asthma Immunol. Res. 10 (2018) 354–362. [177] I. Katayama, M. Aihara, Y. Ohya, H. Saeki, N. Shimojo, S. Shoji, M. Taniguchi, H.
[152] I. Salem, A. Ramser, N. Isham, M.A. Ghannoum, The gut microbiome as a Yamada, Japanese guidelines for atopic dermatitis 2017, Allergol. Int. 66
major regulator of the gut-skin axis, Front. Microbiol. 9 (2018) 1459. (2017) 230–247.
[153] T. Marrs, C. Flohr, The role of skin and gut microbiota in the development of [178] D.J. Atherton, Topical corticosteroids in atopic dermatitis, BMJ 327 (2003)
atopic eczema, Br. J. Dermatol. 175 (Suppl 2) (2016) 13–18. 942–943.
[154] C.A. O’Neill, G. Monteleone, J.T. McLaughlin, R. Paus, The gut-skin axis in [179] S.G. Danby, J. Chittock, K. Brown, L.H. Albenali, M.J. Cork, The effect of
health and disease: a paradigm with therapeutic implications, Bioessays 38 tacrolimus compared with betamethasone valerate on the skin barrier in
(2016) 1167–1176. volunteers with quiescent atopic dermatitis, Br. J. Dermatol. 170 (2014) 914–
[155] J.D. Forbes, G. Van Domselaar, C.N. Bernstein, The gut microbiota in immune- 921.
mediated inflammatory diseases, Front. Microbiol. 7 (2016) 1081. [180] M.A. Ropke, C. Alonso, S. Jung, H. Norsgaard, C. Richter, M.E. Darvin, T. Litman,
[156] C. Capristo, I. Romei, A.L. Boner, Environmental prevention in atopic eczema A. Vogt, J. Lademann, U. Blume-Peytavi, J. Kottner, Effects of glucocorticoids
dermatitis syndrome (AEDS) and asthma: avoidance of indoor allergens, on stratum corneum lipids and function in human skin-A detailed lipidomic
Allergy 59 (Suppl 78) (2004) 53–60. analysis, J. Dermatol. Sci. 88 (2017) 330–338.

155
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

[181] T.A. Luger, M. Lahfa, R. Folster-Holst, W.P. Gulliver, R. Allen, S. Molloy, N. intrapatient randomized trial, J. Allergy Clin. Immunol. 144 (2019) 1274–
Barbier, C. Paul, J.D. Bos, Long-term safety and tolerability of pimecrolimus 1289.
cream 1% and topical corticosteroids in adults with moderate to severe atopic [201] H. He, E. Guttman-Yassky, JAK inhibitors for atopic dermatitis: an update, Am.
dermatitis, J. Dermatolog. Treat. 15 (2004) 169–178. J. Clin. Dermatol. 20 (2019) 181–192.
[182] W.W. Carr, Topical calcineurin inhibitors for atopic dermatitis: review and [202] W. Damsky, B.A. King, JAK inhibitors in dermatology: the promise of a new
treatment recommendations, Paediatr. Drugs 15 (2013) 303–310. drug class, J. Am. Acad. Dermatol. 76 (2017) 736–744.
[183] N.J. Reynolds, W.I. Al-Daraji, Calcineurin inhibitors and sirolimus: [203] A.T. Virtanen, T. Haikarainen, J. Raivola, O. Silvennoinen, Selective JAKinibs:
mechanisms of action and applications in dermatology, Clin. Exp. prospects in inflammatory and autoimmune diseases, BioDrugs 33 (2019)
Dermatol. 27 (2002) 555–561. 15–32.
[184] K. Gutfreund, W. Bienias, A. Szewczyk, A. Kaszuba, Topical calcineurin [204] L.M. Lundquist, S.W. Cole, M.L. Sikes, Efficacy and safety of tofacitinib for
inhibitors in dermatology. Part I: properties, method and effectiveness of treatment of rheumatoid arthritis, World J. Orthop. 5 (2014) 504–511.
drug use, Postepy Dermatol. Alergol. 30 (2013) 165–169. [205] L.L. Levy, J. Urban, B.A. King, Treatment of recalcitrant atopic dermatitis with
[185] A.F. Abdel-Magid, Modulation of TRPV1 receptor for treatment of pain and the oral Janus kinase inhibitor tofacitinib citrate, J. Am. Acad. Dermatol. 73
other disorders, ACS Med. Chem. Lett. 4 (2013) 155–156. (2015) 395–399.
[186] J. Ring, A. Alomar, T. Bieber, M. Deleuran, A. Fink-Wagner, C. Gelmetti, U. [206] BioSpace, Incyte Announces Positive Data From Phase 2b Trial of Ruxolitinib
Gieler, J. Lipozencic, T. Luger, A.P. Oranje, T. Schafer, T. Schwennesen, S. Cream in Patients With Atopic Dermatitis, (2018) . (Accessed 2020
Seidenari, D. Simon, S. Stander, G. Stingl, S. Szalai, J.C. Szepietowski, A. Taieb, September) https://www.biospace.com/article/releases/incyte-announces-
T. Werfel, A. Wollenberg, U. Darsow, Guidelines for treatment of atopic positive-data-from-phase-2b-trial-of-ruxolitinib-cream-in-patients-with-
eczema (atopic dermatitis) part I, J. Eur. Acad. Dermatol. Venereol. 26 (2012) atopic-dermatitis/.
1045–1060. [207] A. Ostojic, R. Vrhovac, S. Verstovsek, Ruxolitinib for the treatment of
[187] L.F. Eichenfield, W.L. Tom, S.L. Chamlin, S.R. Feldman, J.M. Hanifin, E.L. myelofibrosis: its clinical potential, Ther. Clin. Risk Manag. 8 (2012) 95–103.
Simpson, T.G. Berger, J.N. Bergman, D.E. Cohen, K.D. Cooper, K.M. Cordoro, D. [208] C. Harrison, A.M. Vannucchi, Ruxolitinib: a potent and selective Janus kinase
M. Davis, A. Krol, D.J. Margolis, A.S. Paller, K. Schwarzenberger, R.A. Silverman, 1 and 2 inhibitor in patients with myelofibrosis, An update for clinicians,
H.C. Williams, C.A. Elmets, J. Block, C.G. Harrod, W. Smith Begolka, R. Sidbury, Ther. Adv. Hematol. 3 (2012) 341–354.
Guidelines of care for the management of atopic dermatitis: section 1. [209] H. Nakagawa, O. Nemoto, A. Igarashi, T. Nagata, Efficacy and safety of topical
Diagnosis and assessment of atopic dermatitis, J. Am. Acad. Dermatol. 70 JTE-052, a Janus kinase inhibitor, in Japanese adult patients with moderate-
(2014) 338–351. to-severe atopic dermatitis: a phase II, multicentre, randomized, vehicle-
[188] T. Werfel, N. Schwerk, G. Hansen, A. Kapp, The diagnosis and graded therapy controlled clinical study, Br. J. Dermatol. 178 (2018) 424–432.
of atopic dermatitis, Dtsch Arztebl. Int. 111 (2014) 509–520. [210] W. Amano, S. Nakajima, H. Kunugi, Y. Numata, A. Kitoh, G. Egawa, T.
[189] A. Wollenberg, S. Barbarot, T. Bieber, S. Christen-Zaech, M. Deleuran, A. Dainichi, T. Honda, A. Otsuka, Y. Kimoto, Y. Yamamoto, A. Tanimoto, M.
Fink-Wagner, U. Gieler, G. Girolomoni, S. Lau, A. Muraro, M. Czarnecka- Matsushita, Y. Miyachi, K. Kabashima, The Janus kinase inhibitor JTE-052
Operacz, T. Schafer, P. Schmid-Grendelmeier, D. Simon, Z. Szalai, J.C. improves skin barrier function through suppressing signal transducer and
Szepietowski, A. Taieb, A. Torrelo, T. Werfel, J. Ring, European Dermatology activator of transcription 3 signaling, J. Allergy Clin. Immunol. 136 (2015)
Forum (EDF), the European Academy of Dermatology Venereology (EADV), 667–677 e7.
the European Academy of Allergy Clinical Immunology (EAACI), the [211] J. Peppers, A.S. Paller, T. Maeda-Chubachi, S. Wu, K. Robbins, K. Gallagher, J.E.
European Task Force on Atopic Dermatitis (ETFAD), European Federation of Kraus, A phase 2, randomized dose-finding study of tapinarof (GSK2894512
Allergy and Airways Diseases Patients’ Associations (EFA), the European cream) for the treatment of atopic dermatitis, J. Am. Acad. Dermatol. 80
Society for Dermatology and Psychiatry (ESDaP), the European Society of (2019) 89–98 e3.
Pediatric Dermatology (EPD), Global Allergy and Asthma European [212] S.H. Smith, C. Jayawickreme, D.J. Rickard, E. Nicodeme, T. Bui, C. Simmons, C.
Network (GA2LEN), the European Union of Medical Specialists (UEMS), M. Coquery, J. Neil, W.M. Pryor, D. Mayhew, D.K. Rajpal, K. Creech, S. Furst, J.
Consensus-based European guidelines for treatment of atopic eczema Lee, D. Wu, F. Rastinejad, T.M. Willson, F. Viviani, D.C. Morris, J.T. Moore, J.
(atopic dermatitis) in adults and children: part II, J. Eur. Acad. Dermatol. Cote-Sierra, Tapinarof is a natural AhR agonist that resolves skin
Venereol. 32 (2018) 850–878. inflammation in mice and humans, J. Invest. Dermatol. 137 (2017) 2110–2119.
[190] T. Luger, M. Boguniewicz, W. Carr, M. Cork, M. Deleuran, L. Eichenfield, P. [213] M. Schmuth, V. Moosbrugger-Martinz, S. Blunder, S. Dubrac, Role of PPAR,
Eigenmann, R. Folster-Holst, C. Gelmetti, H. Gollnick, E. Hamelmann, A.A. LXR, and PXR in epidermal homeostasis and inflammation, Biochim. Biophys.
Hebert, A. Muraro, A.P. Oranje, A.S. Paller, C. Paul, L. Puig, J. Ring, E. Siegfried, J. Acta 1841 (2014) 463–473.
M. Spergel, G. Stingl, A. Taieb, A. Torrelo, T. Werfel, U. Wahn, Pimecrolimus in [214] R.W.J. Helder, W.A. Boiten, R. van Dijk, G.S. Gooris, A. El Ghalbzouri, J.A.
atopic dermatitis: consensus on safety and the need to allow use in infants, Bouwstra, The effects of LXR agonist T0901317 and LXR antagonist GSK2033
Pediatr. Allergy Immunol. 26 (2015) 306–315. on morphogenesis and lipid properties in full thickness skin models,
[191] X. Huang, B. Xu, Efficacy and safety of tacrolimus versus pimecrolimus for the Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1865 (2020)158546.
treatment of atopic dermatitis in children: a network meta-analysis, [215] M. Schmuth, P.M. Elias, K. Hanley, P. Lau, A. Moser, T.M. Willson, D.D. Bikle, K.
Dermatology 231 (2015) 41–49. R. Feingold, The effect of LXR activators on AP-1 proteins in keratinocytes, J.
[192] M. Meurer, R. Folster-Holst, G. Wozel, G. Weidinger, M. Junger, M. Brautigam, Invest. Dermatol. 123 (2004) 41–48.
Pimecrolimus cream in the long-term management of atopic dermatitis in [216] T. Czarnowicki, A.B. Dohlman, K. Malik, D. Antonini, R. Bissonnette, T.C. Chan,
adults: a six-month study, Dermatology 205 (2002) 271–277. L. Zhou, H.C. Wen, Y. Estrada, H. Xu, C. Bryson, J. Shen, D. Lala, A. Ma’ayan, G.
[193] H. Kyllonen, A. Remitz, J.M. Mandelin, P. Elg, S. Reitamo, Effects of 1-year McGeehan, R. Gregg, E. Guttman-Yassky, Effect of short-term liver X receptor
intermittent treatment with topical tacrolimus monotherapy on skin activation on epidermal barrier features in mild to moderate atopic
collagen synthesis in patients with atopic dermatitis, Br. J. Dermatol. 150 dermatitis: a randomized controlled trial, Ann. Allergy Asthma Immunol. 120
(2004) 1174–1181. (2018) 631–640 e11.
[194] E.C. Siegfried, J.C. Jaworski, A.A. Hebert, Topical calcineurin inhibitors and [217] R. Sidbury, D.M. Davis, D.E. Cohen, K.M. Cordoro, T.G. Berger, J.N. Bergman, S.L.
lymphoma risk: evidence update with implications for daily practice, Am. J. Chamlin, K.D. Cooper, S.R. Feldman, J.M. Hanifin, A. Krol, D.J. Margolis, A.S.
Clin. Dermatol. 14 (2013) 163–178. Paller, K. Schwarzenberger, R.A. Silverman, E.L. Simpson, W.L. Tom, H.C.
[195] S. Kempers, M. Boguniewicz, E. Carter, M. Jarratt, D. Pariser, D. Stewart, M. Williams, C.A. Elmets, J. Block, C.G. Harrod, W.S. Begolka, L.F. Eichenfield,
Stiller, E. Tschen, K. Chon, S. Wisseh, B. Abrams, A randomized investigator- Guidelines of care for the management of atopic dermatitis: section 3.
blinded study comparing pimecrolimus cream 1% with tacrolimus ointment Management and treatment with phototherapy and systemic agents, J. Am.
0.03% in the treatment of pediatric patients with moderate atopic dermatitis, Acad. Dermatol. 71 (2014) 327–349.
J. Am. Acad. Dermatol. 51 (2004) 515–525. [218] E.C. Siegfried, J.C. Jaworski, P. Mina-Osorio, A systematic scoping literature
[196] L.T. Zane, S. Chanda, K. Jarnagin, D.B. Nelson, L. Spelman, L.S. Gold, Crisaborole review of publications supporting treatment guidelines for pediatric atopic
and its potential role in treating atopic dermatitis: overview of early clinical dermatitis in contrast to clinical practice patterns, Dermatol. Ther. (Heidelb.)
studies, Immunotherapy 8 (2016) 853–866. 8 (2018) 349–377.
[197] D.N. Jouan, New Therapeutic Targets in AD, (2019) . (Accessed October 2019) [219] E.L. Simpson, M. Bruin-Weller, C. Flohr, M.R. Ardern-Jones, S. Barbarot, M.
http://www.world-rendezvous-dermatology.com/en/pack-info-live/ Deleuran, T. Bieber, C. Vestergaard, S.J. Brown, M.J. Cork, A.M. Drucker, L.F.
american-academy-of-dermatology-2019/report/new-therapeutic-targets- Eichenfield, R. Foelster-Holst, E. Guttman-Yassky, A. Nosbaum, N.J. Reynolds,
in-ad/. J.I. Silverberg, J. Schmitt, M.M.B. Seyger, P.I. Spuls, J.F. Stalder, J.C. Su, R.
[198] E.L. Simpson, A.S. Paller, M. Boguniewicz, L.F. Eichenfield, S.R. Feldman, J.I. Takaoka, C. Traidl-Hoffmann, J.P. Thyssen, J. van der Schaft, A. Wollenberg, A.
Silverberg, S.L. Chamlin, L.T. Zane, Crisaborole ointment improves quality of D. Irvine, A.S. Paller, When does atopic dermatitis warrant systemic therapy?
life of patients with mild to moderate atopic dermatitis and their families, Recommendations from an expert panel of the International Eczema Council,
Dermatol. Ther. (Heidelb.) 8 (2018) 605–619. J. Am. Acad. Dermatol. 77 (2017) 623–633.
[199] European Medicines Agency, Staquis (Crisaborole) Summary of Product [220] B. Ungar, S. Garcet, J. Gonzalez, N. Dhingra, J. Correa da Rosa, A. Shemer, J.G.
Characteristics, (2020) . (Accessed May 2020) https://www.ema.europa.eu/ Krueger, M. Suarez-Farinas, E. Guttman-Yassky, An integrated model of atopic
en/documents/product-information/staquis-epar-product-information_en. dermatitis biomarkers highlights the systemic nature of the disease, J. Invest.
pdf. Dermatol. 137 (2017) 603–613.
[200] R. Bissonnette, A.B. Pavel, A. Diaz, J.L. Werth, C. Zang, I. Vranic, V.S. Purohit, M. [221] I.M. Majoie, J.M. Oldhoff, H. van Weelden, M. Laaper-Ertmann, M.T. Bousema,
A. Zielinski, B. Vlahos, Y.D. Estrada, E. Saint-Cyr Proulx, W.C. Ports, E. V. Sigurdsson, E.F. Knol, C.A. Bruijnzeel-Koomen, M.S. de Bruin-Weller,
Guttman-Yassky, Crisaborole and atopic dermatitis skin biomarkers: an Narrowband ultraviolet B and medium-dose ultraviolet A1 are equally

156
T. Luger, M. Amagai, B. Dreno et al. Journal of Dermatological Science 102 (2021) 142–157

effective in the treatment of moderate to severe atopic dermatitis, J. Am. [245] M. de Bruin-Weller, D. Thaci, C.H. Smith, K. Reich, M.J. Cork, A. Radin, Q.
Acad. Dermatol. 60 (2009) 77–84. Zhang, B. Akinlade, A. Gadkari, L. Eckert, T. Hultsch, Z. Chen, G. Pirozzi, N.M.H.
[222] M. Ozawa, K. Ferenczi, T. Kikuchi, I. Cardinale, L.M. Austin, T.R. Coven, L.H. Graham, B. Shumel, Dupilumab with concomitant topical corticosteroid
Burack, J.G. Krueger, 312-nanometer ultraviolet B light (narrow-band UVB) treatment in adults with atopic dermatitis with an inadequate response or
induces apoptosis of T cells within psoriatic lesions, J. Exp. Med. 189 (1999) intolerance to ciclosporin A or when this treatment is medically inadvisable:
711–718. a placebo-controlled, randomized phase III clinical trial (LIBERTY AD CAFE),
[223] U. Khanna, S. Khandpur, What is new in narrow-band ultraviolet-B therapy Br. J. Dermatol. 178 (2018) 1083–1101.
for vitiligo? Indian Dermatol. Online J. 10 (2019) 234–243. [246] D.M. Bullens, A. Kasran, X. Peng, K. Lorre, J.L. Ceuppens, Effects of anti-IL-4
[224] F.J. Legat, The antipruritic effect of phototherapy, Front. Med. (Lausanne) 5 receptor monoclonal antibody on in vitro T cell cytokine levels: IL-4
(2018) 333. production by T cells from non-atopic donors, Clin. Exp. Immunol. 113 (1998)
[225] J.G. Krueger, J.T. Wolfe, R.T. Nabeya, V.P. Vallat, P. Gilleaudeau, N.S. Heftler, L. 320–326.
M. Austin, A.B. Gottlieb, Successful ultraviolet B treatment of psoriasis is [247] L.A. Beck, D. Thaçi, J.D. Hamilton, N.M. Graham, T. Bieber, R. Rocklin, J.E. Ming,
accompanied by a reversal of keratinocyte pathology and by selective H. Ren, R. Kao, E. Simpson, M. Ardeleanu, S.P. Weinstein, G. Pirozzi, E.
depletion of intraepidermal T cells, J. Exp. Med. 182 (1995) 2057–2068. Guttman-Yassky, M. Suárez-Fariñas, M.D. Hager, N. Stahl, G.D. Yancopoulos,
[226] H. Sigmundsdottir, A. Johnston, J.E. Gudjonsson, H. Valdimarsson, A.R. Radin, Dupilumab treatment in adults with moderate-to-severe atopic
Narrowband-UVB irradiation decreases the production of pro- dermatitis, N. Engl. J. Med. 371 (2014) 130–139.
inflammatory cytokines by stimulated T cells, Arch. Dermatol. Res. 297 [248] E.L. Simpson, T. Bieber, E. Guttman-Yassky, L.A. Beck, A. Blauvelt, M.J. Cork, J.I.
(2005) 39–42. Silverberg, M. Deleuran, Y. Kataoka, J.P. Lacour, K. Kingo, M. Worm, Y. Poulin,
[227] L.K. Dotterud, T. Wilsgaard, L.H. Vorland, E.S. Falk, The effect of UVB radiation A. Wollenberg, Y. Soo, N.M. Graham, G. Pirozzi, B. Akinlade, H. Staudinger, V.
on skin microbiota in patients with atopic dermatitis and healthy controls, Mastey, L. Eckert, A. Gadkari, N. Stahl, G.D. Yancopoulos, M. Ardeleanu, SOLO
Int. J. Circumpolar Health 67 (2008) 254–260. 1 and SOLO 2 Investigators, Two phase 3 trials of dupilumab versus placebo in
[228] S.P. Hong, M.J. Kim, M.Y. Jung, H. Jeon, J. Goo, S.K. Ahn, S.H. Lee, P.M. Elias, E.H. atopic dermatitis, N. Engl. J. Med. 375 (2016) 2335–2348.
Choi, Biopositive effects of low-dose UVB on epidermis: coordinate [249] J.I. Silverberg, A. Pinter, G. Pulka, Y. Poulin, J.D. Bouaziz, A. Wollenberg, D.F.
upregulation of antimicrobial peptides and permeability barrier Murrell, A. Alexis, L. Lindsey, F. Ahmad, C. Piketty, A. Clucas, Phase 2B
reinforcement, J. Invest. Dermatol. 128 (2008) 2880–2887. randomized study of nemolizumab in adults with moderate-to-severe atopic
[229] S. Matsuda, S. Koyasu, Mechanisms of action of cyclosporine, dermatitis and severe pruritus, J. Allergy Clin. Immunol. 145 (2020) 173–182.
Immunopharmacology 47 (2000) 119–125. [250] E.L. Simpson, C. Flohr, L.F. Eichenfield, T. Bieber, H. Sofen, A. Taieb, R. Owen, W.
[230] B.D. Kahan, Cyclosporine, N. Engl. J. Med. 321 (1989) 1725–1738. Putnam, M. Castro, K. DeBusk, C.Y. Lin, A. Voulgari, K. Yen, T.A. Omachi,
[231] K. Tsuda, K. Yamanaka, H. Kitagawa, T. Akeda, M. Naka, K. Niwa, T. Nakanishi, Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in
M. Kakeda, E.C. Gabazza, H. Mizutani, Calcineurin inhibitors suppress adults with moderate-to-severe atopic dermatitis inadequately controlled by
cytokine production from memory T cells and differentiation of naïve T cells topical corticosteroids: a randomized, placebo-controlled phase II trial
into cytokine-producing mature T cells, PLoS One 7 (2012)e31465. (TREBLE), J. Am. Acad. Dermatol. 78 (2018) 863–871 e11.
[232] T. Bhutani, C. Sue Lee, J. Koo, Cyclosporine, in: S.E. Wolverton (Ed.), [251] A. Wollenberg, M.D. Howell, E. Guttman-Yassky, J.I. Silverberg, C. Kell, K. Ranade,
Comprehensive Dermatologic Drug Therapy, Saunders/Elsevier, Edinburgh, R. Moate, R. van der Merwe, Treatment of atopic dermatitis with tralokinumab,
2013. an anti-IL-13 mAb, J. Allergy Clin. Immunol. 143 (2019) 135–141.
[233] M.Mary P. Maiberger, J.R. Nunley, S. Wolverton, Other systemic drugs: [252] Y. Renert-Yuval, E. Guttman-Yassky, What’s new in atopic dermatitis,
cyclosporine, in: J. Bolognia (Ed.), Dermatology, Elsevier, 2018. Dermatol. Clin. 37 (2019) 205–213.
[234] S. Khattri, A. Shemer, M. Rozenblit, N. Dhingra, T. Czarnowicki, R. Finney, P. [253] A. De Benedetto, M.K. Slifka, N.M. Rafaels, I.H. Kuo, S.N. Georas, M.
Gilleaudeau, M. Sullivan-Whalen, X. Zheng, H. Xu, I. Cardinale, C. de Guzman Boguniewicz, T. Hata, L.C. Schneider, J.M. Hanifin, R.L. Gallo, D.C. Johnson,
Strong, J. Gonzalez, M. Suárez-Fariñas, J.G. Krueger, E. Guttman-Yassky, K.C. Barnes, D.Y. Leung, L.A. Beck, Reductions in claudin-1 may enhance
Cyclosporine in patients with atopic dermatitis modulates activated susceptibility to herpes simplex virus 1 infections in atopic dermatitis, J.
inflammatory pathways and reverses epidermal pathology, J. Allergy Clin. Allergy Clin. Immunol. 128 (2011) 242–246 e5.
Immunol. 133 (2014) 1626–1634. [254] P.M. Elias, Y. Hatano, M.L. Williams, Basis for the barrier abnormality in atopic
[235] T. Kokuhu, K. Fukushima, H. Ushigome, N. Yoshimura, N. Sugioka, Dose dermatitis: outside-inside-outside pathogenic mechanisms, J. Allergy Clin.
adjustment strategy of cyclosporine A in renal transplant patients: Immunol. 121 (2008) 1337–1343.
evaluation of anthropometric parameters for dose adjustment and C0 vs. C2 [255] D. Roosterman, T. Goerge, S.W. Schneider, N.W. Bunnett, M. Steinhoff,
monitoring in Japan, 2001-2010, Int. J. Med. Sci. 10 (2013) 1665–1673. Neuronal control of skin function: the skin as a neuroimmunoendocrine
[236] J. Schmitt, N. Schmitt, M. Meurer, Cyclosporin in the treatment of patients organ, Physiol. Rev. 86 (2006) 1309–1379.
with atopic eczema - a systematic review and meta-analysis, J. Eur. Acad. [256] E. Azimi, V.B. Reddy, P.J.S. Pereira, S. Talbot, C.J. Woolf, E.A. Lerner, Substance P
Dermatol. Venereol. 21 (2007) 606–619. activates Mas-related G protein-coupled receptors to induce itch, J. Allergy
[237] Drugs.com, Cyclosporine Dosage [last Update Sep 2020], (2020) . Accessed Clin. Immunol. 140 (2017) 447–453 e3.
June https://www.drugs.com/dosage/cyclosporine. [257] W. Zieglgänsberger, Substance P and pain chronicity, Cell Tissue Res. 375
html#Usual_Adult_Dose_for_Organ_Transplant___Rejection_Prophylaxis. (2019) 227–241.
[238] Drugs.com, Cyclosporine [last Update Mar 2020], (2019) . Accessed October [258] J. Kim, B.E. Kim, D.Y.M. Leung, Pathophysiology of atopic dermatitis: clinical
https://www.drugs.com/pro/cyclosporine.html#s-34070-3. implications, Allergy Asthma Proc. 40 (2019) 84–92.
[239] P. Di Filippo, A. Scaparrotta, D. Rapino, A. Cingolani, M. Attanasi, M.I.
Petrosino, K. Chuang, S. Di Pillo, F. Chiarelli, Vitamin D supplementation Thomas Luger, MD, is currently Senior Professor,
modulates the immune system and improves atopic dermatitis in children, Department of Dermatology, University of Münster. He
Int. Arch. Allergy Immunol. 166 (2015) 91–96. obtained his MD from the University of Vienna. His
[240] T.T. Wang, F.P. Nestel, V. Bourdeau, Y. Nagai, Q. Wang, J. Liao, L. Tavera- clinical interest is focused on inflammatory and autoim-
Mendoza, R. Lin, J.W. Hanrahan, S. Mader, J.H. White, Cutting edge: 1,25- mune diseases of the skin, and his main areas of research
dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene are the role of cytokines and neuropeptides in the
expression, J. Immunol. 173 (2004) 2909–2912. regulation of inflammation and immune tolerance. He is
[241] J. Schauber, R.L. Gallo, Antimicrobial peptides and the skin immune defense Past-President of the German Society of Dermatology and
system, J. Allergy Clin. Immunol. 122 (2008) 261–266. former Dean of the Medical Faculty of the University of
[242] P.Y. Ong, D.Y. Leung, Bacterial and viral infections in atopic dermatitis: a Münster. He is a member of the Austrian Academy of
comprehensive review, Clin. Rev. Allergy Immunol. 51 (2016) 329–337. Sciences and the German Academy of Sciences Leopol-
[243] T.R. Hata, P. Kotol, M. Jackson, M. Nguyen, A. Paik, D. Udall, K. Kanada, K. dina. He is currently Honorary Editor of the journal
Yamasaki, D. Alexandrescu, R.L. Gallo, Administration of oral vitamin D Experimental Dermatology.
induces cathelicidin production in atopic individuals, J. Allergy Clin.
Immunol. 122 (2008) 829–831.
[244] Drugs.com, Dupilumab, (2017) . (Accessed October 2019) https://www.drugs.
com/history/dupixent.html.

157

You might also like