Content » Vol 100, June

Review

Disease Mechanisms in Atopic Dermatitis: A Review of Aetiological Factors

Jacob P. Thyssen1, Maria Rasmussen Rinnov1 and Christian Vestergaard2

1Department of Dermatology and Allergy, Herlev and Gentofte Hospital, Hellerup, and 2Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark

ABSTRACT

Atopic dermatitis is a prevalent inflammatory skin condition characterized by itch and dry skin, which affects 15–20% of children and 3–5% of adults. This article reviews epidemiological, clinical and experimental data to provide an overview of the most important disease mechanisms in atopic dermatitis. Genetic predisposition, environmental insults, atopic triggers, complex host immune response and skin barrier changes, and altered skin microbiota are discussed. Whilst our understanding of atopic dermatitis has improved dramatically in recent years, many basic aspects are still not understood. Further research is needed to fully understand this complex skin disease.

Key words: atopic dermatitis; aetiology; pathophysiology; pathomechanism; risk.

Accepted May 7, 2020; Epub ahead of print May 15, 2020

Acta Derm Venereol 2020; 100; adv00162.

Corr: Jacob P. Thyssen, Department of Dermatology and Allergy, Herlev and Gentofte Hospital, Hospitalsvej 15, DK-2900 Hellerup, Denmark. E-mail: jacob.p.thyssen@regionh.dk

SIGNIFICANCE

The aetiology of atopic dermatitis is poorly understood, but studies have provided insight into the pathomechanism, which may improve the prediction of onset of atopic dermatitis and its prophylaxis. This review provides an overview of the pathogenesis and pathomechanism of atopic dermatitis.

INTRODUCTION

Atopic dermatitis (AD) is a prevalent inflammatory skin condition characterized by itch and dry skin, which affects 15–20% of children and 3–5% of adults. In large proportions of affected patients AD is chronic or remitting, as shown by epidemiological studies (1).

The pathogenesis of AD is complex and poorly understood. However, in recent years, there has been major advancements in our understanding of the disease mechanism of AD, e.g. through the discovery of common filaggrin gene (FLG) mutations as a strong risk factor for AD, as well as the significant clinical effects of antagonistic therapy against interleukins (IL) 4, 13, 22 and 31.

This review provides a holistic overview of the most important disease mechanisms in AD.

INCIDENCE OF ATOPIC DERMATITIS PEAKS IN EARLY CHILDHOOD

AD predominately begins in early childhood, as indicated by a recent prospective Danish study, which showed that nearly all cases of AD are diagnosed before the age of 7 years (2). It is currently unclear to what degree “late-onset AD” is important in absolute numbers, as studies have shown that patients who present with AD in adulthood may have forgotten about their childhood AD, and that the disease may therefore represent re-activation of previous disease. This notion is strongly emphasized by the finding that approximately 29% of Swedish adults aged 31–42 years with a school health record of AD in childhood did not recall this when asked as adults (3). In asthma, patients with adult onset seem to have different disease mechanisms, and it is possible that this may also be the case for AD. Moreover, the epidemiology of AD may change over time, in concert with new causative exposures. As an example, use of cosmetic products in adolescence has been associated with new onset of AD or recurrence of previous disease (4). Nonetheless, AD normally begins in early childhood; a time where the skin barrier is vulnerable to stress (5–7). This will lead to a decrease in the threshold level against common triggers. As discussed in this review, the skin barrier defect is central to the risk of developing AD.

GENETIC PREDISPOSITION

AD is a clinical syndrome, as indicated by the Hanifin & Rajka criteria for AD (8). These criteria dictate that a certain number of major and minor criteria need to be fulfilled in order to make a diagnosis of AD, including a list of phenotypic and heritable characteristics, such as xerosis, palmar hyperlinearity, keratosis pilaris (all associated with FLG mutations), infra-orbital folds or darkening, as well as facial pallor. Importantly, family predisposition to atopic disease is a major criterion of the Hanifin & Rajka criteria, and twin studies have shown that the heritability of AD is very high (9). The Hanifin & Rajka criteria were unintentionally developed for use in patients with predominately European ancestry, and it is clear that the phenotypic characteristics observed in other ethnic groups are under-represented, and that the criteria may fail when used in these populations (10). An example is the recent observation that pigmentation on the lips is associated with AD in Asian subjects (11).

FLG mutations lead to dry skin, characterized by elevated pH, increased colonization with staphylococci, enhanced penetration and reactivity to chemicals and allergens, and therefore, expectedly, a strongly increased risk of AD (12). Nearly all carriers of FLG mutations with AD develop their skin disease within their first 2 years of life (13), whereas children with later onset do not have these mutations (14). The discovery of FLG mutations provided a new, and much needed, basis for the study of paediatric AD, and led to a strong re-emphasis on primary skin barrier impairment as a crucial factor for the development of AD. Since then, it has been shown that dry skin at birth and at 2 months of age, independent of FLG mutations, can predict AD at 12 months of age, and that daily application of emollients in high-risk infants may reduce the risk of AD (15). Importantly, the normal skin barrier in the 2 first years of infancy is very different from that of adult skin; for example, the levels of natural moisturizing factors (NMF), a degradation product of filaggrin, are much reduced (16). The tendency for AD to begin on the cheeks is also explained by a local, very pronounced, reduction in NMF, which may last until 3 years of age (6). The down-regulation of filaggrin on exposed skin areas, as well as the increased prevalence of FLG mutations in populations that have migrated far from the Equator, is probably explained by evolutionary benefits due to increased synthesis of vitamin D following facilitated penetration of ultraviolet (UV) (17). Importantly, a deficiency of filaggrin, whether primary or secondary, results in increased penetration of allergens and risk of sensitization, which, in turn, may explain the increased risk of allergic asthma, rhinitis and food allergy in carriers of FLG mutations who have AD (18).

ENVIRONMENTAL EXPOSURE

The crucial role of environmental exposure and skin stressors cannot be overemphasized when explaining the aetiology of the AD epidemic. Modern society has resulted in dramatic changes in human exposure, with increased use of, or exposure to, household products, cosmetics, tobacco, processed food, and air pollution, but at the same time reduced exposure to microorganisms and solar irradiation, as a result of increased hygiene, fewer people living together in the same household, and less time spent outside. Epigenetic changes due to environmental changes or insults could explain a large part of the endemic proportions of AD. In support of this theory, large genome-wide association studies have identified only a small proportion of genetic factors associated with AD (19). However, how the environmental changes have influenced the risk of AD at a mechanistic level is largely unknown.

Being born in the autumn or winter in the Northern hemisphere, or being exposed to a dry and cold climate, has been strongly associated with AD (20, 21). This is probably explained by skin exposure to low temperatures, as well as low ambient humidity due to indoor heating, which can negatively affect the skin barrier and result in dermatitis (22). Similarly, bathing infants in hard water may increase the risk of AD, possibly due to increased pH, which, among other aspects, results in premature cleavage of cornedesmosomes (20). Exposure to air pollution and being born in a newly built home have also been associated with AD (23, 24), perhaps because chemicals negatively affect the epidermal barrier. For example, short-term exposure to airborne formaldehyde results in increased water loss from the skin surface (25) in patients with AD, and toluene, a common air pollutant, can directly down-regulate synthesis of filaggrin (26). Interestingly, exposure to solar irradiation, which is normally avoided in infancy, to reduce the risk of skin malignancy, seems to protect against AD (27, 28). This could be explained by the positive effects of sub-erythemogenic doses of UVB irradiation on the skin barrier, which, among other aspects, reduces Staphyloccocus aureus colonization, itch, and T-cell invasion.

EARLY ALTERATIONS IN THE IMMUNE SYSTEM

The crucial role of early-age alterations in immune activity on the development of AD is emphasized by the significantly reduced risk of AD in premature infants (29). Moreover, thymectomy in infancy reduces the risk of AD by 20%, suggesting that removal of the thymus decreases the number of circulating T cells that can act to develop AD (30). In indirect support of this assumption, a study found significantly larger thymus sizes in children with AD compared with controls, although this may also be a consequence of the increased demand for T cells in patients with AD (31). The farm theory suggests that microbial exposure may reduce the risk of diseases mediated by T-helper (Th) cell 2, including AD (32), but it is probably more important for allergic diseases than for AD per se. The finding that neonate exposure to dogs can strongly reduce the risk of AD could be confounded, but it is also possible that changes in the host gut microbiome can affect the tolerance-reactivity balance (33). It is unclear how nutrients and alcohol use in mothers can affect the risk of AD, but is has been suggested that the Th2 skew induced by alcohol intake may lead to a higher prevalence of AD in infants (34). Similarly, nutrients may affect the child’s immune response, but this area is complex, and little evidence exists. Collectively, AD occurs mainly in genetically predisposed individuals who have significant skin barrier impairment and who are exposed to AD triggers (or who are overly protected against the crucial microorganisms that could prevent excessive Th2 skew in childhood) (Fig. 1).


Fig. 1. Theoretical outline of how genetic risk genes and environmental risk exposures interact and may impact the risk of atopic dermatitis (AD). If a child reaches the threshold bar for AD, the disease will manifest. Factors that increase the risk of AD are represented by yellow vertical lines, whereas factors that decrease the risk are represented by green vertical lines. Once AD has manifested, the lines are shown in red.

VICIOUS CYCLE IN ATOPIC DERMATITIS

AD is a skin condition in which primary (or secondary) skin barrier impairment leads to (further) skin inflammation, and in which S. aureus colonization may increase, and in turn may drive both eczema severity and the relentless sensation of itch (35). This leads to scratching and additional barrier impairment, thus creating a vicious cycle. Clinicians attempt to stop this cycle by restoring the skin barrier with emollients, reducing inflammation and itch with use of topical/oral immune suppressants or immune modulating drugs, as well light therapy, and, finally, decreasing the burden of S. aureus by use of disinfectants and antibiotics. Evidence supporting the benefits of emollient use to treat AD is the strongly increased time to subsequent flares in emollient users, and the reduced need for topical corticosteroids (36). However, barrier restoration without simultaneous control of inflammation seems to be inadequate in the treatment of AD (37). Prophylactic use of topical anti-inflammatory agents, e.g. with application twice weekly, works to reduce the risk of new flares (38).

PATHOGENIC ROLE OF STAPHYLOCOCCUS AUREUS

While the exact role of bacteria in the pathogenesis of AD is unclear, colonization with S. aureus is very common in lesional and non-lesional AD skin. Antimicrobial peptides, which work as broad-spectrum antibiotics to kill Gram-negative and Gram-positive bacteria, are reduced in patients with AD, which, in turn, allows bacteria to colonize the skin (39). S. aureus can induce serine protease activity, which will destroy corneodesmosomes, and allow invasion (40). Moreover, the expression of Th2 cytokines is activated by proteases released by S. aureus (41), and S. aureus toxin increases the allergic response by activating mast cells (42), and induces up-regulation of T cells via a superantigen-mediated mechanism (43). S. aureus also release α-toxins, which forms pores in keratinocyte membranes leading to cellular damage (44). Individuals with AD and FLG mutations have a 7-fold higher risk of S. aureus skin infections, in part due to increased pH, but also due to the lack of the direct growth inhibition of the filaggrin proteins (45, 46). The levels of filaggrin degradation products, i.e. NMF, seem to regulate the strength of S. aureus corneocyte adhesion, the first step in skin colonization (47).

SKIN MICROBIOME AND DISEASE CONTROL

While the skin hosts the most diverse commensal community of humans, with over 1,000 different bacterial species, the role of the skin microbiome in AD is poorly understood (48, 49). An animal study showed that filaggrin deficiency and microbial dysbiosis triggered intracellular IL-1α secretion and drove chronic inflammation, hence indicating an important pathogenic role (50). Moreover, following successful treatment of AD, Streptococcus, Propionibacterium, and Corynebacterium species increase in numbers along with microbial diversity (51).

DYSFUNCTIONAL LESIONAL AND NON-LESIONAL SKIN

It is important to understand that non-lesional AD skin is also different from the skin of normal controls (Fig. 2). It shows decreased or altered synthesis of important epidermal proteins, e.g. filaggrin, filaggrin 2, involucrin, loricrin, hornerin, and tight junctions, but also decreased synthesis of antimicrobial peptides and lipids, (52–58) as well as increased expression of high-affinity IgE receptor on dendritic CD1a, along with increased numbers of T cells and their cytokines. Children with AD and food allergy have stratum corneum abnormalities in non-lesional skin that are not found in children with AD and controls without food allergy. Thus, filaggrin and ω-hydroxy fatty acid sphingosine are reduced, and there are important changes in the epidermal lamellar bilayer architecture (59). Thus, skin measurements in non-lesional AD skin show elevated pH, increased water loss from the skin surface, and increased penetration of chemicals (60). Moreover, AD skin displays a reduced reactivity threshold to exogenous stressors, such as skin irritants, allergens and S. aureus, in part due to the creation of resident T-cell populations (61–63). The changes in non-lesional skin are largely determined by disease extent and severity (53), probably reinforcing the impression of AD as a generalized skin disease.


Fig. 2. Important skin barrier changes in atopic dermatitis (AD). Innate and acquired inflammation in AD leads to downregulation and degradation of filaggrin and tight junction proteins, in turn leading to a dry and leaky skin barrier with elevated pH, which allows bacteria to colonize and allergens, irritants and microorganisms to invade. Tight junction reduction further allows antigen presenting cells to move upwards and meet the antigens. Lipid synthesis is compromised at several levels, which acts in concert with protein dysfunction to allow increased loss of water from the skin surface. In an attempt to restore the skin barrier and prevent excessive water loss, acanthosis occurs, often in conjunction with mild spongiosis.

HETEROGENEOUS INFLAMMATORY RESPONSE, DEFICIENT SKIN BARRIER AND EXOGENOUS STRESSORS

Type 2 immunity-associated cytokines, such as IL-4 and IL-13, as well as other cytokines, including, but not limited to, IL-1, IL-17, IL-22, IL-31 IL-33, and thymic stromal lymphopoietin (TSLP) have important roles in AD. It is presently unclear whether significant differences exist between AD skin of children and adults, as well as between different ethnic groups, and to what degree this should affect treatment strategy (64, 65). While certain endotypes of AD are suspected to exist, the heterogeneous cytokine landscape could also, in part, be explained by the crucial pathogenic role of the sustained skin barrier impairment in lesional and non-lesional AD skin. Thus, the continuous bombardment and penetration of microorganisms, chemicals, irritants and allergens into the primary and sustained skin barrier impairment in AD could lead to secretion of various cytokines, and as discussed below, activate the Th1 and Th17 axis in addition to the Th2 axis. The exact immune response would be expected to depend on genetics, age, sites of skin exposure, possible co-infection, climatic effects, and type of elicitor. Interestingly, use of monoclonal antibodies against the IL-4 and IL-13 receptors seems to be slightly less effective in facial skin; an anatomical area which is exposed to environmental pollutants and climatic factors (66).

ATOPIC TRIGGERS

To date, there has been little research into the reactivity to various stressors. A survey in children with AD showed that sweating from exercise was a common exacerbator of AD (67). While the exact mechanisms is unknown (68) and, at least in part, could be explained by the direct effects of heating (69), leaking of sweat into the epidermis due to dysfunctional tight junction function could be relevant (70), as well as obstruction of sweat ducts due to filaggrin deficiency (71). Other well-established triggers for AD include exposure to wool, hot weather, psychological stress and sleep deprivation. Induction of stress leads to scratching behaviour in patients with AD, but not in controls (72). The dysfunctional and partly unresponsive peripheral hypothalamic-pituitary-adrenal axis in AD skin could also be important (73). Moreover, psychological stress reduces the recovery time of the stratum corneum, decreases lipid synthesis, and increases the risk of skin infections (74). Exposure to grass allergens may cause worsening of AD in grass-allergic AD individuals through IL-4 release (75). Contact allergens, e.g. fragrances and certain rubber chemicals, have been shown to elicit Th2 immune activity in patch test reactions, as opposed to many other allergens that elicit Th1 immune response (76, 77). Furthermore, exposure to experimental and environmental contact allergens in patients with AD causes Th2 immune response activity, but Th1 immune response in non-atopic skin (78). How this translates into clinical relevance is currently unclear. A recent study examined the skin immune response to various atopic triggers in individuals with normal skin and found that exposure to hard water is associated with IL-4 secretion in the epidermis (79).

CYTOKINE ANTAGONISM AND THE IMMUNE RESPONSE

It is beyond the scope of this review to describe the immunopathophysiology of AD in detail. Briefly, predominately Th2 (IL-4, IL-5, IL-13, IL-31) and Th22 (IL-22) deviation is observed in acute and chronic AD lesions, which, in turn, down-regulate expression of important skin barrier proteins, such as filaggrin. Innate lymphoid cells also release Th2 cytokines, now increasingly referred to as type 2 immunity. In chronic AD lesions, a parallel activation of the Th1 axis is observed, and in both acute and chronic AD, IL-17 activation can be found (82). Yet, even in healthy skin from patients with AD, there is increased expression of inflammatory cytokines and chemokines, as well as of their receptors, and an increased number of lymphocytes compared with healthy controls, suggesting increased immuno-surveillance in the skin and risk of acute inflammation (53).

Apart from the negative influence on the skin barrier, Th2 inflammation inhibits antimicrobial peptide synthesis and increases S. aureus colonization. The Th2 cells may, in many patients, lead to antibody isotype switching to IgE and recruit mast cells, eosinophils, basophils and dendritic cells. Elevated levels of IgE correlate with AD and atopic co-morbidities, including asthma and food allergies (83). Previously, this has been used to subtype AD into extrinsic AD, where allergic sensitization has taken place, and intrinsic AD, in which patients have normal levels of IgE. However, patients with normal IgE levels may also be sensitized and vice versa. It has even been suggested to use the terms intrinsic factors to describe inborn factors e.g. FLG mutations, Th2 skewing, etc., which affect the skin barrier function or the immune response in terms of AD and extrinsic factors to describe exogenous factors, e.g. S. aureus, detergents, allergens, etc. (82). Interestingly, IgE may target keratinocytes in up to 25% of patients with AD, indicating that IgE may play an important role in impairment of the skin barrier (84).

Regulatory T cells can suppress the Th2 response, and the balance between these 2 cell types is central to development of tolerance. It is not known whether a primary immune-deficiency/imbalance might be the prime cause of AD. Single nucleotide polymorphisms (SNPs) in, for example, ST2 (a member of the interleukin 1 family), IL-13, IL-12, have been reported to be associated with AD, and a huge work in developing a taxonomy for AD subtypes based on serum levels of cytokines has been undertaken (85). A recent work was able to distinguish at least 3 different subtypes of AD, based on analysis of 147 different soluble factors, yet this does not, in itself, show that the immune response is the prime cause of the disease (86). Rather, it indicates that patients with AD have different propensity to react to exogenous stimuli and that, even within the group of patients with AD, this differs slightly and gives rise to different subtypes. The result of this may be the development of personalized medicine for patients with AD (87).

ROLE OF SYSTEMIC INFLAMMATION

Adult patients with AD have significantly elevated levels of circulating cytokines and chemokines (87). While it is intriguing to consider that the systemic inflammation in AD can negatively affect the function of other organs, such as the central nervous system and vascular system, there is currently no convincing evidence to support this. Nonetheless, AD has been associated with anxiety, depression, autism and attention deficit disorders, and it is possible that cytokines may cause a leaky blood–brain barrier and become absorbed into the cerebrospinal compartments and negatively affect cognitive development, by affecting the glia cells and neurogenesis. Decreased sleep quality due to itch is, however, also a major risk factor for ADD and depressive symptoms. The link between asthma and AD is not fully understood, but the shared type 2 immunity and effect of dupilumab on severity of both AD and asthma support that systemic inflammation could play an important role. While some patients with AD experience worsening of their AD during or after asthma attacks, it is unclear whether this is explained by psychological stress or by cytokines reaching the skin.

CONCLUSION

This review highlights some important disease mechanisms of AD. While understanding of AD has improved in recent years, many basic aspects are still not understood. For example, why do AD lesions outside the flexural areas tend to clear once flexural eczema is controlled? Why is AD a flexural disease? What triggers an AD flare? What explains the resolution of AD in the majority of children? What is the role of foods as triggers for AD? Why do AD children have fewer naevi than controls? These are just some of many unanswered questions. In conclusion, more research is needed into this complex skin disease.

ACKNOWLEDGEMENTS

Funding sources: JPT and MRR are financially supported by an unrestricted grant from the Lundbeck Foundation.

Conflicts of interest: JPT has attended advisory boards for Roche, Eli Lilly & Co., Pfizer, Abbvie, LEO Pharma, and Sanofi-Genzyme, been an investigator for Pfizer, Abbvie, Regeneron, Sanofi-Genzyme, and LEO Pharma, and received speaker honorarium from LEO Pharma, Abbvie, Regeneron, and Sanofi-Genzyme. CV has been investigator, speaker, or consultant for Novartis, Abbvie, Sanofi, LeoPharma and Eli Lilly & Co. MRR has no conflicts of interest to declare.

REFERENCES
  1. Margolis JS, Abuabara K, Bilker W, Hoffstad O, Margolis DJ. Persistence of mild to moderate atopic dermatitis. JAMA Dermatol 2014; 150: 593–600.
    View article    Google Scholar
  2. Thorsteinsdottir S, Stokholm J, Thyssen JP, Norgaard S, Thorsen J, Chawes BL, et al. Genetic, clinical, and environmental factors associated with persistent atopic dermatitis in childhood. JAMA Dermatol 2019; 155: 50–57.
    View article    Google Scholar
  3. Moberg C, Meding B, Stenberg B, Svensson A, Lindberg M. Remembering childhood atopic dermatitis as an adult: factors that influence recollection. Br J Dermatol 2006; 155: 557–560.
    View article    Google Scholar
  4. Puangpet P, Lai-Cheong J, McFadden JP. Chemical atopy. Contact Dermatitis 2013; 68: 208–213.
    View article    Google Scholar
  5. Fluhr JW, Darlenski R, Taieb A, Hachem JP, Baudouin C, Msika P, et al. Functional skin adaptation in infancy – almost complete but not fully competent. Exp Dermatol 2010; 19: 483–492.
    View article    Google Scholar
  6. McAleer MA, Jakasa I, Raj N, O’Donnell CPF, Lane ME, Rawlings AV, et al. Early-life regional and temporal variation in filaggrin-derived natural moisturizing factor, filaggrin-processing enzyme activity, corneocyte phenotypes and plasmin activity: implications for atopic dermatitis. Br J Dermatol 2018; 179: 431–441.
    View article    Google Scholar
  7. Wester RC, Noonan PK, Cole MP, Maibach HI. Percutaneous absorption of testosterone in the newborn rhesus monkey: comparison to the adult. Pediat Res 1977; 11: 737–739.
    View article    Google Scholar
  8. Hanifin JM, Rajka G. Diagnostic features of atopic dermatitis. Acta Derm Venereol 1980; Suppl 92: 44–47.
    View article    Google Scholar
  9. Schultz Larsen F. Atopic dermatitis: a genetic-epidemiologic study in a population-based twin sample. J Am Acad Dermatol 1993; 28: 719–723.
    View article    Google Scholar
  10. Cheng R, Guo Y, Huang L, Hao F, Gao X, Bieber T, et al. Current status in diagnosis of atopic dermatitis in China. Allergy 2017; 72: 1277–1278.
    View article    Google Scholar
  11. Kang IH, Jeong KH, Lee MH, Shin MK. Atopic labial pigmentation: a new diagnostic feature in Asian patients with atopic dermatitis. Int J Dermatol 2018; 57: 817–821.
    Google Scholar
  12. Thyssen JP, Kezic S. Causes of epidermal filaggrin reduction and their role in the pathogenesis of atopic dermatitis. J Allergy Clin Immunol 2014; 134: 792–799.
    View article    Google Scholar
  13. Bisgaard H, Simpson A, Palmer CN, Bonnelykke K, McLean I, Mukhopadhyay S, et al. Gene-environment interaction in the onset of eczema in infancy: filaggrin loss-of-function mutations enhanced by neonatal cat exposure. PLoS Med 2008; 5: e131.
    View article    Google Scholar
  14. Rupnik H, Rijavec M, Korosec P. Filaggrin loss-of-function mutations are not associated with atopic dermatitis that develops in late childhood or adulthood. Br J Dermatol 2015; 172: 455–461.
    View article    Google Scholar
  15. Kelleher M, Dunn-Galvin A, Hourihane JO, Murray D, Campbell LE, McLean WH, et al. Skin barrier dysfunction measured by transepidermal water loss at 2 days and 2 months predates and predicts atopic dermatitis at 1 year. J Allergy Clin Immunol 2015; 135: 930–935.e1.
    View article    Google Scholar
  16. Nikolovski J, Stamatas GN, Kollias N, Wiegand BC. Barrier function and water-holding and transport properties of infant stratum corneum are different from adult and continue to develop through the first year of life. J Invest Dermatol 2008; 128: 1728–1736.
    View article    Google Scholar
  17. Thyssen JP. The trade-off: evolutionary benefits of epidermal filaggrin deficiency on cheek skin came at the price of the atopic dermatitis epidemic. Br J Dermatol 2018; 179: 235–236.
    View article    Google Scholar
  18. Rodriguez 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: 1361–1370.e7.
    View article    Google Scholar
  19. Kezic S, Novak N, Jakasa I, Jungersted JM, Simon M, Brandner JM, et al. Skin barrier in atopic dermatitis. Front Biosci (Landmark Ed) 2014; 19: 542–556.
    View article    Google Scholar
  20. Engebretsen KA, Bager P, Wohlfahrt J, Skov L, Zachariae C, Nybo Andersen AM, et al. Prevalence of atopic dermatitis in infants by domestic water hardness and season of birth: Cohort study. J Allergy Clin Immunol 2017; 139: 1568–1574.e1.
    View article    Google Scholar
  21. Silverberg JI, Hanifin J, Simpson EL. Climatic factors are associated with childhood eczema prevalence in the United States. J Invest Dermatol 2013; 133: 1752–1759.
    View article    Google Scholar
  22. McNally NJ, Williams HC, Phillips DR. Atopic eczema and the home environment. Br J Dermatol 2001; 145: 730–736.
    View article    Google Scholar
  23. Wen HJ, Chen PC, Chiang TL, Lin SJ, Chuang YL, Guo YL. Predicting risk for early infantile atopic dermatitis by hereditary and environmental factors. Br J Dermatol 2009; 161: 1166–1172.
    Google Scholar
  24. Hidaka T, Ogawa E, Kobayashi EH, Suzuki T, Funayama R, Nagashima T, et al. The aryl hydrocarbon receptor AhR links atopic dermatitis and air pollution via induction of the neurotrophic factor artemin. Nature Immunol 2017; 18: 64–73.
    Google Scholar
  25. Kim J, Han Y, Ahn JH, Kim SW, Lee SI, Lee KH, et al. Airborne formaldehyde causes skin barrier dysfunction in atopic dermatitis. Br J Dermatol 2016; 175: 357–363.
    Google Scholar
  26. Lee H, Shin JJ, Bae HC, Ryu WI, Son SW. Toluene downregulates filaggrin expression via the extracellular signal-regulated kinase and signal transducer and activator of transcription-dependent pathways. J Allergy Clin Immunol 2017; 139: 355–358.5.
    View article    Google Scholar
  27. Thyssen JP, Zirwas MJ, Elias PM. Potential role of reduced environmental UV exposure as a driver of the current epidemic of atopic dermatitis. J Allergy Clin Immunol 2015; 136: 1163–1169.
    View article    Google Scholar
  28. Rueter K, Jones AP, Siafarikas A, Lim EM, Bear N, Noakes PS, et al. Direct infant UV light exposure is associated with eczema and immune development. J Allergy Clin Immunol 2019; 143: 1012–1020.e2.
    View article    Google Scholar
  29. Barbarot S, Gras-Leguen C, Colas H, Garrot E, Darmaun D, Larroque B, et al. Lower risk of atopic dermatitis among infants born extremely preterm compared with higher gestational age. Br J Dermatol 2013; 169: 1257–1264.
    View article    Google Scholar
  30. Thyssen JP, Andersen YMF, Zhang H, Gislason G, Skov L, Egeberg A. Incidence of pediatric atopic dermatitis following thymectomy: a Danish register study. Allergy 2018; 73: 1741–1743.
    View article    Google Scholar
  31. Olesen AB, Andersen G, Jeppesen DL, Benn CS, Juul S, Thestrup-Pedersen K. Thymus is enlarged in children with current atopic dermatitis. A cross-sectional study. Acta Derm Venereol 2005; 85: 240–243.
    View article    Google Scholar
  32. Flohr C, Yeo L. Atopic dermatitis and the hygiene hypothesis revisited. Curr Probl Dermatol 2011; 41: 1–34.
    View article    Google Scholar
  33. Thorsteinsdottir S, Thyssen JP, Stokholm J, Vissing NH, Waage J, Bisgaard H. Domestic dog exposure at birth reduces the incidence of atopic dermatitis. Allergy 2016; 71: 1736–1744.
    View article    Google Scholar
  34. Halling-Overgaard AS, Hamann CR, Holm RP, Linneberg A, Silverberg JI, Egeberg A, et al. Atopic dermatitis and alcohol use – a meta-analysis and systematic review. J Eur Acad Dermatol Venereol 2018; 32: 1238–1245.
    View article    Google Scholar
  35. Elias PM, Steinhoff M. “Outside-to-inside” (and now back to “outside”) pathogenic mechanisms in atopic dermatitis. J Invest Dermatol 2008; 128: 1067–1070.
    View article    Google Scholar
  36. Wiren K, Nohlgard C, Nyberg F, Holm L, Svensson M, Johannesson A, et al. Treatment with a barrier-strengthening moisturizing cream delays relapse of atopic dermatitis: a prospective and randomized controlled clinical trial. J Eur Acad Dermatol Venereol 2009; 23: 1267–1272.
    View article    Google Scholar
  37. Czarnowicki T, Dohlman AB, Malik K, Antonini D, Bissonnette R, Chan TC, et al. Effect of short-term liver X receptor activation on epidermal barrier features in mild to moderate atopic dermatitis: a randomized controlled trial. Ann Allergy Asthma Immunol 2018; 120: 631–640.e11.
    View article    Google Scholar
  38. Hanifin J, Gupta AK, Rajagopalan R. Intermittent dosing of fluticasone propionate cream for reducing the risk of relapse in atopic dermatitis patients. Br J Dermatol 2002; 147: 528–537.
    View article    Google Scholar
  39. Schauber J, Gallo RL. Antimicrobial peptides and the skin immune defense system. J Allergy Clin Immunol 2009; 124: R13–18.
    View article    Google Scholar
  40. Williams MR, Nakatsuji T, Sanford JA, Vrbanac AF, Gallo RL. Staphylococcus aureus induces increased serine protease activity in keratinocytes. J Invest Dermatol 2017; 137: 377–384.
    View article    Google Scholar
  41. Nakatsuji T, Chen TH, Two AM, Chun KA, Narala S, Geha RS, et al. Staphylococcus aureus exploits epidermal barrier defects in atopic dermatitis to trigger cytokine expression. J Invest Dermatol 2016; 136: 2192–2200.
    View article    Google Scholar
  42. Nakamura Y, Oscherwitz J, Cease KB, Chan SM, Munoz-Planillo R, Hasegawa M, et al. Staphylococcus delta-toxin induces allergic skin disease by activating mast cells. Nature 2013; 503: 397–401.
    View article    Google Scholar
  43. Skov L, Olsen JV, Giorno R, Schlievert PM, Baadsgaard O, Leung DY. Application of Staphylococcal enterotoxin B on normal and atopic skin induces up-regulation of T cells by a superantigen-mediated mechanism. J Allergy Clin Immunol 2000; 105: 820–826.
    View article    Google Scholar
  44. Song L, Hobaugh MR, Shustak C, Cheley S, Bayley H, Gouaux JE. Structure of staphylococcal alpha-hemolysin, a heptameric transmembrane pore. Science 1996; 274: 1859–1866.
    View article    Google Scholar
  45. Miajlovic H, Fallon PG, Irvine AD, Foster TJ. Effect of filaggrin breakdown products on growth of and protein expression by Staphylococcus aureus. J Allergy Clin Immunol 2010; 126: 1184–1190.e3.
    View article    Google Scholar
  46. Cai SC, Chen H, Koh WP, Common JE, van Bever HP, McLean WH, et al. Filaggrin mutations are associated with recurrent skin infection in Singaporean Chinese patients with atopic dermatitis. Br J Dermatol 2012; 166: 200–203.
    View article    Google Scholar
  47. Feuillie C, Vitry P, McAleer MA, Kezic S, Irvine AD, Geoghegan JA, et al. Adhesion of staphylococcus aureus to corneocytes from atopic dermatitis patients is controlled by natural moisturizing factor levels. mBio 2018; 9. pii: e01184–18.
    View article    Google Scholar
  48. Bjerre RD, Bandier J, Skov L, Engstrand L, Johansen JD. The role of the skin microbiome in atopic dermatitis: a systematic review. Br J Dermatol 2017; 177: 1272–1278.
    View article    Google Scholar
  49. Polkowska-Pruszynska B, Gerkowicz A, Krasowska D. The gut microbiome alterations in allergic and inflammatory skin diseases – an update. J Eur Acad Dermatol Venereol 2020; 34: 455–464.
    View article    Google Scholar
  50. Archer NK, Jo JH, Lee SK, Kim D, Smith B, Ortines RV, et al. Injury, dysbiosis, and filaggrin deficiency drive skin inflammation through keratinocyte IL-1alpha release. J Allergy Clin Immunol 2019; 143: 1426–1443.e6.
    View article    Google Scholar
  51. 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: 850–859.
    View article    Google Scholar
  52. Pellerin L, Henry J, Hsu CY, Balica S, Jean-Decoster C, Mechin MC, et al. Defects of filaggrin-like proteins in both lesional and nonlesional atopic skin. J Allergy Clin Immunol 2013; 131: 1094–1102.
    View article    Google Scholar
  53. Suarez-Farinas M, Tintle SJ, Shemer A, Chiricozzi A, Nograles K, Cardinale I, et al. Nonlesional atopic dermatitis skin is characterized by broad terminal differentiation defects and variable immune abnormalities. J Allergy Clin Immunol 2011; 127: 954–964.e1–4.
    View article    Google Scholar
  54. Jensen JM, Folster-Holst R, Baranowsky A, Schunck M, Winoto-Morbach S, Neumann C, et al. Impaired sphingomyelinase activity and epidermal differentiation in atopic dermatitis. J Invest Dermatol 2004; 122: 1423–1431.
    View article    Google Scholar
  55. Di Nardo A, Wertz P, Giannetti A, Seidenari S. Ceramide and cholesterol composition of the skin of patients with atopic dermatitis. Acta Derm Venereol 1998; 78: 27–30.
    View article    Google Scholar
  56. Semper AE, Heron K, Woollard AC, Kochan JP, Friedmann PS, Church MK, et al. Surface expression of Fc epsilon RI on Langerhans’ cells of clinically uninvolved skin is associated with disease activity in atopic dermatitis, allergic asthma, and rhinitis. J Allergy Clin Immunol 2003; 112: 411–419.
    View article    Google Scholar
  57. Wollenberg A, Wen S, Bieber T. Phenotyping of epidermal dendritic cells: clinical applications of a flow cytometric micromethod. Cytometry 1999; 37: 147–155.
    View article    Google Scholar
  58. Hamid Q, Boguniewicz M, Leung DY. Differential in situ cytokine gene expression in acute versus chronic atopic dermatitis. J Clin Invest 1994; 94: 870–876.
    View article    Google Scholar
  59. 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. pii: eaav2685.
    View article    Google Scholar
  60. Halling-Overgaard AS, Kezic S, Jakasa I, Engebretsen KA, Maibach H, Thyssen JP. Skin absorption through atopic dermatitis skin: a systematic review. Br J Dermatol 2017; 177: 84–106.
    View article    Google Scholar
  61. Brunner PM, Emerson RO, Tipton C, Garcet S, Khattri S, Coats I, et al. Nonlesional atopic dermatitis skin shares similar T-cell clones with lesional tissues. Allergy 2017; 72: 2017–2025.
    View article    Google Scholar
  62. Strange P, Skov L, Lisby S, Nielsen PL, Baadsgaard O. Staphylococcal enterotoxin B applied on intact normal and intact atopic skin induces dermatitis. Arch Dermatol 1996; 132: 27–33.
    View article    Google Scholar
  63. Nassif A, Chan SC, Storrs FJ, Hanifin JM. Abnormal skin irritancy in atopic dermatitis and in atopy without dermatitis. Arch Dermatol 1994; 130: 1402–1407.
    View article    Google Scholar
  64. Zhou L, Leonard A, Pavel AB, Malik K, Raja A, Glickman J, et al. Age-specific changes in the molecular phenotype of patients with moderate-to-severe atopic dermatitis. J Allergy Clin Immunol 2019; 144: 144–156.
    View article    Google Scholar
  65. Chan TC, Sanyal RD, Pavel AB, Glickman J, Zheng X, Xu H, et al. Atopic dermatitis in Chinese patients shows TH2/TH17 skewing with psoriasiform features. J Allergy Clin Immunol 2018; 142: 1013–1017.
    View article    Google Scholar
  66. Silverberg JI, Simpson EL, Ardeleanu M, Thaci D, Barbarot S, Bagel J, et al. Dupilumab provides important clinical benefits to patients with atopic dermatitis who do not achieve clear or almost clear skin according to the Investigator’s Global Assessment: a pooled analysis of data from two phase III trials. Br J Dermatol 2019; 181: 80–87.
    View article    Google Scholar
  67. Williams JR, Burr ML, Williams HC. Factors influencing atopic dermatitis – a questionnaire survey of schoolchildren’s perceptions. Br J Dermatol 2004; 150: 1154–1161.
    View article    Google Scholar
  68. Hendricks AJ, Vaughn AR, Clark AK, Yosipovitch G, Shi VY. Sweat mechanisms and dysfunctions in atopic dermatitis. J Dermatol Sci 2018; 89: 105–111.
    View article    Google Scholar
  69. Murota H, Yamaga K, Ono E, Katayama I. Sweat in the pathogenesis of atopic dermatitis. Allergol Int 2018; 67: 455–459.
    View article    Google Scholar
  70. Yamaga K, Murota H, Tamura A, Miyata H, Ohmi M, Kikuta J, et al. Claudin-3 loss causes leakage of sweat from the sweat gland to contribute to the pathogenesis of atopic dermatitis. J Invest Dermatol 2018; 138: 1279–1287.
    View article    Google Scholar
  71. Rerknimitr P, Tanizaki H, Yamamoto Y, Amano W, Nakajima S, Nakashima C, et al. Decreased filaggrin level may lead to sweat duct obstruction in filaggrin mutant mice. J Invest Dermatol 2017; 137: 248–251.
    View article    Google Scholar
  72. Mochizuki H, Lavery MJ, Nattkemper LA, Albornoz C, Valdes Rodriguez R, Stull C, et al. Impact of acute stress on itch sensation and scratching behaviour in patients with atopic dermatitis and healthy controls. Br J Dermatol 2019; 180: 821–827.
    View article    Google Scholar
  73. Slominski AT, Zmijewski MA, Plonka PM, Szaflarski JP, Paus R. How UV light touches the brain and endocrine system through skin, and why. Endocrinol 2018; 159: 1992–2007.
    View article    Google Scholar
  74. Hall JM, Cruser D, Podawiltz A, Mummert DI, Jones H, Mummert ME. Psychological stress and the cutaneous immune response: roles of the HPA axis and the sympathetic nervous system in atopic dermatitis and psoriasis. Dermatology Res Pract 2012; 2012: 403908.
    Google Scholar
  75. Werfel T, Heratizadeh A, Niebuhr M, Kapp A, Roesner LM, Karch A, et al. Exacerbation of atopic dermatitis on grass pollen exposure in an environmental challenge chamber. J Allergy Clin Immunol 2015; 136: 96–103.e9.
    View article    Google Scholar
  76. Newell L, Polak ME, Perera J, Owen C, Boyd P, Pickard C, et al. Sensitization via healthy skin programs Th2 responses in individuals with atopic dermatitis. J Invest Dermatol 2013; 133: 2372–2380.
    Google Scholar
  77. Dhingra N, Shemer A, Correa da Rosa J, Rozenblit M, Fuentes-Duculan J, Gittler JK, et al. Molecular profiling of contact dermatitis skin identifies allergen-dependent differences in immune response. J Allergy Clin Immunol 2014; 134: 362–372.
    View article    Google Scholar
  78. Correa da Rosa J, Malajian D, Shemer A, Rozenblit M, Dhingra N, Czarnowicki T, et al. Patients with atopic dermatitis have attenuated and distinct contact hypersensitivity responses to common allergens in skin. J Allergy Clin Immunol 2015; 135: 712–720.
    View article    Google Scholar
  79. Engebretsen KA, Kezic S, Jakasa I, Hedengran A, Linneberg A, Skov L, et al. Effect of atopic skin stressors on natural moisturizing factors and cytokines in healthy adult epidermis. Br J Dermatol 2018; 179: 679–688.
    View article    Google Scholar
  80. Guttman-Yassky E, Brunner PM, Neumann AU, Khattri S, Pavel AB, Malik K, et al. Efficacy and safety of fezakinumab (an IL-22 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by conventional treatments: A randomized, double-blind, phase 2a trial. J Am Acad Dermatol 2018; 78: 872–881.e6.
    View article    Google Scholar
  81. Kabashima K, Furue M, Hanifin JM, Pulka G, Wollenberg A, Galus R, et al. Nemolizumab in patients with moderate-to-severe atopic dermatitis: randomized, phase II, long-term extension study. J Allergy Clin Immunol 2018; 142: 1121–1130.e7.
    View article    Google Scholar
  82. Eyerich K, Novak N. Immunology of atopic eczema: overcoming the Th1/Th2 paradigm. Allergy 2013; 68: 974–982.
    View article    Google Scholar
  83. Nygaard U, Hvid M, Johansen C, Buchner M, Folster-Holst R, Deleuran M, et al. TSLP, IL-31, IL-33 and sST2 are new biomarkers in endophenotypic profiling of adult and childhood atopic dermatitis. J Eur Acad Dermatol Venereol 2016; 30: 1930–1938.
    View article    Google Scholar
  84. Altrichter S, Kriehuber E, Moser J, Valenta R, Kopp T, Stingl G. Serum IgE autoantibodies target keratinocytes in patients with atopic dermatitis. J Invest Dermatol 2008; 128: 2232–2239.
    View article    Google Scholar
  85. Al-Shobaili HA, Ahmed AA, Alnomair N, Alobead ZA, Rasheed Z. Molecular genetic of atopic dermatitis: an update. Int J Health Sci 2016; 10: 96–120.
    View article    Google Scholar
  86. Thijs JL, Strickland I, Bruijnzeel-Koomen C, Nierkens S, Giovannone B, Csomor E, et al. Moving toward endotypes in atopic dermatitis: identification of patient clusters based on serum biomarker analysis. J Allergy Clin Immunol 2017; 140: 730–737.
    View article    Google Scholar
  87. Thijs JL, Strickland I, Bruijnzeel-Koomen C, Nierkens S, Giovannone B, Knol EF, et al. Serum biomarker profiles suggest that atopic dermatitis is a systemic disease. J Allergy Clin Immunol 2018; 141: 1523–1526.
    View article    Google Scholar