Background: Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by recurrent eczematous lesions and severe pruritus. The economic burden and time penalty caused by the relapse of AD reduce patients’ life quality. Summary: AD has complex pathogenesis, including genetic disorders, epidermal barrier dysfunction, abnormal immune responses, microbial dysbiosis of the skin, and environmental factors. Recently, the role of innate immune cells in AD has attracted considerable attention. This review highlighted recent findings on innate immune cells in the onset and progression of AD. Key Messages: Innate immune cells play essential roles in the pathogenesis of AD and enough attention should be given for treating AD from the perspective of innate immunity in clinics.

AD is a chronic inflammatory skin disease closely associated with host immune system dysfunction, manifested by compromised skin barrier. Innate immune cells present variations in quantity and function, contributing to the pathogenesis of AD via various pathways. Additionally, innate immune cells can interact with adaptive immune cells, further complicating the regulatory network. Moreover, some innate immune cells contribute to induce itching. This review describes the functions of innate immune cells in the onset and progression of AD.

Atopic dermatitis (AD) is one of the most common chronic and recurrent inflammatory skin conditions, characterized by recurrent eczematous lesions and intense itching [1]. Early-onset AD usually occurs in childhood, typically at 3–6 months, whereas AD manifests as persistent and new onset forms in adults [2, 3]. The pathogenesis of AD is complex and multifactorial, including immune dysregulation, genetic disorders, epidermal barrier dysfunction, microbial dysbiosis of skin, and environmental factors [4, 5].

Immune dysregulation plays an essential role in the disorder of AD. One hallmark of AD is the accumulation of inflammatory cells within the lesion, including T cells, macrophages, eosinophils, and mast cells. On the contrary, non-lesional atopic skin has a sparse perivascular T-cell infiltrate in the dermis [6]. T helper 1 (Th1)- and T helper 2 (Th2)-cell imbalances and IgE-mediated hypersensitivity have been reported as essential factors in AD pathology [7]. Notably, several studies have emphasized the role of innate immune cells in the onset and progression of AD [8, 9]. It has been widely accepted that type 2 innate lymphoid cells (ILCs), a member of the innate immune system, contribute to the initiation of AD. Unlike the paradigmatic Th2‐mediated immune response in the early stage, a mixed Th1/Th2 pattern of inflammation can be observed in the chronic phase, which is characterized by a switch toward a Th1/Th17 environment [10]. This article reviews recent research on the role of innate immune cells in AD, which will help further explain the pathogenesis of AD and give insight for treating AD from the perspective of innate immunity in clinics.

As the largest immune organ in the body, the skin protects against numerous external dangers [11, 12]. Skin immunity can also be divided into adaptive immunity and innate immunity. Innate immune response has two main characteristics. One is that it can distinguish structural components of microbial pathogens, while these specific structural components are absent in the normal host cells, and these main structural components are pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). The recognition process is mediated by a variety of proteins present in the host cells such as the PRRs [13, 14]. These PRRs could recognize PAMPs or DAMPs present in microbial pathogens. Then, the relevant cells take their functions. The other characteristic is that the innate immune response does not have the ability to generate immunological memory after the recognition of the pathogen. This is different from adaptive immunity [13]. Besides, innate immune cells could take their function rapidly without the need for cloning or amplification while adaptive immunity does. The cells mainly involved in the innate immunity of the skin are natural killer cells (NK cells), natural killer T cells (NKT cells), dendritic cells (DCs), neutrophils, melanocytes, and keratinocytes. Other innate immune cells, including eosinophils, gamma delta T cells (γδ T cells), ILCs, macrophages, mast cells, and basophils, are also implicated in inflammatory skin diseases such as AD, either in lesional skin or in peripheral blood. Specifically, keratinocytes, macrophages, epidermal and dermal DCs, and mast cells constitute the resident cells (shown in Fig. 1a). Meanwhile, when inflammation happens, mobilized cells such as neutrophils, inflammatory DCs, and eosinophils can be immediately recruited to the skin and produce significant effects (shown in Fig. 1b) [15]. In addition, the epidermis and dermis contain specific immune cells. For example, keratinocytes and epidermal DCs distribute in the epidermis and the latter take residence even before birth [16]. In contrast, mast cells and macrophages are found around the dermal papillary vessel and superficial dermis, respectively, while DCs are found throughout the dermis [17]. Other dermal immune components include basophils and eosinophils [16].

Fig. 1.

Distribution of immune cells in the skin. a In normal skin, resident innate immune cells including keratinocytes, macrophages, epidermal DCs (i.e., LC), dDCs, and mast cells, together with adaptive immune cells like T/B lymphocytes constitute a defense barrier. b When inflammation occurs, inflammatory cells such as neutrophils, inflammatory DCs (IDEC), eosinophils and T/B lymphocytes immediately mobilize to the skin. Both the epidermis and dermis contain specific immune cells. LC, Langerhans cell; dDC, dermal dendritic cell; NK cell, natural killer cell; IDEC, inflammatory dendritic epidermal cell.

Fig. 1.

Distribution of immune cells in the skin. a In normal skin, resident innate immune cells including keratinocytes, macrophages, epidermal DCs (i.e., LC), dDCs, and mast cells, together with adaptive immune cells like T/B lymphocytes constitute a defense barrier. b When inflammation occurs, inflammatory cells such as neutrophils, inflammatory DCs (IDEC), eosinophils and T/B lymphocytes immediately mobilize to the skin. Both the epidermis and dermis contain specific immune cells. LC, Langerhans cell; dDC, dermal dendritic cell; NK cell, natural killer cell; IDEC, inflammatory dendritic epidermal cell.

Close modal

Neutrophils

Neutrophil precursors located in the bone marrow, after differentiation and maturation, enter the circulation as neutrophils. During an immune response in the tissue, neutrophils are recruited to detect pathogens and danger signals and to initiate inflammation. Herein, neutrophils are regarded as key innate immune phagocytes in immune defense [18]. An earlier study showed a significant number of neutrophils in the skin of AD lesions [19]. Furthermore, transcriptomic profiling also revealed increased expression of genes associated with neutrophil chemotaxis, such as CXCL8 and GM-CSF [19]. Neutrophils could secrete S100A8 and S100A9, and the latter binds to Toll-like receptor 4 to participate in the pathogenesis of AD [20‒22]. Staphylococcus aureus infection is a common complication of AD. Increased S. aureus may lead to thickening of the epidermis and expansion of immune response cells, leading to the onset and exacerbation of AD [23]. Interestingly, recruited neutrophils failed to inhibit bacterial growth in the skin; as a result, increased S. aureus colonization and persistence were reported. This observation may be caused by the interaction of neutrophil extracellular traps with keratinocytes [24]. Overall, neutrophils may be involved in the pathological process of AD to some extent, but their contribution to AD is much less than that of psoriasis [25].

Eosinophils

Eosinophils are part of white blood cells that help destroy foreign substances, such as parasites, and promote inflammation in the body [26]. Eosinophils and their secreted mediators have been implicated in various inflammatory diseases, including AD [27]. The deposition of eosinophils and eosinophil granules has long been established [28]. Compared with healthy controls, the peripheral blood eosinophils count of AD patients is significantly increased [25], which is a typical feature of AD and has a value in differentiating AD from similar diseases [29, 30]. The elevated levels of blood eosinophils also correlate with disease severity [29, 31].

Although the exact role of eosinophils in the pathogenesis of AD is still unclear, Mendelian randomization findings indicated that increased eosinophil and basophil counts may be a potential causal risk factor of AD [32]. Conversely, the depletion of eosinophils improved skin barrier function and alleviated symptoms [33]. In addition, the activity of eosinophils can be regulated and controlled by pathological changes. Previous studies have confirmed that Th2-derived IL-5 induced activation and chemotaxis of eosinophils [29]. Recently, the effects of histamine on eosinophils have attracted attention. Eosinophils from AD patients highly expressed the histamine H4 receptor (H4R), and histamine was crucial in activating human eosinophils [34]. Osiecka et al. [35] linked secretory leukocyte protease inhibitor (SLPI) to the migratory function of eosinophils. Eosinophils are one of the major sources of SLPI, which is stored in the crystalline core of eosinophil granules. SLPI may be involved in the recruitment of eosinophils to tissue either through interaction with the cell surface and potentially cell-surface receptors, and/or via modifying intracellular pathways upon uptake by eosinophils [35]. Subsequently, eosinophils may produce leukotriene (LT) C4/D4/E4 (LTC4, LTD4, LTE4) that amplify the inflammatory cascade, produce reactive oxygen species, and release extracellular eosinophil traps, thus promoting inflammation [36].

Innate Lymphoid Cells

ILCs comprise five subsets: NK cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer cells. Group 1 ILCs comprise NK cells and ILC1s; group 2 refers to ILC2s; group 3 includes lymphoid tissue inducer cells and ILC3s [37]. ILCs do not express antigen-specific receptors but respond to infection guided by cytokines and secreted proteins. ILC2s are components of the first line of defense, acting prior to the adaptive immune response [38]. ILC2s and ILC3s are resident cells, while ILC1s constantly traffic and predominantly produce IFN-γ [39].

NK Cells

NK cells are cytotoxic cells of the innate immune systems that limit the spread of infection. They circulate in the blood seeking out infected cells or abnormal cells. NK cells recognize certain molecules on the surface of abnormal cells and release cytotoxic granules to destroy them [40]. Variations in the quantity and quality of NK cells in lesional AD skin and peripheral blood have been reported [41‒44]. Activated NK cells and their chemoattractant (CCL2 and CCL22) were enriched in lesional AD skin [41, 43]. However, total CD56+ NK cells and peripheral IFN-γ production were decreased in AD blood samples [42], possibly due to increased NK-cell apoptosis in the blood or increased extravasation in the inflamed skin [42, 44]. The reduced number of NK cells in the blood can also indicate the severity of AD and is of a certain diagnostic value [43‒45]. Histamine may play a role in NK-cell migration into skin lesions. The histamine level was elevated in the skin lesions, and in vitro studies have shown that stimulation of the H4R expressed by NK cells induces chemotaxis [46]. NK cells can function in regulating immune responses in tissues [47]. In skin lesions of AD, NK cells could regulate ILC2s because the loss of NK cells in mice exacerbated pathogenic ILC2 responses [43]. Furthermore, IFN-γ deficiency shifted the Th1/Th2 polarization toward Th2 responses and increased host susceptibility to viral infection [42, 48].

Several changes occur in NK-cell subsets in AD. For example, the proportion of transforming growth factor (TGF)-β-producing regulatory NK (NKreg) subset decreased in peripheral blood mononuclear cells of AD patients, which significantly suppressed Th2 immunity and ILC2 activation in AD mice [49]. In a mouse AD disease model, adoptive transfer of NKreg alleviates the severity of AD [49]. Blood NK cells consist of CD3-CD56dim and CD3-CD56bright-cell subsets. High-dimensional mass cytometry technology and RNA-seq analysis revealed a selective loss of a subset of mature CD56dim NK cells [43]. Additionally, an imbalance in the expression of NK-cell inhibitory and activating receptors as well as a disbalance between resting and activated NK cells have been reported in lesional AD skins [50]. These facts highlight the importance of studying the imbalance of different NK-cell subsets.

ILC2s and ILC3s

ILC2s were originally found to protect the host against intestinal helminth infection by releasing the typical Th2 cytokine IL-13. Later, their function in the production of type 2 inflammatory cytokines, including IL-4, IL-5, and IL-13, was reported, and scholars began to link them to various inflammatory and allergic diseases [51]. Dermal ILC2s can modulate inflammation as they produce IL-13, which affects mast cell activity in normal situations and, when activated, promotes eosinophil influx and mast cell activation [52], with both pro- and anti-inflammatory potential.

The infiltration of ILC2s in AD lesions increased while ILC2s in the blood were rarely detectable [44, 53]. Moreover, several genes encoding Thymic stromal lymphopoietin (TSLP), IL-4, IL-13, and IL-18R are tested to be the susceptibility loci for AD, and all of them are related to the activated behaviors of ILC2s [37, 54, 55]. Experiments in earlier years have already demonstrated the role of ILC2s in MC903 (vitamin D analog calcipotriol)-induced AD-like dermatitis in a mouse model, and deletion of ILCs attenuated the dermatitis [56]. ILC2s are important sources of type 2 cytokines in AD. A subset of ILC2s that express NKp30 can interact with increased B7-H6 [57], a cognate activating ligand, in AD lesions, inducing rapid production of type 2 cytokines, further leading to skin barrier disruption, entry of outer antigens, and induction of effector Th2 cells [58]. Strikingly, though type 2 cytokines serve as a bridge between ILC2s and adaptive immune cells like Th2 cells, ILC2s promote AD independently of adaptive immunity. In a filaggrin-deficient mouse model that tended to AD-like inflammation, the typical clinical manifestations of AD could still be observed under intentional T/B-cell deficiency conditions [59]. Similarly, in another mouse model lacking T/B lymphocytes, AD-like symptoms did not disappear, accompanied by increased ILC2-cell numbers in the skin. These may prove that innate immune response is more important in triggering skin inflammation [60].

In the past, little attention was paid to the role of ILC3s in AD. Although ILC3s are rarely present in human skin, they are significantly increased in eczema [61, 62]. In an AD mouse model built by Kong et al. [62] increases in ILC3s often preceded increases in ILC2s. Moreover, the presence of inflammatory ILC3s roughly correlated with severity. These facts indicate a possible prerequisite role of type 3 innate immune response in AD. In addition, ILC3s can induce IL-33 production by non-immune skin cells, keratinocytes, and fibroblasts, thus promoting type 2 immune responses [63].

Keratinocytes

Keratinocytes are not considered classical innate immune cells in histology. However, to the extent of infection-related areas, we could classify them into innate immune cells according to the following characteristics: Keratinocytes are the main constituent cells of the epidermis, accounting for more than 80% of the epidermal cells [64]. Besides, keratinocytes form the physical skin barrier and represent the first line of the host defense system by sensing pathogens via PRRs on their cell surfaces. These PRRs include Toll-like receptors (TLRs), nucleotide-binding oligomerization domain-like receptor-like receptors (NLRs) and C-type lectin receptors [65]. These receptors recognize highly conserved structure components of microorganisms, known as PAMPs and DAMPs, initiating anti-microbial responses and producing various chemokines, anti-microbial peptides and Th2-promoting cytokines, including TSLP, IL-25 and IL-33. These keratinocyte-derived cytokines could regulate effector T-cell differentiation in AD. Meanwhile, keratinocytes participate in antigen presentation by expressing major histocompatibility complex class II antigens, phagocytosis, and crude processing of antigens [66]. In summary, we could classify them into innate immune cells due to their functions in AD. Keratinocytes can act as an initiator and subsequently activate other cells in the pathogenesis of AD. In AD patients, type 2 pro-inflammatory chemokines can be released from keratinocytes, contributing to the infiltration of mast cells, ILC2 cells, eosinophils, monocytes, and DCs [67, 68]. These cells produce type 2 cytokines and chemokines involved in aberrant immunity in AD. Furthermore, several conditions common in AD can stimulate the release process, including epidermal barrier defects, signal transmission, and environmental stimuli such as dysbiosis of S. aureus [68].

Dendritic Cells

DCs are professional antigen-presenting cells (APC) that form a link between innate and adaptive immunity [69]. In particular, human skin has a special type of DCs called Langerhans cells (LCs), which reside in the epidermis and act as the first cellular barrier. DCs in the dermis comprise different subsets of dermal DCs (dDCs) [70]. Under the state of AD, a special DC subset called inflammatory dendritic epidermal cells (IDEC) infiltrates the inflammatory lesional skin and participates in the development of AD [70].

DCs are capable of regulating Th2-cell function and maintaining homeostasis. Both epidermal LCs and dDCs can produce CC chemokine ligands CCL17 and CCL22, which play a critical role in Th2-cell recruitment and immune response maintenance [71]. However, when a disorder in the regulation occurs, the overreaction of Th2 cells happens and leads to allergic diseases such as AD [72]. In patients with AD, both LC and IDEC highly express the FcεRI receptor, a high-affinity receptor for IgE. IgE facilitates allergen uptake and binding and offers a possible explanation for abnormal IgE-mediated hypersensitivity [69]. Unlike LCs and dDCs, IDECs present the allergen to Th1 cells and are likely to associate with the chronicity of eczema by IL-12 and IL-18 production [41, 73].

Mast Cells

Skin mast cells are also APCs that involve immune regulation and allergic reactions due to the expression of large numbers of IgE Fc receptors. Mast cells increased in skin lesions in both human and mouse AD models, indicating their participation in AD [74]. Mast cell-deficient mice displayed attenuated skin inflammation, revealing that mast cells are essential in allergen-induced AD-like skin inflammation [75]. In addition, a special receptor, dendritic cell immunoreceptor (DCIR), which mediates allergen binding and uptake, was upregulated, further leading to ROS generation and oxidation of calmodulin kinase II (ox-CaMKII) [75]. Numerous studies also revealed that mast cells are producers of several inflammatory mediators demonstrated to associate with AD, including prostaglandin D2 (PGD2), histamine, IL-31, and IL-33 [76].

γδ T Cells

γδ T cells are a subset of T cells that perform intrinsic immune functions, and their T-cell receptors (TCRs) are composed of γ and δ chains. Though γδ T cells occupy a tiny percentage of total T cells in the peripheral blood, they are enriched in the skin, especially during acute and chronic skin inflammation [77]. However, blood γδ T cells are decreased due to apoptosis induced by monocytes [48]. Meanwhile, γδ T cells may have the activity of maintaining homeostasis. Recent findings demonstrate that innate neonatal-derived IL-17 producing γδ T (Tγδ17)-cell subset has a crucial role in preventing AD. Spontaneous AD was observed in a Tγδ17 cell-deficient mouse model, indicating the dual homeostatic and inflammatory function of IL-17 in AD [78].

Basophils

Basophils, similar to mast cells residing in the skin, also express the high-affinity receptor for IgE, FcεRI, and release similar effector molecules such as histamine, 5-hydroxytryptamine, tryptophan-like enzymes, and LTs upon IgE cross-linking. It is noteworthy that basophils are present in the skin lesions among approximately 60% of patients with AD [79], while in peripheral blood, the number of basophils varied [79, 80], indicating distinct endotypes of AD (with or without involvement of basophils). Recent studies report that FcεRIa is upregulated on basophils in inflammation associated with AD [81]. In addition, for AD involving basophils, a retrospective study shows that basophils are associated with epidermal infiltration of helper T cells [82], and might play a role in pruritus (described below).

NKT Cells

NKT cells express NK receptors and markers of conventional T cells as well [83], and are usually divided into type I and type II. Type I NKT cells are called semi-invariant NKT cells (iNKT) because they express the typical, semi-invariant TCR, whereas type II NKT cells have a different TCR repertoire. Both type I and type II NKT cells can recognize glycolipid antigens on CD1d molecules.

The significant alterations in the number and function of NKT cells in AD suggest a potential link between NKT cells and AD [84‒86]. Decreased intracellular IFN-γ and increased intracellular IL-4 were observed [84]. Besides, surface immune cell markers, including CXCR4 and CD4/CD8, also undergo quantitative changes in NKT cells [86]. Skin-resident NKT cells uniquely expressed CXCR4 and were enriched in AD skin, which was presumed to be recruited by fibroblast-derived CXCL12 [85]. However, although NKT cells play a critical role in many other allergic diseases, they may not compose a requisite part of allergic skin inflammation [87]. The role of NKT cells in AD requires further investigation.

Macrophages

Macrophages derive from monocytes when they invade the dermis. As a member of APCs, dermal macrophages play a role in wound repair and prevention of microbial invasion. Previous studies have revealed increased infiltration of macrophages in AD skin [88]. Furthermore, reduced phagocytic activity and TLR-2 expression of macrophages were detected in patients with AD, contributing to enhanced susceptibility to skin infections [88]. Recently, increasing evidence has shown how macrophages are involved in the pathogenesis of AD. Transient receptor potential vanilloid 4 (TRPV4) expressed by macrophages exhibited an anti-inflammatory effect, and decreased expression of TRPV4 was detected in AD [89]. Chitinase 3-like 1 (CHI3L1), a recognized player in Th2 inflammation, has been proven to drive AD via M2 macrophage activation [90]. Additionally, histamine stimulated M2 macrophages to express chemokine CCL18, a highly expressed chemokine in lesional skin and serum [91]. In conclusion, the infiltration and activation of macrophages may contribute to the maintenance of inflammation [92].

Innate Immune Cells Regulate Innate Immunity

ILC2s play a pivotal role in the immune network concerning innate immunity. ILC2s can be activated by several innate immune cells. In MC903-induced AD mouse models, basophils regulated ILC2 activation via IL-4 production [93]. Mast cells enhanced IL-5 production by ILC2s via miR103a-3p in extracellular vesicles [94]. Additionally, mast cell-derived mediators such as cysteinyl LTs, LTE4 in particular, can activate ILC2s. Meanwhile, the expression of LT receptors CysLT1 increased on ILC2s [95]. Basophil-generated IL-4 and increased expression of CCL11, IL-5, IL-9, and IL-13 by ILC2s after activation by basophils contributed to the recruitment and accumulation of eosinophils [96, 97].

Keratinocytes form the first line of defense and play a role in the upstream of the network. TSLP expression in keratinocytes significantly increases, directly activating iNKT cells to secrete IL-4 and IL-13 [98]. Besides, TSLP is known as a main regulator of skin ILC2s [98]. Innate immune signaling in keratinocytes, like MyD88 signaling, led to T-cell recruitment and LC emigration [99].

Innate Immune Cells Regulate Adaptive Immunity

Since the acute phase of AD is dominated by type 2 immune responses, the effect of Th2 cells cannot be ignored. As previously mentioned, TSLP directly interacted with Th2 cells; NK cells promoted Th2 polarization; and DCs participated in Th2-cell recruitment [42, 48, 71]. In the chronic stage, Type 1 inflammation also plays a part. Infiltration of Th1 cells required IL-12 secreted by macrophages, eosinophils, and DCs [100]. In addition, innate immune cells such as ILCs produced Th1 cytokines (IL-2, IFN-γ, etc.) and Th2 cytokines (IL-5, IL-13, etc.) to promote adaptive immunity [101]. Overall, innate immune cells actively participate in both innate and adaptive immunity, constituting a complex network in AD (shown in Fig. 2).

Fig. 2.

Role of innate immune cells in innate immunity and adaptive immunity. a ILC2s can be activated by basophils, mast cells, NK cells and keratinocytes, and then produce inflammatory cytokines to activate downstream cells, thus playing a central role. Keratinocytes, however, are more often involved in the upstream of the regulatory network. b Innate immune cells have an effect on both Th2 and Th1/17 lymphocytes. ILC, innate lymphoid cell; KC, keratinocytes; TSLP, thymic stromal lymphopoietin; iNKT, semi-invariant NKT cell; LTE4, leukotriene E4; SLPI, secretory leukocyte protease inhibitor. Solid or dashed arrows representatively indicate direct or indirect effects.

Fig. 2.

Role of innate immune cells in innate immunity and adaptive immunity. a ILC2s can be activated by basophils, mast cells, NK cells and keratinocytes, and then produce inflammatory cytokines to activate downstream cells, thus playing a central role. Keratinocytes, however, are more often involved in the upstream of the regulatory network. b Innate immune cells have an effect on both Th2 and Th1/17 lymphocytes. ILC, innate lymphoid cell; KC, keratinocytes; TSLP, thymic stromal lymphopoietin; iNKT, semi-invariant NKT cell; LTE4, leukotriene E4; SLPI, secretory leukocyte protease inhibitor. Solid or dashed arrows representatively indicate direct or indirect effects.

Close modal

Pruritus, transmitted by non-myelinated C-type free nerve endings in the epidermis and upper dermis [102], is one of the major and diagnostic symptoms of AD. During the induction of pruritus, the interactions between sensory neurons and immune cells require pruritogens to act as intermediaries [103]. It is well known that the primary sources of pruritogens are histamine, neuropeptides, and cytokines (including IL-4, IL-13, and IL-31), and innate cells are the important producers. For example, mast cells, basophils, and eosinophils are the primary sources of pruritogens mentioned above [104]. A basophil-LT axis contributes to the acute itch flare in the chronic course of AD because allergen-stimulated basophils produce enhanced LTC4, which then bind to its receptor CysLTR2 on neurons [81]. Besides, eosinophils in the skin of AD patients mainly locate close to peripheral nerves and express BDNF, one of the neurite growth-stimulating factors, and may induce itch via stimulation of skin nerve branching [102].

Neutrophils play many roles in itch as neutrophils produce and release pruritogens like histamine and LTB4, which then induce itch [105]. Crisaborole (phosphodiesterase 4 inhibitor), an ointment approved to treat mild to moderate AD, inhibits itch by reducing neutrophil infiltration and interaction of neutrophils with sensory neurons in the MC903-treated mouse model [106]. Depletion of neutrophils significantly relieved itch in the mouse model of AD, along with a decrease in several hallmarks of chronic itch inflammatory cytokines [107]. However, above-mentioned evidence of neutrophils inducing itch is mainly based on mouse models. Although the role of neutrophils in itch has been demonstrated in other pruritic skin diseases like psoriasis, the role in AD needs further research.

Keratinocytes are the major source of TSLP, which activates primary afferent neurons through TRPA1 activation [108]. TSLP expression was highly upregulated in AD skin, accounting for pruritus induction [109].

M2 macrophages are essential producers of IL-31, a type 2 cytokine related to itching sensation [110]. The scratching due to the sensation of pruritus will eventually damage the skin barrier and increase the likelihood of skin infections (shown in Fig. 3).

Fig. 3.

Role of innate immune cells in pruritus. Innate immune cells induce itch directly or indirectly via secretion of small molecules. The subsequent act of scratching could lead to skin barrier damage and infections, constituting a vicious cycle. Solid or dashed arrows representatively indicate direct or indirect effects.

Fig. 3.

Role of innate immune cells in pruritus. Innate immune cells induce itch directly or indirectly via secretion of small molecules. The subsequent act of scratching could lead to skin barrier damage and infections, constituting a vicious cycle. Solid or dashed arrows representatively indicate direct or indirect effects.

Close modal

AD can be divided into two types: intrinsic type of AD (IAD) and extrinsic type of AD (EAD) [111]. The difference between these two types includes the concentrations of normal total serum IgE levels and allergen-specific IgE antibodies in the blood. IAD may develop into EAD over time. With the elevation of total serum IgE [112], EAD patients showed high levels of Th2 cytokines (IL-4, IL-5, and IL-13) compared with IAD. Besides, strong CCL18 expression was observed in lesional skin and IDEC-like DCs in EAD patients [113]. These phenomena indicate that there may be some differences in the function of cells between two types of AD, including mast cells, DCs, monocytes, etc.

AD is also considered a biphasic disease, with Th2 predominating in acute disease and a switch to Th1 characterizing chronic disease. In the acute phase of AD, a subset of increased terminal differentiation proteins was found, including S100A7, S100A8, and S100A9 [114]. The last two proteins could be secreted by neutrophils and bind to Toll-like receptor 4 to participate in the pathogenesis of AD [20‒22]. These might indicate that the role of neutrophils in the acute phase of AD is more dominant than that in the chronic phase. Since the acute phase of AD is dominated by type 2 immune responses, the effect of Th2 cells cannot be ignored. TSLP could directly interact with Th2 cells; NK cells promoted Th2 polarization; and DCs participated in Th2-cell recruitment [42, 48, 71]. In the chronic stage, type 1 inflammation also plays a part. Infiltration of Th1 cells required IL-12 secreted by macrophages, eosinophils, and DCs [100]. In addition, innate immune cells such as ILCs produced Th1 cytokines (IL-2, IFN-γ, etc.) and Th2 cytokines (IL-5, IL-13, etc.) to promote adaptive immunity [101].

AD could also be divided into adult-onset AD and child-onset AD. About 13% of American children suffer from AD. While 1 in 4 adults with AD report adult-onset disease, many adults with AD might have child-onset AD [115]. These differences may be due to different stages of growth and immune status.

AD is a chronic inflammatory skin disease closely associated with host immune system dysfunction, manifested by compromised skin barrier. Innate immune cells present variations in quantity and function, contributing to the pathogenesis of AD via various pathways. Additionally, innate immune cells can interact with adaptive immune cells, further complicating the regulatory network. Moreover, some innate immune cells contribute to induce itching. Overall, this review helps understand the roles of innate immune cells in AD and is instructive for treatment.

The authors have no conflicts of interest to declare.

This study was not supported by any sponsor or funder.

Yuke Pan, Youyi Wang, Kang Zeng, and Xiaowen Huang reviewed previous related publications. Yuke Pan and Youyi Wang wrote the first draft of the manuscript. Meinian Xu, Meizhen Zhong, Xiaoming Peng, Kang Zeng, and Xiaowen Huang commented on previous versions of the manuscript. Yuke Pan, Youyi Wang, Meinian Xu, Meizhen Zhong, Xiaoming Peng, Kang Zeng, and Xiaowen Huang read and approved the final manuscript.

Additional Information

Yuke Pan and Youyi Wang contributed equally to this work and share first authorship.

1.
Langan
SM
,
Irvine
AD
,
Weidinger
S
.
Atopic dermatitis
.
Lancet
.
2020
;
396
(
10247
):
345
60
.
2.
Abuabara
K
,
Hoffstad
O
,
Troxel
AB
,
Gelfand
JM
,
McCulloch
CE
,
Margolis
DJ
.
Patterns and predictors of atopic dermatitis disease control past childhood: an observational cohort study
.
J Allergy Clin Immunol
.
2018
;
141
(
2
):
778
80.e6
.
3.
Abuabara
K
,
Yu
AM
,
Okhovat
JP
,
Allen
IE
,
Langan
SM
.
The prevalence of atopic dermatitis beyond childhood: a systematic review and meta-analysis of longitudinal studies
.
Allergy
.
2018
;
73
(
3
):
696
704
.
4.
David Boothe
W
,
Tarbox
JA
,
Tarbox
MB
.
Atopic dermatitis: pathophysiology
.
Adv Exp Med Biol
.
2024
;
1447
:
21
35
.
5.
Li
H
,
Zhang
Z
,
Zhang
H
,
Guo
Y
,
Yao
Z
.
Update on the pathogenesis and therapy of atopic dermatitis
.
Clin Rev Allergy Immunol
.
2021
;
61
(
3
):
324
38
.
6.
Bieber
T
.
Atopic dermatitis
.
N Engl J Med
.
2008
;
358
(
14
):
1483
94
.
7.
Sheikhi
A
,
Giti
H
,
Heibor
MR
,
Jafarzadeh
A
,
Shakerian
M
,
Baharifar
N
, et al
.
Lactobacilus delbrueckii subsp. bulgaricus modulates the secretion of Th1/Th2 and treg cell-related cytokines by PBMCs from patients with atopic dermatitis
.
Drug Res (Stuttg)
.
2017
;
67
(
12
):
724
9
.
8.
Yasuda-Sekiguchi
F
,
Shiohama
A
,
Fukushima
A
,
Obata
S
,
Mochimaru
N
,
Honda
A
, et al
.
Single nucleotide variations in genes associated with innate immunity are enriched in Japanese adult cases of face and neck type atopic dermatitis
.
J Dermatol Sci
.
2021
;
101
(
2
):
93
100
.
9.
Hawerkamp
HC
,
Fahy
CMR
,
Fallon
PG
,
Schwartz
C
.
Break on through: the role of innate immunity and barrier defence in atopic dermatitis and psoriasis
.
Skin Health Dis
.
2022
;
2
(
2
):
e99
.
10.
Orciani
M
,
Campanati
A
,
Caffarini
M
,
Ganzetti
G
,
Consales
V
,
Lucarini
G
, et al
.
T helper (Th)1, Th17 and Th2 imbalance in mesenchymal stem cells of adult patients with atopic dermatitis: at the origin of the problem
.
Br J Dermatol
.
2017
;
176
(
6
):
1569
76
.
11.
Bos
JD
,
Kapsenberg
ML
.
The skin immune system its cellular constituents and their interactions
.
Immunol Today
.
1986
;
7
(
7–8
):
235
40
.
12.
Salmon
JK
,
Armstrong
CA
,
Ansel
JC
.
The skin as an immune organ
.
West J Med
.
1994
;
160
(
2
):
146
52
.
13.
Takeuchi
O
,
Akira
S
.
Pattern recognition receptors and inflammation
.
Cell
.
2010
;
140
(
6
):
805
20
.
14.
Wilkins
C
,
Gale
M
Jr
.
Recognition of viruses by cytoplasmic sensors
.
Curr Opin Immunol
.
2010
;
22
(
1
):
41
7
.
15.
Skabytska
Y
,
Kaesler
S
,
Volz
T
,
Biedermann
T
.
The role of innate immune signaling in the pathogenesis of atopic dermatitis and consequences for treatments
.
Semin Immunopathol
.
2016
;
38
(
1
):
29
43
.
16.
Kobayashi
T
,
Naik
S
,
Nagao
K
.
Choreographing immunity in the skin epithelial barrier
.
Immunity
.
2019
;
50
(
3
):
552
65
.
17.
Holken
JM
,
Friedrich
K
,
Merkel
M
,
Blasius
N
,
Engels
U
,
Buhl
T
, et al
.
A human 3D immune competent full-thickness skin model mimicking dermal dendritic cell activation
.
Front Immunol
.
2023
;
14
:
1276151
.
18.
Galli
SJ
,
Borregaard
N
,
Wynn
TA
.
Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils
.
Nat Immunol
.
2011
;
12
(
11
):
1035
44
.
19.
Choy
DF
,
Hsu
DK
,
Seshasayee
D
,
Fung
MA
,
Modrusan
Z
,
Martin
F
, et al
.
Comparative transcriptomic analyses of atopic dermatitis and psoriasis reveal shared neutrophilic inflammation
.
J Allergy Clin Immunol
.
2012
;
130
(
6
):
1335
43 e5
.
20.
Sabroe
I
,
Prince
LR
,
Jones
EC
,
Horsburgh
MJ
,
Foster
SJ
,
Vogel
SN
, et al
.
Selective roles for Toll-Like Receptor (TLR)2 and TLR4 in the regulation of neutrophil activation and life span
.
J Immunol
.
2003
;
170
(
10
):
5268
75
.
21.
Vogl
T
,
Tenbrock
K
,
Ludwig
S
,
Leukert
N
,
Ehrhardt
C
,
van Zoelen
MA
, et al
.
Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock
.
Nat Med
.
2007
;
13
(
9
):
1042
9
.
22.
Wang
Y
,
Weng
H
,
Song
JF
,
Deng
YH
,
Li
S
,
Liu
HB
.
Activation of the HMGB1-TLR4-NF-κB pathway may occur in patients with atopic eczema
.
Mol Med Rep
.
2017
;
16
(
3
):
2714
20
.
23.
Byrd
AL
,
Deming
C
,
Cassidy
SKB
,
Harrison
OJ
,
Ng
WI
,
Conlan
S
, et al
.
Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis
.
Sci Transl Med
.
2017
;
9
(
397
):
eaal4651
.
24.
Bitschar
K
,
Staudenmaier
L
,
Klink
L
,
Focken
J
,
Sauer
B
,
Fehrenbacher
B
, et al
.
Staphylococcus aureus skin colonization is enhanced by the interaction of neutrophil extracellular traps with keratinocytes
.
J Invest Dermatol
.
2020
;
140
(
5
):
1054
65.e4
.
25.
Dhingra
N
,
Suarez-Farinas
M
,
Fuentes-Duculan
J
,
Gittler
JK
,
Shemer
A
,
Raz
A
, et al
.
Attenuated neutrophil axis in atopic dermatitis compared to psoriasis reflects TH17 pathway differences between these diseases
.
J Allergy Clin Immunol
.
2013
;
132
(
2
):
498
501 e3
.
26.
Shin
SH
,
Ye
MK
,
Park
J
,
Geum
SY
.
Immunopathologic role of eosinophils in eosinophilic chronic rhinosinusitis
.
Int J Mol Sci
.
2022
;
23
(
21
):
13313
.
27.
Simon
HU
,
Yousefi
S
,
Germic
N
,
Arnold
IC
,
Haczku
A
,
Karaulov
AV
, et al
.
The cellular functions of eosinophils: Collegium Internationale Allergologicum (CIA) update 2020
.
Int Arch Allergy Immunol
.
2020
;
181
(
1
):
11
23
.
28.
Simon
D
,
Braathen
LR
,
Simon
HU
.
Eosinophils and atopic dermatitis
.
Allergy
.
2004
;
59
(
6
):
561
70
.
29.
Jenerowicz
D
,
Czarnecka-Operacz
M
,
Silny
W
.
Peripheral blood eosinophilia in atopic dermatitis
.
Acta Dermatovenerol Alp Pannonica Adriat
.
2007
;
16
(
2
):
47
52
.
30.
Altaii
HA
,
Al-Tae
FMD
.
Investigation of serum total IgE and eosinophil levels in different allergic diseases together with the study of their correlations with various possible allergens in mosul city
.
Immunol Invest
.
2022
;
51
(
3
):
567
87
.
31.
Radonjic-Hoesli
S
,
Bruggen
MC
,
Feldmeyer
L
,
Simon
HU
,
Simon
D
.
Eosinophils in skin diseases
.
Semin Immunopathol
.
2021
;
43
(
3
):
393
409
.
32.
Zeng-Yun-Ou
Z
,
Zhong-Yu
J
,
Wei
L
.
Bidirectional associations between eosinophils, basophils, and lymphocytes with atopic dermatitis: a multivariable Mendelian randomization study
.
Front Immunol
.
2022
;
13
:
1001911
.
33.
Naidoo
K
,
Jagot
F
,
van den Elsen
L
,
Pellefigues
C
,
Jones
A
,
Luo
H
, et al
.
Eosinophils determine dermal thickening and water loss in an MC903 model of atopic dermatitis
.
J Invest Dermatol
.
2018
;
138
(
12
):
2606
16
.
34.
Beyer
L
,
Kabatas
AS
,
Mommert
S
,
Stark
H
,
Werfel
T
,
Gutzmer
R
, et al
.
Histamine activates human eosinophils via H(2)R and H(4)R predominantly in atopic dermatitis patients
.
Int J Mol Sci
.
2022
;
23
(
18
):
10294
.
35.
Osiecka
O
,
Skrzeczynska-Moncznik
J
,
Morytko
A
,
Mazur
A
,
Majewski
P
,
Bilska
B
, et al
.
Secretory leukocyte protease inhibitor is present in circulating and tissue-recruited human eosinophils and regulates their migratory function
.
Front Immunol
.
2021
;
12
:
737231
.
36.
Marzano
AV
,
Genovese
G
.
Eosinophilic dermatoses: recognition and management
.
Am J Clin Dermatol
.
2020
;
21
(
4
):
525
39
.
37.
Vivier
E
,
Artis
D
,
Colonna
M
,
Diefenbach
A
,
Di Santo
JP
,
Eberl
G
, et al
.
Innate lymphoid cells: 10 Years on
.
Cell
.
2018
;
174
(
5
):
1054
66
.
38.
Gomez-Cadena
A
,
Romero
P
,
Trabanelli
S
,
Jandus
C
.
Detecting and analyzing murine innate lymphoid cells
.
Methods Enzymol
.
2020
;
631
:
329
42
.
39.
Dutton
EE
,
Gajdasik
DW
,
Willis
C
,
Fiancette
R
,
Bishop
EL
,
Camelo
A
, et al
.
Peripheral lymph nodes contain migratory and resident innate lymphoid cell populations
.
Sci Immunol
.
2019
;
4
(
35
):
eaau8082
.
40.
Gutic
B
,
Bozanovic
T
,
Mandic
A
,
Dugalic
S
,
Todorovic
J
,
Stanisavljevic
D
, et al
.
Programmed cell death-1 and its ligands: current knowledge and possibilities in immunotherapy
.
Clinics
.
2023
;
78
:
100177
.
41.
von Bubnoff
D
,
Andres
E
,
Hentges
F
,
Bieber
T
,
Michel
T
,
Zimmer
J
.
Natural killer cells in atopic and autoimmune diseases of the skin
.
J Allergy Clin Immunol
.
2010
;
125
(
1
):
60
8
.
42.
Luci
C
,
Gaudy-Marqueste
C
,
Rouzaire
P
,
Audonnet
S
,
Cognet
C
,
Hennino
A
, et al
.
Peripheral natural killer cells exhibit qualitative and quantitative changes in patients with psoriasis and atopic dermatitis
.
Br J Dermatol
.
2012
;
166
(
4
):
789
96
.
43.
Mack
MR
,
Brestoff
JR
,
Berrien-Elliott
MM
,
Trier
AM
,
Yang
TB
,
McCullen
M
, et al
.
Blood natural killer cell deficiency reveals an immunotherapy strategy for atopic dermatitis
.
Sci Transl Med
.
2020
;
12
(
532
):
eaay1005
.
44.
Worm
M
,
Glatzel
V
,
Baumgart
S
,
Mei
HE
,
Sorensen
T
,
Grutzkau
A
, et al
.
Immune cell profiling reveals natural killer and T cell subpopulations to be associated with atopic dermatitis severity
.
Clin Exp Allergy
.
2023
;
53
(
1
):
105
8
.
45.
Kabashima
K
,
Weidinger
S
.
NK cells as a possible new player in atopic dermatitis
.
J Allergy Clin Immunol
.
2020
;
146
(
2
):
276
7
.
46.
Ehling
S
,
Rossbach
K
,
Dunston
SM
,
Stark
H
,
Baumer
W
.
Allergic inflammation is augmented via histamine H4 receptor activation: the role of natural killer cells in vitro and in vivo
.
J Dermatol Sci
.
2016
;
83
(
2
):
106
15
.
47.
Vivier
E
,
Tomasello
E
,
Baratin
M
,
Walzer
T
,
Ugolini
S
.
Functions of natural killer cells
.
Nat Immunol
.
2008
;
9
(
5
):
503
10
.
48.
Katsuta
M
,
Takigawa
Y
,
Kimishima
M
,
Inaoka
M
,
Takahashi
R
,
Shiohara
T
.
NK cells and gamma delta+ T cells are phenotypically and functionally defective due to preferential apoptosis in patients with atopic dermatitis
.
J Immunol
.
2006
;
176
(
12
):
7736
44
.
49.
Min
KY
,
Koo
J
,
Noh
G
,
Lee
D
,
Jo
MG
,
Lee
JE
, et al
.
CD1d(hi)PD-L1(hi)CD27(+) regulatory natural killer subset suppresses atopic dermatitis
.
Front Immunol
.
2021
;
12
:
752888
.
50.
Mobus
L
,
Rodriguez
E
,
Harder
I
,
Schwarz
A
,
Wehkamp
U
,
Stolzl
D
, et al
.
Elevated NK-cell transcriptional signature and dysbalance of resting and activated NK cells in atopic dermatitis
.
J Allergy Clin Immunol
.
2021
;
147
(
5
):
1959
65.e2
.
51.
Spits
H
,
Mjosberg
J
.
Heterogeneity of type 2 innate lymphoid cells
.
Nat Rev Immunol
.
2022
;
22
(
11
):
701
12
.
52.
Roediger
B
,
Kyle
R
,
Yip
KH
,
Sumaria
N
,
Guy
TV
,
Kim
BS
, et al
.
Cutaneous immunosurveillance and regulation of inflammation by group 2 innate lymphoid cells
.
Nat Immunol
.
2013
;
14
(
6
):
564
73
.
53.
Brunner
PM
,
Guttman-Yassky
E
,
Leung
DY
.
The immunology of atopic dermatitis and its reversibility with broad-spectrum and targeted therapies
.
J Allergy Clin Immunol
.
2017
;
139
(
4S
):
S65
76
.
54.
Tamari
M
,
Hirota
T
.
Genome-wide association studies of atopic dermatitis
.
J Dermatol
.
2014
;
41
(
3
):
213
20
.
55.
Paternoster
L
,
Standl
M
,
Waage
J
,
Baurecht
H
,
Hotze
M
,
Strachan
DP
, et al
.
Multi-ancestry genome-wide association study of 21,000 cases and 95,000 controls identifies new risk loci for atopic dermatitis
.
Nat Genet
.
2015
;
47
(
12
):
1449
56
.
56.
Kim
BS
,
Siracusa
MC
,
Saenz
SA
,
Noti
M
,
Monticelli
LA
,
Sonnenberg
GF
, et al
.
TSLP elicits IL-33-independent innate lymphoid cell responses to promote skin inflammation
.
Sci Transl Med
.
2013
;
5
(
170
):
170ra16
.
57.
Salimi
M
,
Xue
L
,
Jolin
H
,
Hardman
C
,
Cousins
DJ
,
McKenzie
AN
, et al
.
Group 2 innate lymphoid cells express functional NKp30 receptor inducing type 2 cytokine production
.
J Immunol
.
2016
;
196
(
1
):
45
54
.
58.
Honda
T
,
Kabashima
K
.
Reconciling innate and acquired immunity in atopic dermatitis
.
J Allergy Clin Immunol
.
2020
;
145
(
4
):
1136
7
.
59.
Saunders
SP
,
Moran
T
,
Floudas
A
,
Wurlod
F
,
Kaszlikowska
A
,
Salimi
M
, et al
.
Spontaneous atopic dermatitis is mediated by innate immunity, with the secondary lung inflammation of the atopic march requiring adaptive immunity
.
J Allergy Clin Immunol
.
2016
;
137
(
2
):
482
91
.
60.
Chen
LY
,
Yang-Yen
HF
,
Tsai
CC
,
Thio
CL
,
Chuang
HL
,
Yang
LT
, et al
.
Protein palmitoylation by ZDHHC13 protects skin against microbial-driven dermatitis
.
J Invest Dermatol
.
2017
;
137
(
4
):
894
904
.
61.
Bruggen
MC
,
Bauer
WM
,
Reininger
B
,
Clim
E
,
Captarencu
C
,
Steiner
GE
, et al
.
In situ mapping of innate lymphoid cells in human skin: evidence for remarkable differences between normal and inflamed skin
.
J Invest Dermatol
.
2016
;
136
(
12
):
2396
405
.
62.
Kong
WS
,
Tsuyama
N
,
Inoue
H
,
Guo
Y
,
Mokuda
S
,
Nobukiyo
A
, et al
.
Long-chain saturated fatty acids in breast milk are associated with the pathogenesis of atopic dermatitis via induction of inflammatory ILC3s
.
Sci Rep
.
2021
;
11
(
1
):
13109
.
63.
Kim
MH
,
Jin
SP
,
Jang
S
,
Choi
JY
,
Chung
DH
,
Lee
DH
, et al
.
IL-17A-Producing innate lymphoid cells promote skin inflammation by inducing IL-33-driven type 2 immune responses
.
J Invest Dermatol
.
2020
;
140
(
4
):
827
37.e9
.
64.
Russo
B
,
Brembilla
NC
,
Chizzolini
C
.
Contribution of keratinocytes to dermal fibrosis
.
Curr Opin Rheumatol
.
2022
;
34
(
6
):
337
42
.
65.
Coates
M
,
Blanchard
S
,
MacLeod
AS
.
Innate antimicrobial immunity in the skin: a protective barrier against bacteria, viruses, and fungi
.
PLoS Pathog
.
2018
;
14
(
12
):
e1007353
.
66.
Schell
SL
,
Cong
Z
,
Sennett
ML
,
Gettle
SL
,
Longenecker
AL
,
Goldberg
SR
, et al
.
Keratinocytes and immune cells in the epidermis are key drivers of inflammation in hidradenitis suppurativa providing a rationale for novel topical therapies
.
Br J Dermatol
.
2023
;
188
(
3
):
407
19
.
67.
Patel
NA
,
Patel
JA
,
Stins
MF
,
Kim
KS
,
Chang
SL
.
Dexamethasone affects cytokine-mediated adhesion of HL-60 human promyelocytic leukemia cells to cultured dermal microvascular endothelial cells
.
Clin Immunol
.
2001
;
99
(
3
):
387
94
.
68.
Chieosilapatham
P
,
Kiatsurayanon
C
,
Umehara
Y
,
Trujillo-Paez
JV
,
Peng
G
,
Yue
H
, et al
.
Keratinocytes: innate immune cells in atopic dermatitis
.
Clin Exp Immunol
.
2021
;
204
(
3
):
296
309
.
69.
Kumar
S
,
Jeong
Y
,
Ashraf
MU
,
Bae
YS
.
Dendritic cell-mediated Th2 immunity and immune disorders
.
Int J Mol Sci
.
2019
;
20
(
9
):
2159
.
70.
Scheib
N
,
Tiemann
J
,
Becker
C
,
Probst
HC
,
Raker
VK
,
Steinbrink
K
.
The dendritic cell dilemma in the skin: between tolerance and immunity
.
Front Immunol
.
2022
;
13
:
929000
.
71.
Miake
S
,
Tsuji
G
,
Takemura
M
,
Hashimoto-Hachiya
A
,
Vu
YH
,
Furue
M
, et al
.
IL-4 augments IL-31/IL-31 receptor alpha interaction leading to enhanced ccl 17 and ccl 22 production in dendritic cells: implications for atopic dermatitis
.
Int J Mol Sci
.
2019
;
20
(
16
):
4053
.
72.
Liu
P
,
Kang
C
,
Zhang
J
,
Liu
Y
,
Liu
J
,
Hu
T
, et al
.
The role of dendritic cells in allergic diseases
.
Int Immunopharmacol
.
2022
;
113
(
Pt B
):
109449
.
73.
Yoshida
K
,
Kubo
A
,
Fujita
H
,
Yokouchi
M
,
Ishii
K
,
Kawasaki
H
, et al
.
Distinct behavior of human Langerhans cells and inflammatory dendritic epidermal cells at tight junctions in patients with atopic dermatitis
.
J Allergy Clin Immunol
.
2014
;
134
(
4
):
856
64
.
74.
Otsuka
A
,
Kabashima
K
.
Mast cells and basophils in cutaneous immune responses
.
Allergy
.
2015
;
70
(
2
):
131
40
.
75.
Luo
X
,
Chen
J
,
Yang
H
,
Hu
X
,
Alphonse
MP
,
Shen
Y
, et al
.
Dendritic cell immunoreceptor drives atopic dermatitis by modulating oxidized CaMKII-involved mast cell activation
.
JCI Insight
.
2022
;
7
(
5
):
e152559
.
76.
Modena
BD
,
Dazy
K
,
White
AA
.
Emerging concepts: mast cell involvement in allergic diseases
.
Transl Res
.
2016
;
174
:
98
121
.
77.
Castillo-Gonzalez
R
,
Cibrian
D
,
Sanchez-Madrid
F
.
Dissecting the complexity of γδ T-cell subsets in skin homeostasis, inflammation, and malignancy
.
J Allergy Clin Immunol
.
2021
;
147
(
6
):
2030
42
.
78.
Spidale
NA
,
Malhotra
N
,
Frascoli
M
,
Sylvia
K
,
Miu
B
,
Freeman
C
, et al
.
Neonatal-derived IL-17 producing dermal γδ T cells are required to prevent spontaneous atopic dermatitis
.
Elife
.
2020
;
9
:
9
.
79.
Ito
Y
,
Satoh
T
,
Takayama
K
,
Miyagishi
C
,
Walls
AF
,
Yokozeki
H
.
Basophil recruitment and activation in inflammatory skin diseases
.
Allergy
.
2011
;
66
(
8
):
1107
13
.
80.
Mashiko
S
,
Mehta
H
,
Bissonnette
R
,
Sarfati
M
.
Increased frequencies of basophils, type 2 innate lymphoid cells and Th2 cells in skin of patients with atopic dermatitis but not psoriasis
.
J Dermatol Sci
.
2017
;
88
(
2
):
167
74
.
81.
Wang
F
,
Trier
AM
,
Li
F
,
Kim
S
,
Chen
Z
,
Chai
JN
, et al
.
A basophil-neuronal axis promotes itch
.
Cell
.
2021
;
184
(
2
):
422
40.e17
.
82.
Nagashima
N
,
Ugajin
T
,
Miyake
K
,
Walls
AF
,
Namiki
T
,
Yokozeki
H
, et al
.
Cutaneous basophil infiltration in atopic dermatitis is associated with abundant epidermal infiltration of helper T cells: a preliminary retrospective study
.
J Dermatol
.
2024
;
51
(
1
):
130
4
.
83.
Godfrey
DI
,
Hammond
KJ
,
Poulton
LD
,
Smyth
MJ
,
Baxter
AG
.
NKT cells: facts, functions and fallacies
.
Immunol Today
.
2000
;
21
(
11
):
573
83
.
84.
Gyimesi
E
,
Nagy
G
,
Remenyik
E
,
Sipka
S
,
Zeher
M
,
Biro
T
, et al
.
Altered peripheral invariant natural killer T cells in atopic dermatitis
.
J Clin Immunol
.
2011
;
31
(
5
):
864
72
.
85.
Sun
Z
,
Kim
JH
,
Kim
SH
,
Kim
HR
,
Zhang
K
,
Pan
Y
, et al
.
Skin-resident natural killer T cells participate in cutaneous allergic inflammation in atopic dermatitis
.
J Allergy Clin Immunol
.
2021
;
147
(
5
):
1764
77
.
86.
Lee
SW
,
Park
HJ
,
Van Kaer
L
,
Hong
S
.
Roles and therapeutic potential of CD1d-Restricted NKT cells in inflammatory skin diseases
.
Front Immunol
.
2022
;
13
:
979370
.
87.
Elkhal
A
,
Pichavant
M
,
He
R
,
Scott
J
,
Meyer
E
,
Goya
S
, et al
.
CD1d restricted natural killer T cells are not required for allergic skin inflammation
.
J Allergy Clin Immunol
.
2006
;
118
(
6
):
1363
8
.
88.
Kasraie
S
,
Werfel
T
.
Role of macrophages in the pathogenesis of atopic dermatitis
.
Mediators Inflamm
.
2013
;
2013
:
942375
.
89.
Atsumi
Y
,
Toriyama
M
,
Kato
H
,
Nakamura
M
,
Morita
A
,
Takaishi
M
, et al
.
Anti-inflammatory role of TRPV4 in human macrophages
.
Immunohorizons
.
2023
;
7
(
1
):
81
96
.
90.
Kwak
EJ
,
Hong
JY
,
Kim
MN
,
Kim
SY
,
Kim
SH
,
Park
CO
, et al
.
Chitinase 3-like 1 drives allergic skin inflammation via Th2 immunity and M2 macrophage activation
.
Clin Exp Allergy
.
2019
;
49
(
11
):
1464
74
.
91.
Mommert
S
,
Schaper
JT
,
Schaper-Gerhardt
K
,
Gutzmer
R
,
Werfel
T
.
Histamine increases Th2 cytokine-induced CCL18 expression in human M2 macrophages
.
Int J Mol Sci
.
2021
;
22
(
21
):
11648
.
92.
Lee
SB
,
Park
H
,
Lee
JE
,
Kim
KS
,
Jeon
YH
.
In vivo optical reporter-gene-based imaging of macrophage infiltration of DNCB-induced atopic dermatitis
.
Int J Mol Sci
.
2020
;
21
(
17
):
6205
.
93.
Kim
BS
,
Wang
K
,
Siracusa
MC
,
Saenz
SA
,
Brestoff
JR
,
Monticelli
LA
, et al
.
Basophils promote innate lymphoid cell responses in inflamed skin
.
J Immunol
.
2014
;
193
(
7
):
3717
25
.
94.
Toyoshima
S
,
Sakamoto-Sasaki
T
,
Kurosawa
Y
,
Hayama
K
,
Matsuda
A
,
Watanabe
Y
, et al
.
miR103a-3p in extracellular vesicles from FcεRI-aggregated human mast cells enhances IL-5 production by group 2 innate lymphoid cells
.
J Allergy Clin Immunol
.
2021
;
147
(
5
):
1878
91
.
95.
Salimi
M
,
Stoger
L
,
Liu
W
,
Go
S
,
Pavord
I
,
Klenerman
P
, et al
.
Cysteinyl leukotriene E(4) activates human group 2 innate lymphoid cells and enhances the effect of prostaglandin D(2) and epithelial cytokines
.
J Allergy Clin Immunol
.
2017
;
140
(
4
):
1090
100.e11
.
96.
Imai
Y
,
Yasuda
K
,
Nagai
M
,
Kusakabe
M
,
Kubo
M
,
Nakanishi
K
, et al
.
IL-33-Induced atopic dermatitis-like inflammation in mice is mediated by group 2 innate lymphoid cells in concert with basophils
.
J Invest Dermatol
.
2019
;
139
(
10
):
2185
94.e3
.
97.
Iype
J
,
Fux
M
.
Basophils orchestrating eosinophils’ chemotaxis and function in allergic inflammation
.
Cells
.
2021
;
10
(
4
):
895
.
98.
Nakagome
K
,
Nagata
M
.
Innate immune responses by respiratory viruses, including rhinovirus, during asthma exacerbation
.
Front Immunol
.
2022
;
13
:
865973
.
99.
Didovic
S
,
Opitz
FV
,
Holzmann
B
,
Forster
I
,
Weighardt
H
.
Requirement of MyD88 signaling in keratinocytes for Langerhans cell migration and initiation of atopic dermatitis-like symptoms in mice
.
Eur J Immunol
.
2016
;
46
(
4
):
981
92
.
100.
Tanei
R
,
Hasegawa
Y
.
Immunological pathomechanisms of spongiotic dermatitis in skin lesions of atopic dermatitis
.
Int J Mol Sci
.
2022
;
23
(
12
):
6682
.
101.
Yang
G
,
Seok
JK
,
Kang
HC
,
Cho
YY
,
Lee
HS
,
Lee
JY
.
Skin barrier abnormalities and immune dysfunction in atopic dermatitis
.
Int J Mol Sci
.
2020
;
21
(
8
):
2867
.
102.
Guseva
D
,
Rudrich
U
,
Kotnik
N
,
Gehring
M
,
Patsinakidis
N
,
Agelopoulos
K
, et al
.
Neuronal branching of sensory neurons is associated with BDNF-positive eosinophils in atopic dermatitis
.
Clin Exp Allergy
.
2020
;
50
(
5
):
577
84
.
103.
Vander Does
A
,
Ju
T
,
Mohsin
N
,
Chopra
D
,
Yosipovitch
G
.
How to get rid of itching
.
Pharmacol Ther
.
2023
;
243
:
108355
.
104.
Nakashima
C
,
Ishida
Y
,
Kitoh
A
,
Otsuka
A
,
Kabashima
K
.
Interaction of peripheral nerves and mast cells, eosinophils, and basophils in the development of pruritus
.
Exp Dermatol
.
2019
;
28
(
12
):
1405
11
.
105.
Hashimoto
T
,
Rosen
JD
,
Sanders
KM
,
Yosipovitch
G
.
Possible role of neutrophils in itch
.
Itch
.
2018
;
3
(
4
):
e17
.
106.
Pavlenko
D
,
Todurga Seven
Z
,
Bystrom
L
,
Markan
A
,
Verpile
R
,
Ishida
H
, et al
.
Crisaborole inhibits itch and pain by preventing neutrophil infiltration in a mouse model of atopic dermatitis
.
Acta Derm Venereol
.
2023
;
103
:
adv13382
.
107.
Walsh
CM
,
Hill
RZ
,
Schwendinger-Schreck
J
,
Deguine
J
,
Brock
EC
,
Kucirek
N
, et al
.
Neutrophils promote CXCR3-dependent itch in the development of atopic dermatitis
.
Elife
.
2019
;
8
:
e48448
.
108.
Wilson
SR
,
The
L
,
Batia
LM
,
Beattie
K
,
Katibah
GE
,
McClain
SP
, et al
.
The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch
.
Cell
.
2013
;
155
(
2
):
285
95
.
109.
Jariwala
SP
,
Abrams
E
,
Benson
A
,
Fodeman
J
,
Zheng
T
.
The role of thymic stromal lymphopoietin in the immunopathogenesis of atopic dermatitis
.
Clin Exp Allergy
.
2011
;
41
(
11
):
1515
20
.
110.
Hashimoto
T
,
Yokozeki
H
,
Karasuyama
H
,
Satoh
T
.
IL-31-generating network in atopic dermatitis comprising macrophages, basophils, thymic stromal lymphopoietin, and periostin
.
J Allergy Clin Immunol
.
2023
;
151
(
3
):
737
46.e6
.
111.
Schmid-Grendelmeier
P
,
Simon
D
,
Simon
HU
,
Akdis
CA
,
Wüthrich
B
.
Epidemiology, clinical features, and immunology of the “intrinsic” (non-IgE-mediated) type of atopic dermatitis (constitutional dermatitis)
.
Allergy
.
2001
;
56
(
9
):
841
9
.
112.
Ott
H
,
Stanzel
S
,
Ocklenburg
C
,
Merk
HF
,
Baron
JM
,
Lehmann
S
.
Total serum IgE as a parameter to differentiate between intrinsic and extrinsic atopic dermatitis in children
.
Acta Derm Venereol
.
2009
;
89
(
3
):
257
61
.
113.
Park
CO
,
Lee
HJ
,
Lee
JH
,
Wu
WH
,
Chang
NS
,
Hua
L
, et al
.
Increased expression of CC chemokine ligand 18 in extrinsic atopic dermatitis patients
.
Exp Dermatol
.
2008
;
17
(
1
):
24
9
.
114.
Gittler
JK
,
Shemer
A
,
Suárez-Fariñas
M
,
Fuentes-Duculan
J
,
Gulewicz
KJ
,
Wang
CQ
, et al
.
Progressive activation of T(H)2/T(H)22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis
.
J Allergy Clin Immunol
.
2012
;
130
(
6
):
1344
54
.
115.
Lee
HH
,
Patel
KR
,
Singam
V
,
Rastogi
S
,
Silverberg
JI
.
A systematic review and meta-analysis of the prevalence and phenotype of adult-onset atopic dermatitis
.
J Am Acad Dermatol
.
2019
;
80
(
6
):
1526
32.e7
.