SciELO - Scientific Electronic Library Online

 
vol.79 número3Dermatite de Contacto Alérgica Ocupacional na Produção de Pás de Turbinas Eólicas índice de autoresíndice de assuntosPesquisa de artigos
Home Pagelista alfabética de periódicos  

Serviços Personalizados

Journal

Artigo

Indicadores

Links relacionados

  • Não possue artigos similaresSimilares em SciELO

Compartilhar


Revista da Sociedade Portuguesa de Dermatologia e Venereologia

versão impressa ISSN 2182-2395versão On-line ISSN 2182-2409

Rev Soc Port Dermatol Venereol vol.79 no.3 Lisboa set. 2021  Epub 30-Set-2021

https://doi.org/10.29021/spdv.79.3.1405 

Continuous Medical Education

Epidermal Barrier Dysfunction in Atopic Dermatitis

Disfunção da Barreira Epidérmica na Dermatite Atópica

1Dermatology Department, Centro Hospitalar de Leiria, Portugal

2Dermatology Department, Centro Hospitalar e Universitário de Coimbra, Portugal

3Clinic of Dermatology, Faculty of Medicine, University of Coimbra, Portugal


ABSTRACT

Impaired skin barrier is one of the hallmarks of atopic dermatitis (AD), with abnormalities in the cornified envelope, lipid lamellae, tight junctions and cutaneous microbiome. These findings are also present in nonlesional skin of AD individuals, suggesting that epidermal barrier defects may be the initial step towards the development of AD and eventually other atopic diseases (atopic march). It is currently known that pathophysiology of AD involves an interplay between this dysfunctional skin barrier and a predominantly type 2 skewed innate and adaptive immune responses, which further disrupt the skin barrier through type 2 cytokines.

In this setting, there is enhanced penetration of environmental and food allergens through a deficient barrier, leading to an increased susceptibility to sensitization. During the sensitization process, thymic stromal lymphopoietin (TSLP) polarizes skin dendritic cells to a T-helper 2 response, and TSLP seems to be a key cytokine in the sensitization of food allergy, allergic asthma and rhinitis.

In this review, the authors describe the current knowledge of the pathophysiology of the epidermal barrier, its disruption in AD and how it may be involved in the development of atopic comorbidities and the role of barrier repair therapy on the prevention of the atopic march progression.

KEYWORDS: Dermatitis, Atopic; Epidermis; Membrane Proteins; Tight Tunctions; Staphylococcus aureus.

RESUMO

A disrupção da barreira cutânea é um dos achados característicos da dermatite atópica (DA), que inclui alterações no envelope cor-nificado, lamela lipídica, tightjunctions e microbioma cutâneo. Estas alterações estão também presentes na pele não lesada de doentes com DA, o que sugere que os defeitos da barreira epidérmica possam ser a etapa inicial no desenvolvimento da DA e, eventualmente, de outras doenças atópicas (marcha atópica). A evidência atual indica que a fisiopatologia da DA envolve uma interação entre esta barreira cutânea disfuncional e uma resposta imunitária inata e adaptativa predominantemente do tipo 2, que contribuem para a disrupção da barreira epidérmica através da ação de citocinas do tipo 2.

Neste contexto, as anomalias da barreira permitem uma maior penetração dos alergénios ambientais e alimentares, que aumentam a suscetibilidade à sua sensibilização. Durante o processo de sensibilização, a linfopoietina do estroma tímico (TSLP) polariza as células dendríticas da pele para uma resposta T-helper 2, desempenhando também um papel chave na sensibilização da alergia alimentar, asma e rinite alérgicas.

Nesta revisão, os autores descrevem o conhecimento atual da fisiopatologia da barreira epidérmica, a sua alteração na DA e influência no de-senvolvimento de comorbilidades atópicas e o papel da terapêutica de reparação da barreira na prevenção da progressão da marcha atópica.

PALAVRAS-CHAVE: Dermatite Atópica; Epiderme; Junções Íntimas; Proteínas da Membrana; Staphylococcus aureus.

INTRODUCTION

Atopic dermatitis (AD) is a chronic relapsing eczematous dermatosis that affects up to 20% of children and 10% of adults,1,2with 45% beginning within the first 6 months of life and 60% during the first year.1 According to the Global Burden of Disease study, its prevalence has remained stable from 1997 to 2017 in the developed western countries, while it is still rising in the developing countries as industrialization increases.2

Family history of AD is the strongest identifiable risk factor, since the heritability of AD is estimated to be approximately 75%.3,4Polymorphisms of immune response genes include alterations in the T-helper (Th) type 2 signaling pathway and other immune related genes, such as interleukin(IL)-31, IL-33, thymic stromal lymphopoietin (TSLP) and its receptors, Toll-like receptor (TLR) 2 and high affinity IgE receptor.5 Genes encoding skin barrier proteins have also been implicated in AD.6 Filaggrin gene mutations are the strongest risk factor (3-5 times higher risk for AD)4,7,8and may be the initial step towards the development of both AD and other atopic diseases.1,5,9

Pathophysiology involves an interplay between a dysfunctional skin barrier and type 2 skewed innate and adaptive immune responses,4,5,10with an inappropriate activation of Th2 cells and type 2 innate lymphoid (ILC2) cells,11particularly in the acute phase, with production of IL-4, IL-5, IL-13, IL-25, IL-31.12,13 Subsequently, after the sustained activation of Th2, Th22 and, in a smaller degree, Th17 cells, a Th1 activation occurs,4,5,11,12and in chronic lesions, there is an increase in Th1 cells, interferon-γ, IL-5, IL-12 and GM-CSF.10

The main immune pathways involved vary between different ethnicities and may dictate different clinical presentations: higher Th2/Th22 activation in European American patients,14stronger Th17/Th22 pathways in Asians, attenuated Th1/Th17 activation in African Americans and a strong Th17 skewing in early-onset pediatric AD.15,16

AD is frequently associated with other atopic comorbidities, with increasing evidence suggesting that skin defects may trigger further atopic manifestations by enhancing allergen penetration through a defective epidermis and a type 2 skewed immune response triggering IgE-mediated sensitization to food and environmental allergens and predisposing individuals to other atopic diseases that integrate the so called “atopic march”.

In this review, the authors describe the current knowledge of the pathophysiology of the epidermal barrier and its disruption in AD, how it may be involved in the development of atopic comorbidities and eventual preventive measures.

1. Atopic March

The traditional atopic march model is defined as a progression of AD to food allergy, allergic asthma and rhinitis during the first years of life.17AD is considered the first step of this progression with barrier disfunction, inflammatory immune profile and microbial dysbiosis enhancing allergen sensitization.18,19However, according to other authors an alternative atopic march model considers AD followed by any of the other atopic manifestations.19 Belgrave et al investigated two prospective cohorts, with a total of 9801 children, and concluded that only 3.1% of children followed the classic progression whereas according to the alternative model the prevalence of the atopic march increases to 10.5%.20 Apart from the genetic background, environmental factors also influence the atopic march.18

2. Skin Barrier in AD

The skin provides an excellent barrier to prevent pathogen and allergen invasion, minimizing physical and chemical insults, and controlling the normal insensible water loss.21,22

Skin barrier comprises the stratum corneum (air-liquid barrier), tight junctions (liquid-liquid barrier), Langerhans cells and the innate immune cells (immunological barrier) and the microbiome (biological barrier).23,24Chemical and physical properties of the stratum corneum (SC) and tight junctions control permeation of environmental insults, while the competitive microbiome and the interplay between innate and adaptive immune systems allow an immediate and long-lasting protection against pathogens.25

Impaired skin barrier in AD includes abnormalities in the cornified envelope, lipid lamellae, tight junctions and cutaneous microbiome, which are also present in nonlesional AD skin, thus suggesting that epidermal barrier defects precede the development of the clinical AD manifestations,26and, eventually, other related allergic diseases.

2.1 The stratum corneum in AD

The SC composed of a continuous sheet of protein-enriched cells (corneocytes) connected by corneodesmosomes, embedded in an intercellular matrix of multilamellar organized lipids (Fig. 1),21is the main barrier against the entry of external agents. It limits pathogen colonization through its low water content, acidic pH, resident microflora and surface antimicrobial lipids and peptides.21,27

Figure 1 Skin barrier. Stratum corneum is composed of a continuous sheet of corneocytes connected by corneodesmosomes, embedded in an intercellular matrix of multilamellar organized lipids. Lamellar bodies, present in the keratinocytes of the upper stratum spinosum and granulosum, contain the lipid precursors that are secreted at the interface with stratum corneum. Within the stratum granulosum, tight-junctions seal the paracellular pathway to reduce epithelial permeability. Epidermal dendritic cells mediate the link between the innate and adaptive immune system. Langerhans cells elongate their dendritic processes to uptake environmental antigens on the outer side of tight-junctions and then migrate to the regional lymph nodes, where antigen presentation to lymphocytes occurs. 

2.1.1 Cornified envelope

Corneocytes have a cornified envelope, a tough protein/lipid polymer structure21 composed of involucrin, loricrin, small prolinerich proteins (SPRP), envoplakin, periplakin and cysteine protease inhibitor A, that are cross-linked by transglutaminase.28 The internal surface of the cornified envelope is linked to the intracellular keratin filaments,22and involucrin, envoplakin and periplakin of the external surface form covalent ester linkages with ω-hydroxyceramides in the intercellular lipid cement.21,22The external lipid bilayer of the plasma membrane is replaced by a layer of acylceramides, forming the corneocyte lipid envelope.29

The surface area of AD corneocytes in lesional skin is significantly smaller than non-lesional and normal control skin, due to aberrant cornified envelope which impairs the adequate flattening of corneocytes.30 Actually, proteins of the cornified envelope such as loricrin, involucrin and SPRP3 (and their encoding genes) are reduced,31-36translating a very late onset of terminal differentiation in granular keratinocytes.33Moreover, lack or low levels of SPRP, specialized in cross-bridging loricrins, were responsible for bilayer disorganization31,34and were correlated with pruritus and concomitant asthma.31

2.1.2 Filaggrin

Filaggrin (FLG) is a protein which aggregates keratin filaments and forms tight bundles within the keratinocyte cytoskeleton, therefore contributing to corneocyte collapse and flattening.7,37,38Upon degradation, FLG releases hygroscopic amino acids and other molecules, including transurocanic acid and pyrrolidone-5-carboxylic acid, which together with sodium and chloride ions, urea and lactate form the natural moisturizing factor (NMF). This is essential for the low skin pH, the acidic epidermal pH gradient, cutaneous hydration, ultraviolet protection and epidermal barrier integrity.7,37,38FLG deficiency causes disorganized keratin filaments, reduced NMF levels, impaired lamellar body loading and abnormal architecture of the lamellar bilayer.37,39,40

FLG loss of functions variants cause Ichthyosis vulgaris that presents with xerosis, scaling, keratosis pilaris, palmar and plantar hyperlinearity and a strong association with atopic disorders.41 Several loss of function mutations in the FLG gene occur in AD,6,8,42mostly in European American patients.14,43FLG mutations are present in 7%-10% of Europeans, being more frequent in Northern European populations37,42than in East and Southern Europe,44-46and with different frequencies and mutations reported in other ethnic groups, such as Asian and African American.47-50

FLG mutations are especially associated with early-onset AD,8,43,51palmar hyperlinearity,38,52some variants are associated with a mode-rate to severe AD,43,50and FLG expression both in lesional and non-lesional skin is inversely correlated with AD severity.31 Also NMF and FLG status correlate with hyperlinearity in moderate to severe AD,38 suggesting that palmar hyperlinearity may be a clue to FLG mutations in AD patients.

Age-of-onset is highly correlated with FLG status, implicating skin barrier disfunction as a potential cause of the atopic disorder.8 AD patients with food allergy have lower levels of FLG breakdown products in nonlesional skin53 and FLG mutations confer an increased risk of allergic sensitization in children with eczema,43 and greater risk of asthma, allergic rhinitis and food allergy. This suggests that a deficient cutaneous barrier may enhance percutaneous allergen exposure, further development of sensitization and expression of the allergic response through the barriers of others organs (nasal, conjunctival and oral mucosa, digestive tract and lungs).38,39,54,55

Although a few studies show a lack of FLG downregulation in childhood AD,15,16filaggrin deficiency is of paramount importance in a significant percentage of patients, especially in the American European endotype, either due to a loss of function or also due to Th2-driven downregulation.

Actually proinflammatory cytokines, namely IL-4, also cause filaggrin deficiency in AD.36 Dupilumab, a fully human monoclonal antibody directed against the α-subunit of the IL-4 receptor, shared for both IL-4 and IL-13,56 induces an increase of FLG as well as other barrier disruption markers, namely LOR, claudins and ELOVL3, al-ready by week 4 and in a dose-dependent way.57 A similar effect was found with janus kinase (JAK) inhibitors and with an oral dual JAK/spleen tyrosine kinase inhibitor that increased FLG staining and improved epidermal hyperplasia along with suppression of Th2, Th17/Th22 and Th1 cytokine pathways.58 Nevertheless, as none of these drugs have been used in very young children, their effects on the atopic march are still unknown.

2.1.3 Corneodesmosomes

Corneodesmosomes are the main intercellular adhesive structure in the SC and provide tensile strength for corneocytes to resist shea-ring forces.59,60Their extracellular parts contain two desmosomal cadherins, desmoglein 1 and desmocollin 1, and corneodesmosin, the major constitutional difference from desmosomes.59,61CDSN gene is downregulated in AD skin,33,62but corneodesmosome integrity and CDSN gene expression can also be modulated by Th2 cytokines (IL-4, IL-13, IL-22, IL-25 and IL-31).62 In murine skin and cultured human keratinocytes, IL-4 decreases the number of corneodesmosomes and downregulates desmoglein 1 expression.63

Exogenous and endogenous proteases control corneodesmo-some cleavage. Kallikrein-related peptidases (KLK) and cathepsins produced by keratinocytes, are involved in the cleavage of the corneodesmosome junctions,59-61whereas protease inhibitors like the lympho-epithelial Kazal type inhibitor (LEKTI) encoded by the serine protease inhibitor Kazal-type 5 gene (SPINK5), inhibit KLKs.9,64

Desquamation follows corneodesmosome cleavage in a pH-dependent way. In the deep stratum corneum, neutral pH allows a strong LEKTI and KLK interaction and prevents corneodesmosome cleavage. However, as pH lowers in the superficial SC, LEKTI/KLK dissociate, allowing corneodesmosome degradation and desquamation.60,61

SPINK5 polymorphisms are associated with AD in certain populations,65-67and AD is associated with increased activity of SC serine proteases,68,69whose activity was correlated with total serum IgE and peripheral blood eosinophilia.69 Moreover, since KLK5 upregulates TSLP mRNA expression, lack of LEKTI is also associated with keratinocyte overexpression of TSLP,70a cytokine that enhances Th2 skewing not only during the sensitization process, but also during the inflammatory loop that perpetuates AD.

2.1.4 Lipid lamellae

Intercellular lipids, which represent 10%-15% of the total SC mass, form the extracellular SC matrix with densely packed lipid layers (lipid lamellae)28,29that control water retention within the SC and prevent allergen penetration.71

Lipid lamellae are formed by an equimolar ratio of cholesterol, free fatty acids (FFA) and ceramides,29,72,73which by weight represent 45%-50% of ceramides, 25% of cholesterol, 10%-15% of free fatty acids, 5% of cholesterol sulphate and a small percentage of triacylglycerol species.22,71Aberrant lipid proportion results in skin barrier disfunction,28,74and cholesterol, ceramides and FFA must be supplied in the adequate proportion to recover barrier disruption. In an acutely damaged skin, topical applications of a single or two of the three key lipids actually delays barrier recovery, whereas topical equimolar mixtures normalize recovery rates.72

Glucosylceramides, sphingomyelin and phospholipids stored in lamellar bodies inside the keratinocytes of the upper stratum spinosum and granulosum are the precursors of these SC lipids.73 At the interface with the SC, lamellar bodies fuse with the cell membrane and secrete these lipid precursors into the extracellular space as well as β-glucocerebrosidase, acid sphingomyelinase and phospholipase A, which are further metabolized to give rise to ceramides and FFAs.28,73Ceramides contain mostly very long fatty acid chains linked via an amide to a sphingosine chain74 and have substantial quantities of highly hydrophobic ω-esterified ceramides arranged in lamellar membranes that control water retention.75 Cholesterol sulfate is broken down by sterol sulfatase to cholesterol.76 A reduction in the length of the chains of ceramides and FFAs,75,77creates shorter and less condensed ceramide membranes with higher water permeability.78 This may be due to an altered expression of fatty acid elongases (ELOVL),79 as shown by a reduced expression of ELOVL1, ELOVL3 and ELOVL6 in lesional skin.75,80

SC lipids (both ceramides and FFAs) are decreased in AD,77 and ceramide levels are significantly correlated with SCORAD and TEWL.81 Reduction in ω-esterified fatty acid sphingosine ceramides in the SC of AD patients was correlated with high TEWL, sensitization to food allergens and clinical manifestations of food allergy.53

The immune response, namely Th2 cytokines (IL-4/IL-13) in AD, also play a negative role in ceramide metabolism,75,81by reducing mRNA levels encoding sphingomyelinase and glucocerebrosidase and downregulating ELOVL1, ELOVL3 and ELOVL6 expression.75,80,82

These lipid barrier defects have been associated with both pediatric and adult AD. However, some lipid-associated mediators, such as fatty acyl-CoA reductase 2 and fatty acid 2-hydroxylase are preferentially downregulated in pediatric AD.15

2.2 Tight Junctions

Tight junctions (TJ) are cell-cell junctions located in stratum granulosum, that seal the paracellular pathway to reduce epithelial permeability and restrict the movement of molecules within the intercellular space,9,83namely water, ions, proteins and also Langerhans cells’ dendrites.9,26,84TJ are formed by transmembrane proteins (claudins, ocludins, junctional adhesion molecule A and tricellunin) and cytosolic plaque proteins (Zonula occludens (ZO)-1, ZO-2, ZO-3, multi-PDZ domain protein 1, membrane-associate guanylate kinase and cingulin).85-87

The epidermis is very rich in claudin-1 and claudin-488 but their levels are reduced in AD, causing barrier disfunction assessed by lower transepithelial electrical resistance and higher paracellular permeability.89,90Claudin-1 knockout mice die within the first day of life due to a severely defective epidermal barrier,91 altered ceramide composition and insufficient filaggrin processing, supporting that a defective stratum granulosum may lead to an aberrant stratum corneum.90,92,93Also, low claudin-1 levels in AD lesional skin90,92-94correlate both with TJ barrier93 and abnormal epidermal differentiation and inflammation.88 However, claudin-1 levels in nonlesional skin have shown both reduced or normal values in cohorts from similar ancestries. 89,92-94

Variants of claudin-1 gene (CLDN1) have been associated with AD risk in an African-American cohort,26,89early onset AD in an Ethio-pian cohort,95 and AD with specific IgE to environmental molds,96 but not in a Finish cohort.51 There is also evidence that levels of claudin-4, claudin-23 and ZO-1, may be decreased in some AD patients.89,94

As for filaggrin and lipid lamellae, TJs influence and are influenced by the immune response, although contradictory results have been reported. De Benedetto et al observed an inverse correlation between epidermal claudin-1 expression and markers of Th2 polarity (blood eosinophilia and serum total IgE),89 whereas Tokumasu et al found no correlation between claudin-1 level and IL-4 in AD pa-tients.90 IL-33 down-regulates CLDN1 expression in keratinocytes,97 and low claudin-1 levels induce IL-1β production and promote in-flammation in the presence of Staphylococci, even non-pathogenic strains.93

TJs are also critical in containing viral spread, and claudin-1 reduction enhances susceptibility to herpes simplex virus 1 (HSV-1) infection.98

2.3 Immune skin barrier and antimicrobial peptides

The epidermal immunological barrier includes both cellular and humoral components. Resident skin cells have a vast group of survei-llance receptors that recognize pathogens and other insults and activate innate immunity with cytokine and chemokine production and recruitment of neutrophils, monocytes, macrophages, dendritic cells (DC) and T lymphocytes, that will then be involved in the adaptative immune response.25

Keratinocytes bear membrane and cytosolic pathogen recognition receptors (PRRs), namely TLRs, RIG-I-like receptors, NOD-like receptors and DNA receptors25,99that recognize pathogen-associated molecular patterns (PAMPs), such as bacterial lipopolysaccharides or viral RNA, and damage associated molecular patterns (DAMPs), namely hyaluronic acid, heat shock proteins, oxidized lipids or lipo-proteins.25,83,100Upon activation by PAMPs or DAMPs, keratinocytes produce antimicrobial peptides and inflammatory cytokines of the innate immunity,25,100namely defensins, cathelicidins, S100 proteins, ribonucleases and dermcidin, with a broad spectrum activity against bacteria, virus, fungi and parasites. They also have immuno-modulatory properties and are involved in skin barrier maintenance.101 Human-β-defensins (HBD) and cathelicidins (LL-37) increase upon cutaneous infection, inflammation or wound, but their expres-sion is lower in AD skin.102,103Furthermore, Th2-derived cytokines suppress HBD expression,103,104with TSLP inhibiting HBD-2 through a JAK2/STAT3-dependent pathway.102 Lower expression of LL-37, HBD-2 and HBD-3 in lesional skin has been associated with eczema herpeticum and bacterial skin infections,102,105and the susceptibility to the latter is further aggravated by the reduced dermcidin levels in AD sweat.106

Epidermal DC, namely Langerhans cells (LC), have similar PRRs, which upon activation, induce dendrite elongation to uptake extra-TJ antigens, before TJ barriers seal again the intercellular space.84Then, DC migrate to regional lymph nodes where they present the antigen to lymphocytes.107 Therefore, the innate immune system mounts an effective defense against pathogens and initiates the adaptive im-mune response.99 Dermal innate lymphoid cells (ILC) also contribute to natural immune defense and liaison with the acquired immune response,108 but in AD there is a very significant predominance of IL-5/IL-13 producing ILC (ILC2), causing a shift to a type 2 immune response.109,110

2.4 The biological barrier and Staphylococcus aureus in AD

Skin commensals are essential for skin microbiome equilibrium and activation of resident T cells and keratinocytes, as they educa-te them in combatting skin pathogens.111 In AD this equilibrium is impaired and significantly compromises this biological barrier and enhances infectious complications.112,113

Staphylococcus aureus (S. aureus) colonizes AD skin in an extent significantly correlated with disease activity.114,115Risk factors for S. aureus colonization include decreased levels of filaggrin and filaggrin degradation products, lower levels of coagulase-negative Staphylococcus, altered lipid profiles, deficiency of antimicrobial peptides and overexpression of Th2 cytokines116-119as IL-4 and IL-13 downregulate LL-37 and HBD-3.117 Also, fewer coagulase-negative Staphylococci, such as S. epidermidis and S. hominis, allow a shift to S. aureus predominance during AD flares. This can be reversed by topical application of coagulase-negative Staphylococcus strains and other non-pathogenic bacteria,116,120which also decrease SCORAD and pruritus, and alleviate cutaneous inflammation in AD.121,122Moreover, antimicrobial or anti-inflammatory treatment of AD flares also increases skin microbiome diversity.120

S. aureus exotoxins increase proinflammatory cytokines and proteases that affect keratinocytes and various immune cells in AD skin.117Toxic shock syndrome toxin-1 (TSST-1), staphylococcal ente-rotoxins (SEA, SEB, SEC, SED), exfoliative toxins (ETA and ETB), and leucocidin,123behave as superantigens and induce polyclonal T and B cell proliferation and class switching to IgE and production of allergen-specific IgE in mucosal B cells,117,124and SEB increases IL-31 expression, involved in pruritus, inhibits keratinocyte differentiation and suppresses filaggrin expression.117

S. aureus may impair the usual tolerance to food and enhance food sensitization.125 Nasal carriage of S. aureus and skin coloniza-tion occur more frequently in children with AD and food allergy and patients with allergic rhinitis.126-128MRSA colonization is associated with higher levels of peanut specific IgE,129and increased and persistent levels of specific IgE to egg white and peanut, independent of eczema severity.125 In mice models, S. aureus enterotoxins, like SEB, in combination with ovalbumin enhance epicutaneous sensitization with specific IgE against SEB and ovalbumin,124and augment ovalbumin induced airway hyperresponsiveness and lung inflammation, via an IL-17A-dependent pathway.130 Also, oral administration of SEB with ovalbumin resulted in immune responses to ovalbumin, with decreased regulatory T-cell function, impairment of immune tolerance and a predominant Th2 response.131

In AD, vicious cycles between S. aureus infection and AD exacerbation induce TSLP and favor Th2/Th17-type inflammation, IgE specific allergen sensitization and tissue damage.117

In summary, S. aureus colonization and dysbiosis in AD influence disease severity (IgE anti-S. aureus toxins), further disrupt the skin barrier, enhance food sensitization, inhibit oral tolerance induction and promote allergic responses in other organs, which may, therefore, be regarded as one of drivers of the atopic march.125

3. Environmental allergens and epidermal sensitization through AD skin

Physical, chemical and immunological epidermal barrier defects in AD may explain an increased sensitization to environmental and food allergens, due to their enhanced penetration but some of these allergens also contribute to skin barrier defects, as shown for S. aureus.

3.1 Environmental allergens and protease activity

Many aeroallergens (pollen, housedust mite (HDM), cockroaches, Aspergillus sp, Penicillium sp) and some food-derived allergens have protease activity,132,133which in addition to endogenous proteases further disrupt the skin barrier and enhance inflammation, IgE production and allergen sensitization.134 Epidermal cytokines and chemokines induced by mite proteases attract and activate inflammatory cells and APC, suggesting the role of mite proteases in the initiation of sensitization through the skin.134

Proteases from cockroaches and mites activate protease-activated receptor 2, delay epidermal barrier recovery and lamellar body secretion after acute barrier disruption,135 therefore enhancing penetration of allergens, irritants and molecules from other environmental allergen and microbes, that initiate and perpetuate inflammation.134

Moreover, aeroallergens and food-derived allergens with protease activity enhance specific IgE production and are further specifically recognized by IgE in sensitized patients.134 In human keratinocytes, HDM allergens stimulate innate IL-33 production, and environmental proteases promote Th2 skewing and IgE production.134

Therefore, barrier disruption aggravated by environmental proteases increases accessibility and IgE sensitization to these proteases, with subsequent initiation and perpetuation of inflammation both in the skin and other organs exposed to similar environmental allergens.134

3.2 Impaired cutaneous barrier and percutaneous sensitization

As previously referred, epidermal barrier disruption, further aggravated by itching, allows skin-resident APC, such as LC or DC, to reach the upper epidermis with their dendrites and capture environmental antigens more easily.9 Furthermore, barrier disrupted keratinocytes release alarmins and type-2 cytokines which behave as immune adjuvants to activate, mature DC and enhance their sensitizing capacity.9 In this setting, skin DC express high affinity IgE receptors, become coated with IgE, easily recognize and process aeroallergens or food allergens and cause both immediate and delayed allergic reactions.136 Many AD patients have increased IgE-media-ted reactivity to aeroallergens, food proteins and microbial antigens which may precipitate atopic flares in IgE sensitized patients. However, there is not enough evidence to recommend strict avoidance of such allergens to improve or prevent AD.4

Other cutaneous disorders with skin barrier disruption have an increased risk of allergen sensitization, corroborating the relation between skin barrier disruption and allergic sensitization. In Netherton syndrome, an autosomal recessive disorder caused by loss of function mutations in SPINK5 with consequent deficiency of LEKTI-1/KLK inhibition, increased protease activity, enhanced degradation of corneodesmosomes and SC detachment,137 there is severe skin inflammation and allergic manifestations with elevated IgE, TSLP and TNF-α production.64,138Peeling skin syndrome type B, another rare autosomal recessive genodermatosis caused by CDSN mutations,139,140is associated with severe food allergies, allergic asthma and rhinitis, high IgE and eosinophilia.139,141

3.3 TSLP and Th2 cytokines in food allergy, allergic asthma and allergic rhinitis

TSLP, an IL-7-like cytokine mainly expressed by epithelial cells and keratinocytes at barrier surfaces,70 is critically involved in body-environment interactions, Th2 responses and type 2 inflammation in multiple disease settings, including food allergy, allergic asthma and allergic rhinitis.142,143

TSLP is highly expressed in acute and chronic AD lesions144,145where it can be induced by environmental (allergens, virus, helminths, cigarette smoke or chemicals) or endogenous triggers (Th2-related cytokines and IgE), suggesting its effect on Th2 amplification.70 TSLP polarizes skin DCs to a Th2 response during sensitization to food and aeroallergens presented through the damaged skin barrier of AD skin (face, lips, hands).142 Actually, in the cutaneous ambience rich in ILC2 and TSLP, IL-5 and IL-4/IL-13, APC that migrate to the draining lymph nodes will sensitize naïve CD4+ T cells that will differentiate into allergen-specific Th2 cells that will promote class switching to IgE and form allergen-specific IgE memory B and plasma cells.111,146,147In a further exposure, allergens recognized by specific IgEs bound to high affinity FcεRI receptors on mast cells and baso-phils,111 cause immediate symptoms of food allergy (localized or generalized urticaria, gastrointestinal symptoms or even anaphylaxis), allergic asthma (bronchospasm, mucus production, and dyspnea), allergic rhinitis (pruritus, sneezing and rhinorrhea), and allergic conjunctivitis (pruritus, red eye and weeping).

TSLP seems to be a key cytokine in sensitization to food exposed through the skin, as it increases antigen-specific serum IgE levels and enhances accumulation of intestinal mast cells involved in intestinal food allergy.142,148In murine models, sensitization to ovalbumin and peanut through AD-like skin induced TSLP increase in the skin, whereas knock-out mice lacking TSLP receptor were not prone to food allergy.148,149

Similar pathways of sensitization to aeroallergens through AD skin and development of allergic asthma also involve TSLP production by keratinocytes,142 and asthmatic patients have high expres-sion of TSLP in the serum and airway epithelium.142,150

Experimental models also implicate TSLP in allergic rhinitis, with TSLP receptor-deficient mice having less IgE production and less severe early responses in allergic rhinitis.143

A phase III trial with tezepelumab, a human monoclonal antibody against TSLP, in patients with severe uncontrolled asthma, showed a significantly lower annualized rate of asthma exacerbations compa-red to the placebo group.151 However, in a phase IIa study, tezepelumab in combination with topical steroids did not achieve statistically significant improvement in moderate to severe AD.152

Dupilumab as well as JAK inhibitors progressively reverse the lesional transcriptome in AD and significantly reduce type 2 inflammation genes expression, epidermal hyperplasia, T cells, DC and Th17/Th22 activity.57,58,153However, these drugs have not been introduced early enough to evaluate their capacity to prevent sensitization to environmental allergens and, consequently, influence the atopic march.

4. Early barrier intervention and immune modulation - future perspectives on AD prevention and treatment

In the last decades, several studies have tried to modify the IgE-sensitization-promoting microenvironment of AD skin to prevent the atopic march, but results have not been convincing.154-158

Emollients and other general measures that enhance skin barrier repair from the very early days of life have generated contradictory results, eventually related to the diversity of emollients used. In fact, paraffinbased/alcohol-based/petroleum-based emollients can be detrimental in AD, since the occlusion inhibits keratinocyte differentiation and enhances S. aureus colonization,159 whereas triple physiologic lipid-based barrier repair therapies composed of a ceramide-dominant mixture of the 3 key SC lipids (a 3:1:1 molar ratio of ceramides, cholesterol and free fatty acids) seem more effective. They reduce TEWL in AD children as they amplify lipid production and delivery to the SC intercellular spaces, reple-nish the lamellar bilayers,159,160and prevent Th2 cytokine-induced reduction in SC ceramide content.160 Ongoing large randomized trials (PEBBLES and PreventADALL) are evaluating the effect of such emollients in preventing AD and food allergy in the first 12 months of life in infants with a family history of allergic disease.156,161

CONCLUSION

Epidermal defects causing chemical, physical, immunological and biological disruption of the skin barrier are a key feature of AD. Abnormalities in the cornified envelope, filaggrin, lipid lamellae and tight junctions lead to a vulnerable epidermis, itching and abnormal innate immune response with fewer antimicrobial peptides and dysbiosis. A Th2 shifted immune response further compromise the barrier by downregulating the synthesis of relevant epidermal components.

There is increasing evidence suggesting that a disrupted skin barrier in AD, along with a type 2 skewed immune response, allows IgE sensitization and subsequent development of food allergy and airway hyperreactivity, and is, therefore, the initial event in the atopic march.

Nevertheless, a better characterization of AD skin defects and how they really influence progression to other atopic diseases still needs further study. Therapies targeting these defects also need to be further studied as they may have beneficial implications in the prevention of AD and other atopic diseases162 that represent a real burden for the patients and the society.2

REFERENCES

1. Bieber T. Atopic Dermatitis. N Engl J Med. 2008;358:1483-94. doi:10.1056/NEJMra074081 [ Links ]

2. Laughter MR, Maymone MB, Mashayekhi S, Arents BW, Karimkhani C, Langan SM, et al. The global burden of atopic dermatitis: lessons from the GBD Study - 1990 to 2017. Br J Dermatol. 2020;184:304-9. doi:10.1111/bjd.19580 [ Links ]

3. Elmose C, Thomsen SF. Twin Studies of Atopic Dermatitis: Interpretations and Applications in the Filaggrin Era. J Allergy. 2015;2015:1-7. doi:10.1155/2015/902359 [ Links ]

4. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396:345-60. doi:10.1016/S0140-6736(20)31286-1 [ Links ]

5. Kim J, Kim BE, Leung DY. Pathophysiology of atopic dermatitis: Clinical implications. Allergy Asthma Proc. 2019;40:84-92. doi:10.2500/aap.2019.40.4202 [ Links ]

6. Bin L, Leung DY. Genetic and epigenetic studies of atopic dermatitis. Allergy, Asthma Clin Im-munol. 2016;12:52. doi:10.1186/s13223-016-0158-5 [ Links ]

7. Cabanillas B, Novak N. Atopic dermatitis and filaggrin. Curr Opin Immunol. 2016;42:1-8. doi:10.1016/j.coi.2016.05.002 [ Links ]

8. Smieszek SP, Welsh S, Xiao C, Wang J, Polymeropoulos C, Birznieks G, et al. Correlation of age-of-onset of Atopic Dermatitis with Filaggrin loss-of-function variant status. Sci Rep. 2020;10:2721. doi:10.1038/s41598-020-59627-7 [ Links ]

9. De Benedetto A, Kubo A, Beck LA. Skin barrier disruption: a requirement for allergen sensitization? J Invest Dermatol. 2012;132:949-63. doi:10.1038/jid.2011.435 [ Links ]

10. Fujii M. Current understanding of pathophysiological mechanisms of atopic dermatitis: interactions among skin barrier dysfunction, immune abnormalities and pruritus. Biol Pharm Bull. 2020;43:12-9. doi:10.1248/bpb.b19-00088 [ Links ]

11. Moyle M, Cevikbas F, Harden JL, Guttman-Yassky E. Understanding the immune landscape in atopic dermatitis: The era of biologics and emerging therapeutic approaches. Exp Dermatol. 2019;28:756-68. doi:10.1111/exd.13911 [ Links ]

12. Torres T, Ferreira EO, Gonçalo M, Mendes-Bastos P, Selores M, Filipe P. Update on Atopic Dermatitis. Acta Med Port. 2019;32:606-13. doi:10.20344/amp.11963 [ Links ]

13. Grobe W, Bieber T, Novak N. Pathophysiology of atopic dermatitis. J Dtsch Dermatol Ges. 2019;17:433-40. doi:10.1111/ddg.13819 [ Links ]

14. Nomura T, Wu J, Kabashima K, Guttman-Yassky E. Endophenotypic Variations of Atopic Dermatitis by Age, Race, and Ethnicity. J Allergy Clin Immunol Pract. 2020;8:1840-52. doi:10.1016/j.jaip.2020.02.022 [ Links ]

15. Brunner PM, Israel A, Zhang N, Leonard A, Wen HC, Huynh T, et al. Early-onset pediatric atopic dermatitis is characterized by TH2/TH17/TH22-centered inflammation and lipid alterations. J Allergy Clin Immunol. 2018;141:2094-106. doi:10.1016/j.jaci.2018.02.040 [ Links ]

16. Esaki H, Brunner PM, Renert-Yuval Y, Czarnowicki T, Huynh T, Tran G, et al. Early-onset pediatric atopic dermatitis is T H 2 but also T H 17 polarized in skin. J Allergy Clin Immunol. 2016;138:1639-51. doi:10.1016/j.jaci.2016.07.013 [ Links ]

17. Spergel JM. From atopic dermatitis to asthma: The atopic march. Ann Allergy, Asthma Immunol. 2010;105:99-106. [ Links ]

18. Aw M, Penn J, Gauvreau GM, Lima H, Sehmi R. Atopic March: Collegium Internationale Allergologicum Update 2020. Int Arch Allergy Immunol. 2020;181:1-10. doi:10.1159/000502958 [ Links ]

19. Hill DA, Camargo CA, Paller AS, Spergel JM. A march by any other name. Ann Allergy, Asthma Immunol. 2018;121:137-8. doi:10.1016/j.anai.2018.04.014 [ Links ]

20. Belgrave DC, Granell R, Simpson A, Guiver J, Bishop C, Buchan I, et al. Developmental profiles of eczema, wheeze, and rhinitis: two population-based birth cohort studies. PLoS Med. 2014;11:e1001748. doi:10.1371/journal.pmed.1001748. [ Links ]

21. Proksch E, Brandner JM, Jensen JM. The skin: an indispensable barrier. Exp Dermatol. 2008;17:1063-72. doi:10.1111/j.1600-0625.2008.00786.x [ Links ]

22. Madison KC. Barrier Function of the Skin: »,» ®,® §,§ ­,­ ¹,¹ ²,² ³,³ ß,ß Þ,Þ þ,þ ×,× Ú,Ú ú,ú Û,Û û,û Ù,Ù ù,ù ¨,¨ Ü,Ü ü,ü Ý,Ý ý,ý ¥,¥ ÿ,ÿ ¶,¶ La Raison d'Être »,» ®,® §,§ ­,­ ¹,¹ ²,² ³,³ ß,ß Þ,Þ þ,þ ×,× Ú,Ú ú,ú Û,Û û,û Ù,Ù ù,ù ¨,¨ Ü,Ü ü,ü Ý,Ý ý,ý ¥,¥ ÿ,ÿ ¶,¶ of the Epidermis. J Invest Dermatol. 2003;121:231-41. doi:10.1046/j.1523-1747.2003.12359.x [ Links ]

23. Kubo A, Nagao K, Amagai M. Epidermal barrier dysfunction and cutaneous sensitization in atopic diseases. J Clin Invest. 2012;122:440-7. [ Links ]

24. Skabytska Y, Kaesler S, Volz T, Biedermann T. How the innate immune system trains immu-nity: lessons from studying atopic dermatitis and cutaneous bacteria. J Dtsch Dermatol Ges. 2016;14:153-6. doi:10.1111/ddg.12843 [ Links ]

25. Sun L, Liu W, Zhang L. The Role of Toll-Like Receptors in Skin Host Defense, Psoriasis, and Atopic Dermatitis. J Immunol Res. 2019;2019:1-13. doi:10.1155/2019/1824624 [ Links ]

26. Brandner J, Zorn-Kruppa M, Yoshida T, Moll I, Beck L, De Benedetto A. Epidermal tight junctions in health and disease. Tissue Barriers. 2015;3:e974451. doi:10.4161/21688370.2014 .974451 [ Links ]

27. Elias PM. The skin barrier as an innate immune element. Semin Immunopathol. 2007;29:3-14. [ Links ]

28. Lee A-Y. Molecular Mechanism of Epidermal Barrier Dysfunction as Primary Abnormalities. Int J Mol Sci. 2020;21:1194. doi:10.3390/ijms21041194 [ Links ]

29. Kubo A, Amagai M. Skin Barrier. In: Kang S, Amagai M, Bruckner A, et al., editors. Fitzpatrick’s Dermatology. 9th ed. New York: McGraw-Hill; 2019: p.206-31. [ Links ]

30. Igawa S, Kishibe M, Honma M, Murakami M, Mizuno Y, Suga Y, et al. Aberrant distribution patterns of corneodesmosomal components of tape-stripped corneocytes in atopic dermatitis and related skin conditions (ichthyosis vulgaris, Netherton syndrome and peeling skin syndro-me type B). J Dermatol Sci. 2013;72:54-60. doi:10.1016/j.jdermsci.2013.05.004 [ Links ]

31. Trzeciak M, Sakowicz-Burkiewicz M, Wesserling M, Dobaczewska D, Glen J, Nowicki R, et al. Expression of Cornified Envelope Proteins in Skin and Its Relationship with Atopic Dermatitis Phenotype. Acta Derm Venereol. 2017;97:36-41. doi:10.2340/00015555-2482 [ Links ]

32. Trzeciak M, Olszewska B, Sakowicz-Burkiewicz M, Sokołowska-Wojdyło M, Jankau J, et al. Expression Profiles of Genes Encoding Cornified Envelope Proteins in Atopic Dermatitis and Cutaneous T-Cell Lymphomas. Nutrients. 2020;12:862. doi:10.3390/nu12030862 [ Links ]

33. Guttman-Yassky E, Suárez-Fariñas M, Chiricozzi A, Nograles KE, Shemer A, Fuentes-Duculan J, et al. Broad defects in epidermal cornification in atopic dermatitis identified through genomic analysis. J Allergy Clin Immunol. 2009;124:1235-44.e58. doi:10.1016/j.jaci.2009.09.031 [ Links ]

34. Carregaro F, Stefanini ACB, Henrique T, Tajara EH. Study of small proline-rich proteins (SPRRs) in health and disease: a review of the literature. Arch Dermatol Res. 2013;305:857-66. doi:10.1007/s00403-013-1415-9 [ Links ]

35. Kim BE, Leung DYM, Boguniewicz M, Howell MD. Loricrin and involucrin expression is down-regulated by Th2 cytokines through STAT-6. Clin Immunol. 2008;126:332-7. [ Links ]

36. Howell MD, Kim BE, Gao P, Grant AV, Boguniewicz M, DeBenedetto A, et al. Cytokine modu-lation of atopic dermatitis filaggrin skin expression. J Allergy Clin Immunol. 2009;124:7-12. doi:10.1016/j.jaci.2009.07.012 [ Links ]

37. Brown SJ, Irwin McLean WH. One Remarkable Molecule: Filaggrin. J Invest Dermatol. 2012;132:751-62. doi:10.1038/jid.2011.393 [ Links ]

38. Drislane C, Irvine AD. The role of filaggrin in atopic dermatitis and allergic disease. Ann Allergy, Asthma Immunol. 2020;124:36-43. doi:10.1016/j.anai.2019.10.008 [ Links ]

39. Kawasaki H, Nagao K, Kubo A, Hata T, Shimizu A, Mizuno H, et al. Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrinnull mice. J Allergy Clin Immunol. 2012;129:1538-46.e6. doi:10.1016/j.jaci.2012.01.068 [ Links ]

40. Vávrová K, Henkes D, Strüver K, Sochorová M, Školová B, Witting MY, et al. Filaggrin Deficiency Leads to Impaired Lipid Profile and Altered Acidification Pathways in a 3D Skin Construct. J Invest Dermatol. 2014;134:746-53. doi:10.1038/jid.2013.402 [ Links ]

41. Thyssen JP, Godoy-Gijon E, Elias PM. Ichthyosis vulgaris: the filaggrin mutation disease. Br J Dermatol. 2013;168:1155-66. doi:10.1111/bjd.12219 [ Links ]

42. Rodríguez E, Baurecht H, Herberich E, Wagenpfeil S, Brown SJ, Cordell HJ, et al. Meta-analysis of filaggrin polymorphisms in eczema and asthma: Robust risk factors in atopic disease. J Allergy Clin Immunol. 2009;123:1361-70.e7. doi:10.1016/j.jaci.2009.03.036 [ Links ]

43. Martin MJ, Estravís M, García-Sánchez A, Dávila I, Isidoro-García M, Sanz C. Genetics and Epigenetics of Atopic Dermatitis: An Updated Systematic Review. Genes. 2020;11:442. doi:10.3390/genes11040442 [ Links ]

44. Giardina E, Paolillo N, Sinibaldi C, Novelli G. R501X and 2282del4 Filaggrin Mutations Do Not Confer Susceptibility to Psoriasis and Atopic Dermatitis in Italian Patients. Dermatology. 2008;216:83-4. doi:10.1159/000109365 [ Links ]

45. Jurakic Toncic R, Kezic S, Jakasa I, Ljubojevic Hadzavdic S, Balic A, et al. Filaggrin loss-of-function mutations and levels of filaggrin degradation products in adult patients with atopic dermatitis in Croatia. J Eur Acad Dermatology Venereol. 2020;34:1789-94. doi:10.1111/jdv.16232 [ Links ]

46. González-Tarancón R, Sanmartín R, Lorente F, Salvador-Rupérez E, Hernández-Martín A, Rello L, et al. Prevalence of FLG loss-of-function mutations R501X, 2282del4, and R2447X in Spanish children with atopic dermatitis. Pediatr Dermatol. 2020;37:98-102. doi:10.1111/pde.14025 [ Links ]

47. Chen H, Common JEA, Haines RL, Balakrishnan A, Brown SJ, Goh CS, et al. Wide spectrum of filaggrin-null mutations in atopic dermatitis highlights differences between Singaporean Chinese and European populations. Br J Dermatol.2011;165:106-14. doi:10.1111/j.1365-2133.2011.10331.x [ Links ]

48. Margolis DJ, Apter AJ, Gupta J, Hoffstad O, Papadopoulos M, Campbell LE, et al. The persis-tence of atopic dermatitis and filaggrin (FLG) mutations in a US longitudinal cohort. J Allergy Clin Immunol. 2012;130:912-17. doi:10.1016/j.jaci.2012.07.008 [ Links ]

49. Margolis DJ, Gupta J, Apter AJ, Hoffstad O, Papadopoulos M, Rebbeck TR, et al. Exome Se-quencing of Filaggrin and Related Genes in African-American Children with Atopic Dermatitis. J Invest Dermatol. 2014;134:2272-4. doi:10.1038/jid.2014.126 [ Links ]

50. Margolis DJ, Mitra N, Wubbenhorst B, D'Andrea K, Kraya AA, Hoffstad O, et al. Association of Filaggrin Loss-of-Function Variants With Race in Children With Atopic Dermatitis. JAMA Dermatol. 2019;155:1269-76. doi:10.1001/jamadermatol.2019.1946.doi:10.1001/jama-dermatol.2019.1946 [ Links ]

51. Luukkonen T, Kiiski V, Ahola M, Mandelin J, Virtanen H, Pöyhönen M, et al. The Value of FLG Null Mutations in Predicting Treatment Response in Atopic Dermatitis: An Observational Study in Finnish Patients. Acta Derm Venereol.2017;97:456-63. doi:10.2340/00015555-2578 [ Links ]

52. Lowe AJ, Lee B, Orchard D, King E, Abramson MJ, Allen KJ, et al. Palm reading and water divining: A cross-sectional study of the accuracy of palmar hyperlinearity and transepidermal water loss to identify individuals with a filaggrin gene null mutation. J Am Acad Dermatol. 2020;83:1186-8. doi:10.1016/j.jaad.2020.01.08 [ Links ]

53. Leung DYM, Calatroni A, Zaramela LS, LeBeau PK, Dyjack N, Brar K, et al. The nonlesional skin surface distinguishes atopic dermatitis with food allergy as a unique endotype. Sci Transl Med. 2019;11. doi:10.1126/scitranslmed.aav2685 [ Links ]

54. Esparza-Gordillo J, Weidinger S, Fölster-Holst R, Bauerfeind A, Ruschendorf F, Patone G, et al. A common variant on chromosome 11q13 is associated with atopic dermatitis. Nat Genet. 2009;41:596-601. doi:10.1038/ng.347 [ Links ]

55. Weidinger S, O’Sullivan M, Illig T, Baurecht H, Depner M, Rodriguez E, et al. Filaggrin mutations, atopic eczema, hay fever, and asthma in children. J Allergy Clin Immunol. 2008;121:1203-9. doi:10.1016/j.jaci.2008.02.014 [ Links ]

56. Wu J, Guttman-Yassky E. Efficacy of biologics in atopic dermatitis. Expert Opin Biol Ther. 2020;20:525-38. doi:10.1080/14712598.2020.1722998 [ Links ]

57. Guttman-Yassky E, Bissonnette R, Ungar B, Suárez-Fariñas M, Ardeleanu M, Esaki H, et al. Dupilumab progressively improves systemic and cutaneous abnormalities in patients with atopic dermatitis. J Allergy Clin Immunol.2019;143:155-72. doi:10.1016/j.jaci.2018.08.022 [ Links ]

58. Pavel AB, Song T, Kim HJ, Del Duca E, Krueger JG, Dubin C, et al. Oral Janus kinase/SYK inhibition (ASN002) suppresses inflammation and improves epidermal barrier markers in patients with atopic dermatitis. J Allergy Clin Immunol.2019;144:1011-24. doi:10.1016/j. jaci.2019.07.013 [ Links ]

59. Ishida-Yamamoto A, Igawa S. The biology and regulation of corneodesmosomes. Cell Tissue Res. 2015;360:477-82. doi:10.1007/s00441-014-2037-z [ Links ]

60. Rawlings AV, Voegeli R. Stratum corneum proteases and dry skin conditions. Cell Tissue Res. 2013;351:217-35. doi:10.1007/s00441-012-1501-x [ Links ]

61. Ishida-Yamamoto A, Igawa S, Kishibe M. Order and disorder in corneocyte adhesion. J Dermatol. 2011 ;38:645-54. doi:10.1111/j.1346-8138.2011.01227.x [ Links ]

62. Lee UH, Kim BE, Kim DJ, Cho YG, Ye YM, Leung DY. Atopic dermatitis is associated with reduced corneodesmosin expression: role of cytokine modulation and effects on viral penetration. Br J Dermatol. 2017;176:537-40. doi:10.1111/bjd.15010 [ Links ]

63. Hatano Y, Adachi Y, Elias PM, Crumrine D, Sakai T, Kurahashi R, et al. The Th2 cytokine, interleukin-4, abrogates the cohesion of normal stratum corneum in mice: implications for pathogenesis of atopic dermatitis. Exp Dermatol.2013;22:30-5. doi:10.1111/exd.12047 [ Links ]

64. Furio L, Hovnanian A. Netherton syndrome: defective kallikrein inhibition in the skin leads to skin inflammation and allergy. Biol Chem. 2014;395:945-58. doi:10.1515/hsz-2014-0137 [ Links ]

65. Zhao LP, Di Z, Zhang L, Wang L, Ma L, Lv Y, et al. Association of SPINK5 gene polymorphisms with atopic dermatitis in Northeast China. J Eur Acad Dermatology Venereol. 2012;26:572-7. doi:10.1111/j.1468-3083. 2011 .04120.x [ Links ]

66. Kusunoki T, Okafuji I, Yoshioka T, Saito M, Nishikomori R, Heike T, et al. SPINK5 polymorphism is associated with disease severity and food allergy in children with atopic dermatitis. J Allergy Clin Immunol. 2005;115:636-8. doi:10.1016/j.jaci.2004.12.1114 [ Links ]

67. Li Y, Li Y, Li W, Guo X, Zhou S, Zheng H. Genetic polymorphisms in serine protease inhibitor Kazal-type 5 and risk of atopic dermatitis. Medicine. 2020;99:e21256. doi:10.1097/MD.0000000000021256 [ Links ]

68. Voegeli R, Rawlings AV, Breternitz M, Doppler S, Schreier T, Fluhr JW. Increased stratum corneum serine protease activity in acute eczematous atopic skin. Br J Dermatol. 2009;161:70-7. doi:10.1111/j.1365-2133.2009.09142.x [ Links ]

69. Nomura H, Suganuma M, Takeichi T, Kono M, Isokane Y, Sunagawa K, et al. Multifaceted analyses of epidermal serine protease activity in patients with atopic dermatitis. Int J Mol Sci. 2020;21:913. doi:10.3390/ijms21030913 [ Links ]

70. Takai T. TSLP Expression: Cellular Sources, Triggers, and Regulatory Mechanisms. Allergol Int. 2012;61:3-17. doi:10.2332/allergolint.11-RAI-0395 [ Links ]

71. Wohlrab J, Gebert A, Neubert RH. Lipids in the Skin and pH. Curr Probl Dermatol. 2018;54:64-70. doi:10.1159/000489519 [ Links ]

72. Elias PM. Stratum Corneum Defensive Functions: An Integrated View. J Invest Dermatol. 2005;125:183-200. doi:10.1111/j.0022-202X.2005.23668.x [ Links ]

73. van Smeden J, Janssens M, Gooris GS, Bouwstra JA. The important role of stratum corneum lipids for the cutaneous barrier function. Biochim Biophys Acta - Mol Cell Biol Lipids. 2014;1841:295-313. doi:10.1016/j.bbalip.2013.11.006 [ Links ]

74. Janssens M, van Smeden J, Gooris GS, Bras W, Portale G, Caspers PJ, et al. Lamellar lipid organization and ceramide composition in the stratum corneum of patients with atopic eczema. J Invest Dermatol. 2011 ;131:2136-8. doi:10.1038/jid.2011.175 [ Links ]

75. Berdyshev E, Goleva E, Bronova I, Dyjack N, Rios C, Jung J, et al. Lipid abnormalities in atopic skin are driven by type 2 cytokines. JCI Insight. 2018;3:e98006. doi:10.1172/jci.insight.98006 [ Links ]

76. Rabionet M, Gorgas K, Sandhoff R. Ceramide synthesis in the epidermis. Biochim Biophys Acta Mol Cell Biol Lipids. 2014;1841:422-34. doi:10.1016/j.bbalip.2013.08.011 [ Links ]

77. Boer DEC, van Smeden J, Al-Khakany H, Melnik E, van Dijk R, Absalah S, et al. Skin of atopic dermatitis patients shows disturbed β-glucocerebrosidase and acid sphingomyelinase activity that relates to changes in stratum corneum lipid composition. Biochim Biophys Acta - Mol Cell Biol Lipids. 2020;1865:158673. doi:10.1016/j.bbalip.2020.158673 [ Links ]

78. Pullmannová P, Pavlíková L, Kováčik A, Maixner J, Vávrová K. Permeability and microstructure of model stratum corneum lipid membranes containing ceramides with long (C16) and very long (C24) acyl chains. Biophys Chem.2017;224:20-31. doi:10.1016/j.bpc.2017.03.004 [ Links ]

79. Elias PM. Lipid abnormalities and lipid-based repair strategies in atopic dermatitis. Biochim Biophys Acta - Mol Cell Biol Lipids. 2014;1841:323-30. doi:10.1016/j.bbalip.2013.10.001 [ Links ]

80. Danso M, Boiten W, van Drongelen V, Gmelig Meijling K, Gooris G, El Ghalbzouri A, et al. Altered expression of epidermal lipid bio-synthesis enzymes in atopic dermatitis skin is accompanied by changes in stratum corneum lipid composition. J Dermatol Sci. 2017;88:57-66. doi:10.1016/j.jdermsci.2017.05.005 [ Links ]

81. Toncic RJ, Jakasa I, Hadzavdic SL, Goorden SM, Vlugt KJG, Stet FS,et al. Altered levels of sphingosine, sphinganine and their ceramides in atopic dermatitis are related to skin barrier function, disease severity and local cytokine milieu. Int J Mol Sci. 2020;21:1958. doi:10.3390/ijms21061958 [ Links ]

82. Hatano Y, Katagiri K, Arakawa S, Fujiwara S. Interleukin-4 depresses levels of transcripts for acid-sphingomyelinase and glucocerebrosidase and the amount of ceramide in acetone-wounded epidermis, as demonstrated in a living skin equivalent. J Dermatol Sci. 2007;47:45-7. doi:10.1016/j.jdermsci.2007.02.010 [ Links ]

83. Bäsler K, Bergmann S, Heisig M, Naegel A, Zorn-Kruppa M, Brandner JM. The role of tight junctions in skin barrier function and dermal absorption. J Control Release. 2016;242:105-18. doi:10.1016/j.jconrel.2016.08.007 [ Links ]

84. Kubo A, Nagao K, Yokouchi M, Sasaki H, Amagai M. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J Exp Med. 2009;206:2937-46. doi:10.1084/jem.20091527 [ Links ]

85. Zaniboni MC, Samorano LP, Orfali RL, Aoki V. Skin barrier in atopic dermatitis: beyond filaggrin. An Bras Dermatol. 2016;91:472-8. doi:10.1590/abd1806-4841.20164412 [ Links ]

86. Kezic S, Novak N, Jakasa I, Jungersted JM, Simon M, Brandner JM, et al. Skin barrier in atopic dermatitis. Front Biosci. 2014;19:542-56. doi: 10.2741/4225. [ Links ]

87. Niessen CM. Tight Junctions/Adherens Junctions: Basic Structure and Function. J Invest Dermatol. 2007;127:2525-32. doi:10.1038/sj.jid.5700865 [ Links ]

88. Tokumasu R, Tamura A, Tsukita S. Time- and dose-dependent claudin contribution to biological functions: Lessons from claudin-1 in skin. Tissue Barriers. 2017;5:e1336194. doi:10.1080/21 688370.2017.1336194 [ Links ]

89. Benedetto A De, Rafaels NM, Mcgirt LY, Ivanov AI, Georas SN, Cheadle C, et al. Tight junction defects in patients with atopic dermatitis. J Allergy Clin Immunol. 2011 ;127:773-86. [ Links ]

90. Tokumasu R, Yamaga K, Yamazaki Y, Murota H, Suzuki K, Tamura A,et al. Dose-dependent role of claudin-1 in vivo in orchestrating features of atopic dermatitis. Proc Natl Acad Sci U S A. 2016;113:E4061-8. doi:10.1073/pnas.1525474113 [ Links ]

91. Furuse M, Hata M, Furuse K, Yoshida Y, Haratake A, Sugitani Y, et al. Claudin-based tight junctions are crucial for the mammalian epidermal barrier. J Cell Biol. 2002;156:1099-111. doi:10.1083/jcb.200110122 [ Links ]

92. Gruber R, Börnchen C, Rose K, Daubmann A, Volksdorf T, Wladykowski E, et al. Diverse Regulation of Claudin-1 and Claudin-4 in Atopic Dermatitis. Am J Pathol. 2015;185:2777-89. doi:10.1016/j.ajpath.2015.06.021 [ Links ]

93. Bergmann S, von Buenau B, Vidal-Y-Sy S, Haftek M, Wladykowski E, Houdek P, et al. Claudin-1 decrease impacts epidermal barrier function in atopic dermatitis lesions dose-dependently. Sci Rep. 2020;10:2024. doi:10.1038/s41598-020-58718-9 [ Links ]

94. Yuki T, Tobiishi M, Kusaka-Kikushima A, Ota Y, Tokura Y. Impaired Tight Junctions in Atopic Dermatitis Skin and in a Skin-Equivalent Model Treated with Interleukin-17. Koval M, ed. PLoS One. 2016;11:e0161759. doi:10.1371/journal.pone.0161759 [ Links ]

95. Asad S, Winge MC, Wahlgren CF, Bilcha KD, Nordenskjöld M, Taylan F, et al. The tight junction gene Claudin-1 is associated with atopic dermatitis among Ethiopians. J Eur Acad Dermatology Venereol. 2016;30:1939-41. doi:10.1111/jdv.13806 [ Links ]

96. Yu HS, Kang MJ, Kwon JW, Lee SY, Lee E, Yang SI, et al. Claudin-1 polymorphism modifies the effect of mold exposure on the development of atopic dermatitis and production of IgE. J Allergy Clin Immunol. 2015;135:827-30.e5. doi:10.1016/j.jaci.2014.10.040 [ Links ]

97. Ryu WI, Lee H, Bae HC, Jeon J, Ryu HJ, Kim J, et al. IL-33 down-regulates CLDN1 expres-sion through the ERK/STAT3 pathway in keratinocytes. J Dermatol Sci. 2018;90:313-22. doi:10.1016/j.jdermsci.2018.02.017 [ Links ]

98. De Benedetto A, Slifka MK, Rafaels NM, Kuo IH, Georas SN, Boguniewicz M, et al. Reductions in claudin-1 may enhance susceptibility to herpes simplex virus 1 infections in atopic dermatitis. J Allergy Clin Immunol. 2011 ;128:242-6.e5. doi:10.1016/j.jaci. 2011 .02.014 [ Links ]

99. Kumar H, Kawai T, Akira S. Pathogen Recognition by the Innate Immune System. Int Rev Immunol. 2011 ;30:16-34. doi:10.3109/08830185.2010.529976 [ Links ]

100. Chambers ES, Vukmanovic-Stejic M. Skin barrier immunity and ageing. Immunology. 2020;160:116-25. doi:10.1111/imm.13152 [ Links ]

101. Niyonsaba F, Kiatsurayanon C, Chieosilapatham P, Ogawa H. Friends or Foes? Host defense (antimicrobial) peptides and proteins in human skin diseases. Exp Dermatol. 2017;26:989-98. doi:10.1111/exd.13314 [ Links ]

102. Chieosilapatham P, Ogawa H, Niyonsaba F. Current insights into the role of human β-defensins in atopic dermatitis. Clin Exp Immunol. 2017;190:155-66. doi:10.1111/cei.13013 [ Links ]

103. Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, et al. Endogenous Antimi-crobial Peptides and Skin Infections in Atopic Dermatitis. N Engl J Med. 2002;347:1151-60. doi:10.1056/NEJMoa021481 [ Links ]

104. Nomura I, Goleva E, Howell MD, Hamid QA, Ong PY, Hall CF, et al. Cytokine milieu of atopic dermatitis, as compared to psoriasis, skin prevents induction of innate immune response genes. J Immunol. 2003;171:3262-9. doi:10.4049/jimmunol.171.6.3262 [ Links ]

105. Hata TR, Kotol P, Boguniewicz M, Hata TR, Kotol P, Boguniewicz M, et al. History of eczema herpeticum is associated with the inability to induce human β-defensin (HBD)-2, HBD-3 and cathelicidin in the skin of patients with atopic dermatitis. Br J Dermatol. 2010;163:659-61. doi:10.1111/j.1365-2133.2010.09892.x [ Links ]

106. Rieg S, Steffen H, Seeber S, Humeny A, Kalbacher H, Stevanovic S, et al. Deficiency of dermcidinderived antimicrobial peptides in sweat of patients with atopic dermatitis correlates with an impaired innate defense of human skin in vivo. J Immunol. 2005;174:8003-10. doi:10.4049/jimmunol.174.12.8003 [ Links ]

107. Schwarz T. Immunology. In: Bolognia JL, Schaffer JV, Cerroni L, editors. Dermatology. 4th Ed. Amsterdam: Elsevier; 2018: p. 81-99. [ Links ]

108. Doherty TA, Broide DH. Group 2 innate lymphoid cells: new players in human allergic diseases. J Investig Allergol Clin Immunol. 2015;25:1-11. [ Links ]

109. Bao K, Reinhardt RL. The differential expression of IL-4 and IL-13 and its impact on type-2 immunity. Cytokine. 2015;75:25-37. doi:10.1016/j.cyto.2015.05.008 [ Links ]

110. Rafei-Shamsabadi DA, Klose CS, Halim TY, Tanriver Y, Jakob T. Context Dependent Role of Type 2 Innate Lymphoid Cells in Allergic Skin Inflammation. Front Immunol. 2019;10:1-14. doi:10.3389/fimmu.2019.02591 [ Links ]

111. Brough HA, Nadeau KC, Sindher SB, Alkotob SS, Chan S, Bahnson HT, et al. Epicutaneous sensitization in the development of food allergy: What is the evidence and how can this be prevented? Allergy. 2020;75:2185-205. doi:10.1111/all.14304 [ Links ]

112. Wang V, Boguniewicz J, Boguniewicz M, Ong PY. The infectious complications of atopic dermatitis. Ann Allergy Asthma Immunol. 2021;126:3-12. doi:10.1016/j.anai.2020.08.002 [ Links ]

113. Williams MR, Gallo RL. The role of the skin microbiome in atopic dermatitis. Curr Allergy Asthma Rep. 2015;15:65. doi:10.1007/s11882-015-0567-4 [ Links ]

114. Roll A, Cozzio A, Fischer B, Schmid-Grendelmeier P. Microbial colonization and atopic dermatitis. Curr Opin Allergy Clin Immunol. 2004;4:373-8. doi:10.1097/00130832-200410000-00008 [ Links ]

115. Hill SE, Yung A, Rademaker M. Prevalence of Staphylococcus aureus and antibiotic resistance in children with atopic dermatitis: A New Zealand experience. Australas J Dermatol. 2011 ;52:27-31.doi:10.1111/j.1440-0960.2010.00714.x [ Links ]

116. Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med. 2017;9:eaah4680. doi:10.1126/scitranslmed.aah4680 [ Links ]

117. Kim J, Kim BE, Ahn K, Leung DY. Interactions between atopic dermatitis and Staphylococ-cus aureus infection: clinical implications. Allergy Asthma Immunol Res. 2019;11:593-603. doi:10.4168/aair.2019.11.5.593 [ Links ]

118. Feuillie C, Vitry P, McAleer MA, Kezic S, Irvine AD, Geoghegan JA, et al. Adhesion of Staphylo-coccus aureus to corneocytes from atopic dermatitis patients is controlled by natural moisturizing factor levels. MBio. 2018;9: e01184-18. doi:10.1128/mBio.01184-18 [ Links ]

119. Geoghegan JA, Irvine AD, Foster TJ. Staphylococcus aureus and atopic dermatitis: a complex and evolving relationship. Trends Microbiol. 2018;26:484-97. doi:10.1016/j.tim.2017.11.008 [ Links ]

120. Kong HH, Oh J, Deming C, Conlan S, Grice EA, Beatson MA, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22:850-9. doi:10.1101/gr.131029.111 [ Links ]

121. Gueniche A, Knaudt B, Schuck E, Volz T, Bastien P, Martin R, et al. Effects of nonpathogenic gram-negative bacterium Vitreoscilla filiformis lysate on atopic dermatitis: a prospective, randomized, double-blind, placebo-controlled clinical study. Br J Dermatol. 2008;159:1357-63. doi:10.1111/j.1365-2133.2008.08836.x [ Links ]

122. Volz T, Skabytska Y, Guenova E, Chen KM, Frick JS, Kirschning CJ, et al. Nonpathogenic bacteria alleviating atopic dermatitis inflammation induce IL-10-producing dendritic cells and regulatory Tr1 Cells. J Invest Dermatol.2014;134:96-104. doi:10.1038/jid.2013.291 [ Links ]

123. Błazewicz I, Jaskiewicz M, Piechowicz L, Neubauer D, Nowicki RJ, Kamysz W, et al. Activity of an-timicrobial peptides and conventional antibiotics against superantigen positive Staphylococcus aureus isolated from patients with atopic dermatitis. Adv Dermatol Allergol. 2018;35:74-82. doi:10.5114/ada.2018.62141 [ Links ]

124. Gould HJ, Takhar P, Harries HE, Chevretton E, Sutton BJ. The Allergic March from Staphylo-coccus aureus Superantigens to Immunoglobulin E. In: Marone G, editor. Superantigens and Superallergens. Berlin: KARGER; 2007. p.106-36. doi:10.1159/000100861 [ Links ]

125. Tsilochristou O, du Toit G, Sayre PH, Roberts G, Lawson K, Sever ML, et al. Association of Staphylococcus aureus colonization with food allergy occurs independently of eczema severity. J Allergy Clin Immunol. 2019;144:494-503. doi:10.1016/j.jaci.2019.04.025 [ Links ]

126. Riechelmann H, Essig A, Deutschle T, Rau A, Rothermel B, Weschta M. Nasal carriage of Staphylococcus aureus in house dust mite allergic patients and healthy controls. Allergy. 2005;60:1418-23. doi:10.1111/j.1398-9995.2005.00902.x [ Links ]

127. Shiomori T, Yoshida S, Miyamoto H, Makishima K. Relationship of nasal carriage of Staphylococcus aureus to pathogenesis of perennial allergic rhinitis. J Allergy Clin Immunol. 2000;105:449-54. doi:10.1067/mai.2000.104256 [ Links ]

128. Zeldin Y, Weiler Z, Cohen A, Kalinin M, Schlesinger M, Kidon M, et al. Efficacy of nasal Staphylo-coccus aureus eradication by topical nasal mupirocin in patients with perennial allergic rhinitis. Ann Allergy Asthma Immunol. 2008;100:608-11. doi:10.1016/S1081-1206(10)60053-1 [ Links ]

129. Jones AL, Curran-Everett D, Leung DYM. Food allergy is associated with Staphylococcus aureus colonization in children with atopic dermatitis. J Allergy Clin Immunol. 2016;137:1247-1248. e3. doi:10.1016/j.jaci.2016.01.010 [ Links ]

130. Yu J, Oh MH, Park JU, Myers AC, Dong C, Zhu Z, et al. Epicutaneous exposure to Staphylococ-cal superantigen enterotoxin B enhances allergic lung inflammation via an IL-17A dependent mechanism. PLoS One. 2012;7:e39032. doi:10.1371/journal.pone.0039032 [ Links ]

131. Ganeshan K, Neilsen CV, Hadsaitong A, Schleimer RP, Luo X, Bryce PJ. Impairing oral tolerance promotes allergy and anaphylaxis: A new murine food allergy model. J Allergy Clin Immunol. 2009;123:231-8.e4. doi:10.1016/j.jaci.2008.10.011 [ Links ]

132. Celebi Sözener Z, Cevhertas L, Nadeau K, Akdis M, Akdis CA. Environmental factors in epi-thelial barrier dysfunction. J Allergy Clin Immunol. 2020;145:1517-28. doi:10.1016/j. jaci.2020.04.024 [ Links ]

133. Nakamura T, Hirasawa Y, Takai T, Mitsuishi K, Okuda M, Kato T, et al. Reduction of skin barrier function by proteolytic activity of a recombinant house dust mite major allergen Der f 1. J Invest Dermatol. 2006;126:2719-23. doi:10.1038/sj.jid.5700584 [ Links ]

134. Takai T, Ikeda S. Barrier dysfunction caused by environmental proteases in the pathogenesis of allergic diseases. Allergol Int. 2011 ;60:25-35. [ Links ]

135. Jeong SK, Kim HJ, Youm JK, Ahn SK, Choi EH, Sohn MH, et al. Mite and cockroach allergens activate protease-activated receptor 2 and delay epidermal permeability barrier recovery. J Invest Dermatol. 2008;128:1930-9. doi:10.1038/jid.2008.13 [ Links ]

136. Weidinger S, Novak N. Atopic dermatitis. Lancet. 2016;387:1109-22. doi:10.1016/S0140-6736(15)00149-X [ Links ]

137. Sarri CA, Roussaki-Schulze A, Vasilopoulos Y, Zafiriou E, Patsatsi A, Stamatis C, et al. Netherton syndrome: a genotype-phenotype review. Mol Diagn Ther. 2017;21:137-52. doi:10.1007/s40291-016-0243-y [ Links ]

138. Hovnanian A. Netherton syndrome: skin inflammation and allergy by loss of protease inhibi-tion. Cell Tissue Res. 2013;351:289-300. doi:10.1007/s00441-013-1558-1 [ Links ]

139. Mallet A, Kypriotou M, George K, Leclerc E, Rivero D, Mazereeuw-Hautier J, et al. Identification of the first nonsense CDSN mutation with expression of a truncated protein causing peeling skin syndrome type B. Br J Dermatol.2013;169:1322-5. doi:10.1111/bjd.12593 [ Links ]

140. Bowden PE. Peeling skin syndrome: genetic defects in late terminal differentiation of the epider-mis. J Invest Dermatol. 2011 ;131:561-4. doi:10.1038/jid.2010.434 [ Links ]

141. Arjona Aguilera C, Albarrán Planelles C, Tercedor Sánchez J. Differential Diagnosis of genetic disorders associated with moderate to severe refractory eczema and elevated immunoglobulin E. Actas Dermo-Sifiliográficas 2016;107:116-24. doi:10.1016/j.adengl.2016.01.004 [ Links ]

142. Adhikary PP, Tan Z, Page BDG, Hedtrich S. TSLP as druggable target - a silver-lining for atopic diseases? Pharmacol Ther. 2021;217:107648. doi:10.1016/j.pharmthera.2020.107648 [ Links ]

143. Akasaki S, Matsushita K, Kato Y, Fukuoka A, Iwasaki N, Nakahira M, et al. Murine allergic rhini-tis and nasal T h2 activation are mediated via TSLP- and IL-33-signaling pathways. Int Immunol. 2015;28(2):65-76. doi:10.1093/intimm/dxv055 [ Links ]

144. Kim JH, Bae HC, Ko NY, Lee SH, Jeong SH, Lee H, et al. Thymic stromal lymphopoietin down-regulates filaggrin expression by signal transducer and activator of transcription 3 (STAT3) and extracellular signal-regulated kinase (ERK) phosphorylation in keratinocytes. J Allergy Clin Immunol. 2015;136:205-8.e9. doi:10.1016/j.jaci.2015.04.026 [ Links ]

145. Sano Y, Masuda K, Tamagawa-Mineoka R, Matsunaka H, Murakami Y, Yamashita R, et al. Thymic stromal lymphopoietin expression is increased in the horny layer of patients with atopic dermatitis. Clin Exp Immunol. 2013;171:330-7. doi:10.1111/cei.12021 [ Links ]

146. Gour N, Wills-Karp M. IL-4 and IL-13 signaling in allergic airway disease. Cytokine. 2015;75:68-78. doi:10.1016/j.cyto.2015.05.014 [ Links ]

147. Georas SN, Guo J, De Fanis V, Casolaro U. T-helper cell type-2 regulation in allergic disease. Eur Respir J. 2005;26:1119-37. doi:10.1183/09031936.05.00006005 [ Links ]

148. Noti M, Kim BS, Siracusa MC, Rak GD, Kubo M, Moghaddam AE, et al. Exposure to food allergens through inflamed skin promotes intestinal food allergy through the thymic stromal lymphopoietin-basophil axis. J Allergy Clin Immunol.2014;133:1390-9. doi:10.1016/j. jaci.2014.01.021 [ Links ]

149. Frossard CP, Zimmerli SC, Rincon Garriz JM, Eigenmann PA. Food allergy in mice is modu-lated through the thymic stromal lymphopoietin pathway. Clin Transl Allergy. 2015;6:1-8. doi:10.1186/s13601-016-0090-2 [ Links ]

150. Li Y, Wang W, Lv Z, Li Y, Chen Y, Huang K, et al. Elevated expression of IL-33 and TSLP in the airways of human asthmatics in vivo: a potential biomarker of severe refractory disease. J Immunol. 2018;200:2253-62. doi:10.4049/jimmunol.1701455 [ Links ]

151. Menzies-Gow A, Corren J, Bourdin A, Chupp G, Israel E, Wechsler ME, et al. Tezepelumab in Adults and Adolescents with Severe, Uncontrolled Asthma. N Engl J Med. 2021;384:1800-9. doi:10.1056/NEJMoa2034975 [ Links ]

152. Simpson EL, Parnes JR, She D, Crouch D, Rees W, Mo M, et al. Tezepelumab, an antithymic stromal lymphopoietin monoclonal antibody, in the treatment of moderate to severe atopic dermatitis: A randomized phase 2a clinical trial. J Am Acad Dermatol. 2019;80:1013-21. doi:10.1016/j.jaad.2018.11.059 [ Links ]

153. Hamilton JD, Suárez-Fariñas M, Dhingra N, Cardinale I, Li X, Kostic A, et al. Dupilumab im-proves the molecular signature in skin of patients with moderate-to-severe atopic dermatitis. J Allergy Clin Immunol. 2014;134:1293-1300. [ Links ]

154. Fukuie T, Hirakawa S, Narita M, Nomura I, Matsumoto K, Tokura Y, et al. Potential preventive effects of proactive therapy on sensitization in moderate to severe childhood atopic dermatitis: A randomized, investigator-blinded, controlled study. J Dermatol. 2016;43:1283-92. doi:10.1111/1346-8138.13408 [ Links ]

155. Chalmers JR, Haines RH, Bradshaw LE, Montgomery AA, Thomas KS, Brown SJ, et al. Daily emollient during infancy for prevention of eczema: the BEEP randomised controlled trial. Lancet. 2020;395:962-72. doi:10.1016/S0140-6736(19)32984-8 [ Links ]

156. Skjerven HO, Rehbinder EM, Vettukattil R, LeBlanc M, Granum B, Haugen G, et al. Skin emollient and early complementary feeding to prevent infant atopic dermatitis (PreventADALL): a factorial, multicentre, cluster-randomised trial. Lancet. 2020;395:951-61. doi:10.1016/S0140-6736(19)32983-6 [ Links ]

157. Kelleher MM, Cro S, Van Vogt E, C ornelius V, Lodrup Carlsen KC, Ove Skjerven H, et al. Skincare interventions in infants for preventing eczema and food allergy: A cochrane systema-tic review and individual participant data meta-analysis. Clin Exp Allergy. 2021;51:402-418. doi:10.1111/cea.13847 [ Links ]

158. Simpson EL, Chalmers JR, Hanifin JM, Thomas KS, Cork MJ, McLean WH, et al. Emollient enhancement of the skin barrier from birth offers effective atopic dermatitis prevention. J Allergy Clin Immunol. 2014;134:818-23. doi:10.1016/j.jaci.2014.08.005. [ Links ]

159. Sindher S, Alkotob SS, Shojinaga MN, Hamilton R, Chan S, Cao S, et al. Pilot study measuring transepidermal water loss (TEWL) in children suggests trilipid cream is more effective than a paraffin-based emollient. Allergy. 2020;75:2662-4. doi:10.1111/all.14275 [ Links ]

160. Elias PM, Wakefield JS, Man MQ. Moisturizers versus current and next-generation barrier repair therapy for the management of atopic dermatitis. Skin Pharmacol Physiol. 2019;32:1-7. doi:10.1159/000493641 [ Links ]

161. Lowe A, Su J, Tang M, Matheson M, Allen KJ, Varigos G,et al. PEBBLES study protocol: a randomised controlled trial to prevent atopic dermatitis, food allergy and sensitisation in infants with a family history of allergic disease using a skin barrier improvement strategy. BMJ Open. 2019;9:e024594. doi:10.1136/bmjopen-2018-024594 [ Links ]

162. Czarnowicki T, Krueger JG, Guttman-Yassky E. Novel concepts of prevention and treatment of atopic dermatitis through barrier and immune manipulations with implications for the atopic march. J Allergy Clin Immunol. 2017;139:1723-34. doi:10.1016/j.jaci.2017.04.004 [ Links ]

1Conflicts of Interest: The authors have no conflicts of interest to declare. Financing Support: This work has not received any contribution, grant or scholarship. Provenance and Peer Review: Not commissioned; externally peer reviewed. Conflitos de Interesse: Os autores declaram não possuir conflitos de interesse. Suporte Financeiro: O presente trabalho não foi suportado por nenhum subsídio ou bolsa. Proveniência e Revisão por Pares: Não comissionado; revisão externa por pares

2© Author(s) (or their employer(s)) 2021 SPDV Journal. Re-use permitted under CC BY-NC. No commercial re-use. © Autor (es) (ou seu (s) empregador (es)) 2021 Revista SPDV. Reutilização permitida de acordo com CC BY-NC. Nenhuma reutilização comercial

Received: April 30, 2021; Accepted: August 16, 2021

Corresponding Author: Tiago Fernandes Gomes Address: Dermatology Department - Centro Hospitalar de Leiria Rua das Olhalvas - 2410-197, Leiria - Portugal E-mail: tiagofgomesd@gmail.com

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License