Skip to main content
Springer logoLink to Springer
. 2024 Sep 18;67(1-3):21–39. doi: 10.1007/s12016-024-09004-3

Neutrophils in Atopic Dermatitis

Chih-Chao Chiang 1,2,#, Wei-Jen Cheng 3,4,#, Joseph Renz Marion Santiago Dela Cruz 5,#, Thiyagarajan Raviraj 6, Nan-Lin Wu 7,8,, Michal Korinek 6,9,, Tsong-Long Hwang 5,9,10,11,
PMCID: PMC11638293  PMID: 39294505

Abstract

Neutrophils have a critical role in inflammation. Recent studies have identified their distinctive presence in certain types of atopic dermatitis (AD), yet their exact function remains unclear. This review aims to compile studies elucidating the role of neutrophils in AD pathophysiology. Proteins released by neutrophils, including myeloperoxidase, elastase, and lipocalin, contribute to pruritus progression in AD. Neutrophilic oxidative stress and the formation of neutrophil extracellular traps may further worsen AD. Elevated neutrophil elastase and high-mobility group box 1 protein expression in AD patients' skin exacerbates epidermal barrier defects. Neutrophil-mast cell interactions in allergic inflammation steer the immunological response toward Th2 imbalance and activate the Th17 pathway, particularly in response to allergens or infections linked to AD. Notably, drugs alleviating pruritic symptoms in AD inhibit neutrophilic inflammation. In conclusion, these findings underscore that neutrophils may be therapeutic targets for AD symptoms, emphasizing their inclusion in AD treatment strategies.

Keywords: Neutrophil; Atopic dermatitis; Inflammation; Pruritus, Skin lesions

Introduction

The immune response is a cascade of defense systems that protect the body from pathogens and foreign substances. It triggers extravasation to alert and recruit nearby immune cells to arrive at a localized area and prevent the further spread of an infection. Neutrophils migrate to the inflammatory tissue in a cascade-like manner. These cells are the most abundant leukocytes in circulation and are capable of protecting the body through phagocytosis, formation of neutrophil extracellular traps (NETs), generating reactive oxygen species (ROS), and/or release of granules [1, 2]. Neutrophils play an essential role in skin inflammation, migrating rapidly from the bloodstream to the inflamed site via chemotaxis. In the tissue, they move in an amoeboid fashion, responding to various chemokines and chemoattractants. The migration involves a complex interaction with other cells and the microenvironment. Neutrophils can also form swarms, enhancing their recruitment to the affected area [3]. Although neutrophils protect our body against infection or abnormal cells, they are also considered a double-edged sword. When immune cells are deregulated, confusion in the body causes uncontrolled inflammation, which leads to severe tissue damage [4]. Neutrophils play a role in acute and chronic diseases and autoimmune diseases [5, 6]. Uncontrolled neutrophils are also the culprit for allergic sensitization and allergic inflammation [7]. Allergic inflammation, such as atopic dermatitis (AD), is usually accompanied by itching, swelling, burning sensation, redness, mucus production, and pain [8]. The global prevalence of AD is estimated at 2.6%, impacting a total of 204.05 million individuals. This includes 101.27 million adults and 102.78 million children worldwide, with prevalence rates of 2.0% and 4.0%, respectively [9]. Notably, females exhibit a higher susceptibility, with a prevalence of 2.8%, affecting 108.29 million individuals, compared to males, with a prevalence of 2.4%, impacting 95.76 million individuals [10]. The exact cause of AD is unknown, but several risk factors such as family history of hypersensitivity, genetics (disruption of skin barrier function), exposure to environmental pollutants or irritants, hypersensitivity to food, or excessive hygiene are found to contribute to the development of this condition [11]. Furthermore, pruritogens from leukocyte degranulation were also reported to play a significant role in the pathology of AD inflammation [12].

Early studies suggested that neutrophils were not involved in the pathophysiology of AD causing their potential role in AD pathogenesis to be underestimated and unexplored for years. However, emerging evidence has shown that neutrophils, even in a small number, could exacerbate symptoms in AD [1315]. Of importance, translational studies have demonstrated a significant presence of neutrophils in skin lesions of Asian AD when compared to those of the European American (EA) AD [16]. Furthermore, the intrinsic type of AD shows an elevated level of neutrophil elastase (NE) in skin lesions, whereas the extrinsic AD shows an elevated eosinophil and plasmacytoid dendritic cells (DCs) [17]. Despite recent findings, the connection between neutrophils and AD has yet to be thoroughly analyzed and summarized. Thus, the primary objective of this study is to systematically review and explore the available literature on the mechanism of neutrophils in AD pathology and their clinical impact. We hope this review can shed light on and encourage future researchers to study neutrophils' importance in AD further.

Atopic Dermatitis and Its Endotypes

The pathogenesis of AD involves complicated interactions of genes, immunity, and environment, although their relationship still needs to be fully understood [18]. A notable immune feature of AD is the presence of Th2 cells (expressing cytokines such as IL-4 and IL-13) in skin lesions and elevated concentrations of immunoglobulin E (IgE) in serum [19, 20]. The majority of affected patients exhibit increased IgE antibodies that specifically target allergens in the air, such as house dust mites, pollen, and animal dander [21]. AD is commonly found in early childhood but can manifest later in life. Symptoms may be resolved before adulthood; However, a certain percentage of the population will continue to experience this condition or may develop other atopic conditions, such as asthma and allergic rhinitis, in adulthood [22].

Clinical manifestations of AD vary among different age groups. Typically, infants develop lesions on their face, extensor sites of limbs, and trunk, but the diaper area is usually spared. In adolescents and adults, AD lesions are mainly on the flexural sites, wrists, ankles, hands, upper trunk, shoulders, neck, and head [23]. A study on early-onset pediatric AD showed that children with AD exhibited more significant epidermal hyperplasia and higher expression of Th17-related cytokines and interleukin-8 (IL-8) compared to adults with AD. Furthermore, filaggrin downregulation, a characteristic of AD in adults, was absent in pediatric AD. Another feature of adult AD that differs from that of pediatric AD is the reduction of antimicrobial peptide (AMP) levels. Pediatric AD is characterized by increased AMP levels (LL37, DEFB4B, and lipocalin 2) in both non-lesion and lesion skin, which is even higher than that observed in patients with psoriasis [24]. Moreover, while pediatric AD is clustered around patients with psoriasis, adult AD is not [25]. These findings suggest that pediatric AD's skin phenotype significantly differs from adult AD.

The differentiation between extrinsic AD and intrinsic AD is based on IgE-mediated sensitization. Intrinsic AD, or non-allergic atopic eczema/dermatitis syndrome, has normal IgE levels and lacks other atopic conditions unrelated to allergens [26]. These two endotypes exhibit nearly identical Th1 and Th2 responses, however, intrinsic AD is distinguished by a markedly elevated Th17 and Th22 immune response. Furthermore, the mRNA expression of IL-17-regulated CCL20 and elafin, along with an increased number of neutrophils in intrinsic AD, indicates higher IL-17 activation in intrinsic lesions. On the other hand, extrinsic AD involves elevated serum IgE and an identifiable external trigger or allergen contributing to the development or exacerbation of the condition [17]. It has been reported that the incidence of intrinsic AD is higher among children than adults [27]. However, severe cases of pediatric AD show an increase in the total and specific IgE levels [28]. It is important to note that the level of IgE is associated with the degree of severity of AD [29] but is not the sole factor contributing to AD progression.

The onset of acute skin lesions is associated with significant increases in the expression of genes linked to Th2 and Th22 cytokines with minor increases in Th17-related cytokines, along with notable elevations in IL-17 and IL-22 cytokines. In chronic lesions, there is a significant increase in the expression of products associated with the Th2 response, such as IL-5, IL-13, IL-10, IL-31, CCL5, CCL13, and CCL18. At the same time, there is an elevation in Th1 response products, including IFN-γ, MX1, and CXCL9-11, without a notable increase in Th17 response products [30]. Interestingly, IL-4 and its receptor levels decrease from acute to chronic lesions [31]. The transition to chronic AD is characterized by the heightened activation of immune pathways initially upregulated in acute lesions, notably Th22 and Th2. While IL-17 cytokine expression is less pronounced in chronic AD, several IL-17-regulated genes (CCL20, PI3, and LCN) are elevated in both acute and chronic AD lesions. Increased IL-17 levels are observed in the skin lesions of AD patients, with IL-17-producing cells mainly located in the upper papillary dermis and epidermis [32, 33].

This complicated cocktail of Th pathways interaction ultimately damages the skin barrier, causing dryness and itching, which is a perfect recipe for pruritus. Scratching exacerbates damage, perpetuating the cycle of allergy-induced inflammation, dry skin, and itching in AD. Chronic condition boosts cytokine expression, fueling more inflammation and immune activity. Notably, Th17, Th1, and Th22 cells are also involved in the development of some subtypes of AD, including intrinsic AD, Asian AD, and pediatric AD [34].

The Role of Neutrophils in Atopic Dermatitis

The notable elevation of skin neutrophils is not typically prominent in AD compared to psoriasis. However, neutrophils may contribute to the inflammatory process, especially during acute disease if a secondary infection occurs [35]. Moreover, neutrophil infiltration is significantly increased, and genes encoding neutrophil chemoattractants are highly expressed in the AD lesional skin [36]. In addition, the rapid infiltration of neutrophils into irritated skin was observed [37]. Recent studies also demonstrate the apparent increase of neutrophil infiltration in the epidermis of AD, particularly in the Chinese, Japanese, and Korean AD [16].

The neutrophil-to-lymphocyte ratio (NLR), a serum inflammatory parameter, is a prognostic factor in numerous diseases such as COVID-19 [38], sepsis [39], and chronic obstructive pulmonary disease (COPD) [40]. NLR in patients with AD presents a higher level than that of an average individual [13]. Furthermore, NLR correlates with the severity of skin inflammation and lesional skin area [14]. NLR in children with severe AD is higher than that in children with mild AD [15]. Patients with AD had significantly higher neutrophil counts and NLR [41]. Therefore, elevated circulating neutrophils may be associated with AD severity, and a high NLR may serve as a parameter for AD severity.

Increasing evidence implicates neutrophils in AD's pathogenesis (Fig. 1). AD involves Th1/Th2 imbalance, with Th17 and Th22 participation. The Th2 cytokines (IL-4 and IL-5) are required for IgE synthesis but suppress Th1. Th2 exposure increases Type 2 cytokines (IL-4, IL-5, IL-9, and IL-13), vital to initial inflammation [42]. Th2 leads to decreased protective type 1 immunity against a wide range of viral, bacterial, and protozoan pathogens and facilitates uncontrolled or persistent infection [43]. Th2 cytokines have been shown to affect the mobilization of neutrophils via the Th2-STAT6-C3 complement-NETs cascade [44]. Mast cells, which are key players in AD pathogenesis, release pro-inflammatory mediators like histamine, cytokines, and proteases, that contribute to AD symptoms [29]. They also interact with T cells and dendritic cells, modulating skin inflammation [45]. Tumor Necrosis Factor-alpha (TNF-α) released by mast cells can directly prime circulating neutrophils, enhancing their ability to attach and migrate to the surrounding tissues [46]. Furthermore, these immune cells contribute to S. aureus-induced skin inflammation, leading to spongiosis, parakeratosis, and neutrophil infiltration. This suggests that S. aureus-stimulated mast cells worsen AD conditions [47]. Several mechanisms of mast cell-regulated neutrophil accumulation in tissues were described, involving the defensive response of the host against microbial infection [48]. Neutrophils prominent feature NET formation was regulated by mast cell tryptase in vivo, indicating an intimate functional communication bridge between mast cells and neutrophils may contribute to the pathogenesis of AD [49].

Fig. 1.

Fig. 1

Neutrophils in atopic dermatitis. AD originates from a complex interplay of both internal and external factors, which include allergic reactions, exposure to antigens, physical trauma, infections, and emotional stress. Immune responses are orchestrated by neutrophils, B cells, mast cells, and T cells, which release various chemokines and cytokines, such as IgE, IL-4 and IL-13, CXCL1, CXCL2, CXCL8, TNF-α, or LTB4. These substances mediate the recruitment and activation of neutrophils, a crucial component of inflammation. Neutrophils utilize multiple mechanisms during inflammation, including respiratory burst, degranulation, and the formation of neutrophil extracellular traps (NETs). In patients with AD, enhanced neutrophil activity is observed during flare-ups and colonization by S. aureus, leading to excessive oxidative stress. The respiratory burst involves the activation of NADPH oxidase (NOX2) and myeloperoxidase (MPO), which generate superoxide anion (O2•−) and reactive oxygen species (ROS). Upon activation, neutrophils release granular compounds, including neutrophil elastase (NE) and MPO, through a process known as degranulation. NETs, composed of extruded chromatin and antimicrobial compounds such as MPO and NE, contribute to inflammatory processes and are implicated in the pathogenesis of AD. Continued production of IL-17 by NETs and the subsequent stimulation of Th17 cells to release more IL-17 is a crucial process. This plays a pivotal role in initiating and enhancing neutrophil infiltration, which is a fundamental aspect of the immune response in AD. During the transition from acute to chronic lesions, there is an elevation in products related to the Th1 (including IFN-γ) and Th17 responses, which amplify the migration of neutrophils in the skin. IL-4, a regulator of neutrophil migration, is decreased in chronic AD. NETs continuously supply IL-17 and induce Th17 cells to release more IL-17, leading to T cell polarization in AD. This shift from a Th2-predominant acute phase to a Th1-characterized chronic phase facilitates increased skin neutrophil infiltration and pruritus lichenification. Lastly, neutrophils trigger itch by activating sensory neurons via CXCR3 signaling

Several cytokines are known to play a crucial role in AD development. IL-33, part of the Interleukin-1 family, is linked to genetic and functional activation of neutrophils. NE and cathepsin G activate IL-33. IL-8 plays a role in the generation of leukotriene B4 (LTB4) and cysteinyl leukotriene (LTC4) by neutrophils in patients with AD. IL-8 enhances the responsiveness of neutrophils to receptor-specific stimuli, contributing to the pathogenesis of AD. Both acute and chronic inflammatory responses in AD patients are influenced by IL-3 and IL-8. The skin levels of IL-8 in AD patients indicate the severity of inflammation, with a reduction in IL-8 levels observed in recovering patients [50]. Furthermore, IL-8 levels were significantly elevated in patients with severe AD compared to those with mild or moderate AD [51]. Interestingly, the anti-IL-33 monoclonal antibody etokimab has shown unanticipated CXCR1-dependent impacts on IL-8-driven neutrophil migration in AD [35]. Another marker, claudin-1 (CLDN1), a crucial component of the epidermal tight junction barrier, plays a significant role in human skin diseases, particularly AD. Reduced CLDN1 expression is linked to AD pathogenesis, as evidenced by AD-like skin features and increased neutrophil and macrophage recruitment in CLDN1-deficient mice. This underscores CLDN1 potential impact on AD disease development [52]. High-mobility group box 1 protein (HMGB1) has a proinflammatory function in various skin inflammation-related pathological conditions. Recent studies have found a correlation between elevated serum levels of HMGB1 in AD patients and the severity of the disease [53]. HMGB1 facilitates the recruitment of neutrophils and the formation of NETs in skin wounds. Furthermore, extracellular HMGB1 can cause tissue damage by triggering NET formation in inflammatory conditions [54, 55]. The prolonged presence of neutrophils and NETs in inflamed skin triggers oxidative stress in keratinocytes of AD patients. This stress results in the release of damage-associated molecular patterns like HMGB1, causing dysfunction in the epidermal barrier. Consequently, this dysfunction facilitates the colonization of S. aureus on the skin. The heightened S. aureus colonization, in turn, amplifies skin inflammation, thus promoting a vicious cycle in AD [56]. The nuclear HMGB1 has been described as a critical “gatekeeper” of dermal homeostasis. It suppresses skin inflammation by site-specific chromatin remodeling for the promoter region of the Il24 gene in the nucleus of keratinocytes. IL-24 stimulates acanthosis and the release of chemokines, leading to an influx of neutrophils. Interestingly, conflicting evidence shows that HMGB1 deficiency in keratinocytes led to elevated expression of CXCL1, resulting in infiltration of neutrophils observed in the ear of transgenic HMGB1 knockout mice using a 2,4-dinitrofluorobenzene-induced allergic contact dermatitis model [57]. β-Galactoside-binding protein galectin-1 (Gal-1), a class of carbohydrate-binding proteins, has elevated expression in the lesional skin of AD patients. This protein is distributed in nearly every tissue and exhibits robust and differential modulatory effects on neutrophils and T cells in AD [58]. Recombinant Gal-1 (rGal-1) treatment has been shown to reduce Th1 cytokines such as IL-1β, IFN-γ, and TNF-α, which leads to decreased neutrophil influx. Consequently, this reduction results in diminished clinical signs and skin thickness associated with AD [59].

The interplay between basophils and neutrophils is pivotal in the pathophysiology of AD. Basophils, stimulated by thymic stromal lymphopoietin (TSLP), interact with sensory neurons and T cells, leading to chronic inflammation and itch in AD skin [60]. Conversely, neutrophils are early recruited in the development of AD lesions through CXCR3 signaling, which is initiated by inflammatory cytokines such as CXCL1 and CXCL10 [61]. These neutrophils then release additional cytokines that amplify the inflammatory response. The combined actions of basophils and neutrophils drive the progression and severity of AD [60]. The reduction of basophils significantly decreases the infiltration of eosinophils and neutrophils, as well as skin thickness [62].

Neutrophils are recruited in the upper dermis and epidermis of lesional skin [36] and trigger itch in the early stages of AD [61]. It was observed that neutrophil-derived ROS and leukotrienes promote pain and/or itch in inflammatory conditions [63, 64]. Filaggrin is a crucial protein in the skin, playing a significant role in maintaining the skin barrier and hydration. In the context of AD, filaggrin dysfunction or deficiency has been strongly implicated [65]. In the AD mouse model, it was found that lesional skin has significantly increased epidermal thickness, dermal infiltration with eosinophils and neutrophils, and Il17a [66]. The enhanced presence of NETs in the skin promotes dysfunction of the skin barrier by downregulating FLG, which in return promotes S. aureus skin colonization [56].

Neutrophil Chemoattractant in Atopic Dermatitis

In the skin lesions of patients with AD, the increased expression of genes that attract neutrophils is similar to that seen in the skin lesions of psoriasis patients. Additionally, the number of neutrophils in AD skin lesions is comparable to those found in psoriasis skin lesions [36]. CXCL1 was reported to be associated with AD [67]. Th17-related biomarker CXCL1 was detected in the lesional and non-lesional skin of AD patients [68]. An analysis of the chemokine profiles from human skin biopsies revealed an increased concentration of CXCL1 in patients with AD, showing neutrophil infiltration [69]. The CXCL1 gene is highly expressed in MC903 mice with similar characteristics to human AD, suggesting that neutrophils are the first immune effectors infiltrating the lesional skin [61]. Administration of CXCL1 induces robust scratching and promotes neutrophil infiltration in mouse models. Neutrophil depletion using an anti-Gr1 antibody significantly alleviates itch-evoked scratching on the skin, attenuates CXCL10 induction, and deactivates sensory neurons via CXCR3, suggesting that neutrophils are required for the expression of itch-inducing chemokine CXCL10 [61, 70]. CXCL10 exerts pruritus on the skin by directly exciting sensory neurons through its receptor CXCR3 or by activating and recruiting immune cells such as T cells, eosinophils, monocytes, and NK cells to release inflammatory mediators that target sensory neurons [71, 72]. The participation of markers like CXCL1, CXCL2, IL-8, and CSF2 (GM-CSF) suggests a shared mechanism of neutrophilic inflammation in psoriasis and AD. A potential contributing factor in AD may involve tissue damage, resulting in the release of neutrophil-chemotactic factors. The potent chemokine IL-8, also known as CXCL8, was identified in the lesional stratum corneum of AD patients and showed a correlation with skin-barrier dysfunction. This chemokine can activate multiple signaling pathways by binding to CXCR1 and CXCR2 receptors expressed on human neutrophils [73]. Furthermore, CD4 + TRM cells enhance the expression of INF-γ and TNF-α, potentially leading to the upregulation of CXCL1 level. This, in turn, attracts neutrophils and contributes to the chronic recurrent inflammation observed in AD [37].

Leukotrienes (LTs) recruit and activate an array of leukocytes, including neutrophils, in several acute and chronic inflammatory reactions [74, 75]. LTs are pruritogens abundantly expressed in skin lesions of patients with AD [76]. These lipid mediators belong to the eicosanoid inflammatory mediators released by several immune cells, such as basophils and mast cells. They are known to induce pruritus in AD [7779]. Patients with atopic disease have significant LT levels such as LTB4 in the skin lesions [80] and serum [81] and LTE4 in urine [82]. Repeated mechanical abrasion of the skin in AD resulted in robust neutrophil infiltration and increased levels of LTB4 and its receptor, BLT1, in the dermis. In an AD mouse model, LTB4 was derived from neutrophils and critical for establishing allergic skin inflammation [63].

Degranulation and Atopic Dermatitis

Neutrophilic granules are essential in different inflammatory diseases, including psoriasis [1]; however, their function in AD is still unclear. A proteomic study has found that myeloperoxidase (MPO) is upregulated in the serum of young and adult AD patients [83, 84]. It has been suggested that MPO and its ability to induce the overproduction of ROS could contribute to inflammation and pruritus in several diseases [84, 85]. The levels of NE and lipocalin-2 detected in the papillary epidermis of patients with AD were comparable to those observed in psoriasis [36]. Elevated NE activity has also been found in the lesional epidermis of AD patients [86]. NE, a neutral serine protease, can activate protease-activated receptor 2 (PAR2) and transient receptor potential vanilloid 4, thus leading to inflammation and pain [87]. PAR2 has been identified to be widely distributed throughout the human body and is expressed by several cell types present in the skin, including fibroblasts, endothelial cells, and keratinocytes [88]. PAR2 plays an essential role in altered epidermal barrier function, inflammation, and pruritic sensation of the skin, which are hallmarks of AD [89, 90]. Patients with AD exhibit higher levels of PAR-2 receptor expression than healthy donors. The percentage of PAR2+ neutrophils increases with the severity of AD [91]. Activation of PAR2 in keratinocytes via serine proteases induces the expression of ICAM-1, thymic stromal lymphopoietin, and several cytokine inflammatory mediators, which increase immune cell infiltration [90, 92]. This series of leukocyte recruitment (particularly Th2) leads to skin inflammation and pruritus [93]. These findings indicate that neutrophil granule proteases, such as MPO and NE, may have a role in the onset of AD. However, this mechanism is inferred from indirect observations, necessitating further studies to substantiate this claim.

Oxidative Stress, NETs, and Atopic Dermatitis

Oxidative stress caused by oxidants, such as ROS and superoxide anions, contributes to chronic skin inflammation. Oxidative stress significantly increases when patients with AD experience a flare-up [94]. Activated neutrophils could lead to oxidative stress due to respiratory bursts, producing ROS and superoxide anions [95]. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) and MPO are pivotal enzymes in the respiratory burst of neutrophils. Serum MPO level is significantly increased in AD patients [84].

To date, many studies have revealed that NETs play an essential role in numerous diseases such as COVID-19 [96], thrombotic disorders [97], COPD [98], cardiovascular diseases [99], cancers [100], systemic lupus erythematosus [101], rheumatoid arthritis [102], and psoriasis [1]. NETs are marginally increased in the isolated neutrophils from AD patients, and the NETs level is significantly lower than those in the neutrophils from psoriasis patients [103]. Detection of NETs in the stratum corneum of individuals with AD was confirmed through immunofluorescence staining for MPO and the enzyme 8-oxoguanine glycosylase. In vitro studies revealed that neutrophils and NETs can induce ROS production in primary human keratinocytes. The prolonged presence of neutrophils and NETs causes oxidative stress, leading to the release of HMGB1 and the activation of nuclear factor κB (NF-κB) signaling in primary human keratinocytes. This disrupts the skin barrier and promotes S. aureus colonization [56]. There is adequate evidence of the increased ROS in AD [104].

Bacterial Infection in Atopic Dermatitis Associated with Neutrophils

AD condition is associated with impaired barrier function and low levels of AMPs which could contribute to an increased susceptibility to skin infections with fungi, viruses, and bacteria (i.e., S. aureus) [105, 106]. On the other hand, although psoriasis exhibits similarly low AMP levels, it does not show an increased infection rate. Furthermore, AD has a lower level of IL-17 compared to psoriasis which is essential for stimulating AMPs and chemicals that attract neutrophils [107]. The presence of higher neutrophil counts in S. aureus-infected AD lesions, compared to uninfected AD lesions, underscores the complex immune responses in AD [108].

S. aureus is often isolated from AD lesional skin, with persistent overgrowth during a flare-up of AD [109]. Migration and phagocytosis of neutrophils were poor in patients with AD, which led patients to become more susceptible to infection [110]. Staphylococcus species colonize AD skin lesions, such as S. aureus and S. epidermidis. Bacteria form biofilms, which serve as a protection film against innate immune cells, especially macrophages and neutrophils. Biofilm formation leads to high resistance to antibiotics. Previous studies have shown that neutrophilic lysins produced by S. aureus, namely α-toxin, inhibit neutrophilic activity [111]. In contrast, other research studies have shown that neutrophil levels are enhanced in S. aureus-infected skin of AD patients. S. aureus skin infection leads to neutrophil recruitment in patients with AD [112]. A recent study indicated that the serine protease-like protein A (SplA) from S. aureus works in synergy with IL-17A to enhance IL-8 release and promote neutrophil migration in human keratinocytes of STAT3-HIES patients with clinical aspects similar to a Th2-dominated AD [113]. S. aureus can evade neutrophils-induced oxidative stress by sensing oxidants, utilizing antioxidant components, and repairing oxidative damage [114]. Moreover, inhibition of NETs formation by pretreatment of neutrophils with PAD4 inhibitors or DNase impairs the survival of S. aureus in the co-culture model with keratinocytes, suggesting NETs released by neutrophils could be responsible for enhanced S. aureus colonization in skin [115]. These contrasting findings highlight the complexity and heterogeneity of AD. The variability in immune responses emphasizes the multifaceted nature of AD, making it a challenging condition to fully understand.

Human neutrophil α-defensins (HNP) and dermcidin are microbicidal peptides that can modulate innate immunity. In patients with AD, there is an increase in the levels of antimicrobial peptide dermcidin, HNP, and Th2 chemokines (CCL17, CCL22, and CCL27) in the bloodstream. Compromised innate immunity against S. aureus in the skin may allow bacteria to enter the bloodstream, triggering systemic innate immunity, particularly involving neutrophils, which leads to higher levels of HNP and dermcidin in the circulation [116]. Furthermore, Helicobacter pylori neutrophil-activating protein (HP-NAP) can regulate Th1 and Th2 immune responses. When administered through intraperitoneal injection, HP-NAP has been shown to alleviate symptoms associated with AD, including erythema and swelling. HP-NAP reduces the infiltration of lymphocytes and mast cells and the secretion of IgE, IL-4, and various inflammatory cytokines, such as IL-1β, IL-5, IL-6, and TNF-α [117]. As previously stated, neutrophils show a pro-neutrophilic response in the pathogenesis of AD. The neutrophilic inflammatory response comprises the synthesis of cytokines, the degranulation of vesicles, and the formation of ROS.

TSLP, a cytokine derived from epithelial cells, is generated in reaction to pro-inflammatory signals. The level of TSLP expression in AD skin is linked to both the severity of the disease and the extent of epidermal barrier disruption [118]. TSLP directly stimulates dendritic cells to polarize naive T cells into Th2 cells, which secrete IL-4, IL-5, and IL-13, further promoting TSLP release [119]. Activation of TSLP mediates neutrophil killing of methicillin-resistant S. aureus [120].

Drugs Used in Atopic Dermatitis Affecting Neutrophilic Function

As the incidence of AD increases, the discovery and development of new treatments are increasingly important [121]. According to the American Academy of Family Physicians (AFP) 2022, corticosteroids, particularly topical corticosteroids (TCSs), have been and remain the first-line treatment of choice for AD flare-ups [122, 123]. TCSs were first introduced in 1952 by Sulzberger and Witten and are currently available in various concentrations and potencies [124, 125]. Corticosteroids reduce skin inflammation by modifying the function of dermal and epidermal cells and leukocytes involved in skin inflammation [126]. Glucocorticoids exert their effects by binding to the classic glucocorticoid receptor (GR), with neutrophils primarily expressing the GRβ variant, unlike other tissues that predominantly express the GRα variant [127]. This elevated GRβ expression (effect of splicing factor SRp30c) in neutrophils is linked to glucocorticoid resistance observed in allergic and inflammatory diseases [128]. Ligand-activated GR modulates the transcription of thousands of genes through direct binding to DNA response elements, affecting other transcription factors and causing a wide range of effects [128]. Chronic corticosteroid use raises concerns about side effects and rebound flares, especially in pediatric patients [129]. This has driven the development of new agents with fewer adverse effects, better tolerated by young patients [130132].

The limited efficacy and significant side effects of traditional AD treatments have accelerated research into AD pathogenesis, leading to a translational revolution and rapid expansion in treatment strategies in the last decade [133]. It is important to clarify the role of each cytokine and immune pathway in AD that will lead the path to personalized medicine in the future. This review summarizes potential AD drugs and market-available products targeting neutrophils, supported by evidence from in vivo studies, human trials, and clinical research (Table 1, Fig. 2).

Table 1.

Pharmacological therapies for atopic dermatitis affecting neutrophils

Name Neutrophil target Neutrophil function Route Clinical Stage in AD Clinical Trial related to AD and neutrophils Reference
Crisaborole PDE4 Decrease chemotaxis and activation Topical FDA-approved [134]
Roflumilast Inhibit neutrophil recruitmentb Topical Clinical trial NCT04845620 (phase 3) [135, 136]
Apremilast Decrease the accumulation of neutrophilsb Oral Clinical trial NCT02087943 (phase 2) [137, 138]
Delgocitinib Pan-JAK Decrease respiratory burst and NETs Topical or oral Approved in Japan NCT03725722 (phase 2) [139]
Tofacitinib Decrease neutrophil counts Topical or oral Clinical trial NCT02001181 (phase 2) [140]
Abrocitinib JAK1 Decrease neutrophil counts Oral FDA-approved [141, 142]
Upadacitinib Decrease neutrophil counts Oral FDA-approved [143]
Baricitinib JAK1/2 Decrease neutrophil counts Oral Approved in Europe and Japan NCT03435081 (phase 3) [144]
Ruxolitinib JAK1/JAK2 Decrease neutrophil counts Topical FDA-approved [145]
Tacrolimus Calcineurin inhibitor Decrease MPO and chemotaxis Topical FDA-approved [146147]
Pimecrolimus Decrease chemotaxisc Topical FDA-approved [148]
Cyclosporine Inhibit neutrophil migration and secretion of ROSb Oral FDA-approved (off-label) NCT00445081 (phase 4) [149151]
Azathioprine Immunosuppressant (unknown) Decrease neutrophil infiltration Oral Clinical trial (recruiting) NCT05078294 [150, 152, 153]
Dupilumaba Anti-IL-4 and IL-13 mAb Decrease neutrophil counts Subcutaneous injection FDA-approved [154]
Zileuton Leukotriene pathway Inhibit adhesion and chemotaxis of neutrophils Topical or oral Clinical trial NCT03571620 (phase 2) [155, 156]
Montelukast Oral Clinical trial NCT00557284, NCT00903357 (phase 2) [157]
Etokimab Anti-IL-33 mAb (first-in-class) Decrease neutrophil migration Subcutaneous, intravenous i.v Clinical trial NCT03533751 (phase 2, terminated) [35]

FDA Food and Drug Administration, IL interleukin, JAK Janus kinase, LOX lipoxygenase, NETs neutrophil extracellular traps; PDE phosphodiesterase; mAB, monoclonal antibody, AD atopic dermatitis

aDupilumab is the only approved mAb for the treatment of AD in patients six years and older. Dupilomab has completed many Clinical Trials in AD patients, such as NCT05680298 (phase 4), NCT04345367 (phase 3), etc.

bThe neutrophil function was reported in another disease model

cThe function was derived from a study on T cells

Fig. 2.

Fig. 2

Drugs targeting neutrophils in atopic dermatitis. Several FDA-approved AD drugs or under clinical trials exhibit anti-neutrophilic inflammatory activity. PDE4 inhibitors such as crisaborole and roflumilast inhibit neutrophil chemotaxis. Pan-JAK inhibitors such as delgocitinib, tofacitinib, abrocitinib, ruxolitinib, and baricitinib attenuate respiratory burst and NETs formation in activated neutrophils. Calcineurin inhibitors such as cyclosporine and tacrolimus reduce NETs formation and release of various proinflammatory cytokines. 5-Lipoxygenase inhibitors such as zileuton inhibit adhesion and chemotaxis of neutrophils. Apart from exhibiting antioxidant effects, N-acetylcysteine also ameliorates adhesion and respiratory burst of neutrophils. Vitamin E may inhibit neutrophil chemotaxis

Phosphodiesterase 4 Inhibitors

Phosphodiesterase 4 (PDE4), an enzyme regulating cyclic adenosine monophosphate (cAMP) levels, is present in immune cells like neutrophils and plays a key role in the inflammatory responses in AD [158]. PDE4 inhibitors effectively control neutrophilic inflammation by preventing neutrophil migration and activation [159, 160]. Crisaborole, a nonsteroidal topical PDE4 inhibitor approved by the FDA in 2016 [161] for the treatment of mild-to-moderate AD from 2 years of age and older [162164] (Table 1), has demonstrated efficacy in reducing itching in patients with AD and reducing spontaneous itch-related responses in mice with chronic atopy-like dermatitis [165, 166]. By inhibiting cAMP degradation, crisaborole reduces the production of inflammatory cytokines and chemokines leading to reduced neutrophil infiltration in AD [167]. The accumulation of intracellular cAMP inhibits the inflammatory reactions of innate and adaptive immune cells, thereby reducing the production of various inflammatory cytokines and AD-related pathways (Th1, Th2, Th17, and Th22) [159]. Crisaborole prevented infiltration of neutrophils in the skin and neutrophilic chemokine expressions such as CXCL1, CXCL2, and CXCL5, thus suppressing itch in a mouse model of AD. Despite its benefits, crisaborole did not significantly affect the density of epidermal nerve fibers even though previous findings demonstrating an increase in animal models of persistent itch [134] and AD patients [168]. Thus, the success story of crisaborole highlights the potential of PDE4 inhibitors in targeting neutrophil-related inflammation in AD.

Roflumilast (Table 1), another PDE4 inhibitor, is currently in phase 3 clinical trial for treating AD. It has already been approved for treating psoriasis by reducing neutrophil infiltration and treating COPD by impeding the migration of neutrophils in the lungs [135]. Topical application of a PDE4 inhibitor attenuates the expression of inflammatory cytokines and chemokines and prevents neutrophil infiltration in mouse models of psoriasis, though similar data for AD is still pending [169]. Apremilast (Table 1), an FDA-approved PDE4 inhibitor for psoriasis and ulcers in Behçet’s disease, is currently in phase 2 clinical trials for AD. Its potential to modulate neutrophil activity makes it a promising candidate for AD treatment [137]. Apremilast reduced neutrophil activation, including NETosis in Behçet’s disease, an autoimmune disease with unclear onset [170]. PDE4 inhibitors, particularly crisaborole, significantly modulate neutrophil function and show considerable promise in treating AD, making them a key target for further therapeutic development.

Janus Kinase Inhibitors

Janus kinase (JAK) inhibitors are novel drugs for treating autoimmune diseases such as rheumatoid or psoriatic arthritis [171]. This type of drug regulates the activity of JAKs and disrupts the receptor-mediated signaling of various cytokines with proinflammatory activity [172]. Together with signal transducer and activator of transcription proteins (STATs) they modulate various key pathways (cytokines) in AD, including Th1 (IFN-γ, IL-2, TNF-β), Th2 (IL-5, IL-4, IL-13), Th17 (IL-17A, IL-17F, IL-21), and Th22 (IL-22) [132]. There are four members of JAK-STAT family, JAK1, JAK2, JAK3, and TYK2 (tyrosine kinase 2). Their phosphorylation activates STAT1 to STAT6, which after translocation to the nucleus, leads to downstream activation of the above-mentioned cytokines. For instance, Th1 differentiation, IL-13 and IL-31 signaling is mediated by JAK1/2, while IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 is mediated by JAK1 and JAK3. IL-4 is mediated by STAT3, STAT5, and STAT6 [173]. JAK1 inhibition shows potential in controlling itch, with substantial antipruritic effects observed. Proposed mechanisms include the activation of JAK1 via IL-4Rα stimulation on sensory neurons, which may drive itch transmission [174]. Importantly, pan-JAK inhibitors inhibited respiratory bursts and NET formation in neutrophils [175].

Topical delgocitinib (Table 1), a pan-JAK inhibitor, has been approved for the treatment of AD (Corectim®) in Japan [139] and completed clinical trials in the USA (NCT03725722, phase 2). In addition, a pan-JAK inhibitor, tofacitinib, was found to decrease neutrophil counts [140]. Furthermore, several other topical and oral pan-JAK inhibitors were found effective in treatment and reducing the severity of AD symptoms, showing rapid and pro-longed anti-pruritic effects (Table 1) [132, 176].

Oral selective JAK1 inhibitor abrocitinib (Table 1) was approved by the FDA in 2022 for moderate to severe AD treatment in adults and was effective at 100 mg to 200 mg and well tolerated in patients with moderate to severe AD [177]. Abrocitinib decreased neutrophil counts, probably due to IL‐6 signaling inhibition via JAK1 [141, 142]. Another JAK1 inhibitor, upadacitinib, is an FDA-approved oral drug for treating AD for patients older than 12 years old [143], that is still undergoing over 20 clinical trials (NCT03661138, NCT03738397) [178].

Ruxolitinib, an FDA-approved topical JAK1/JAK2 inhibitor for treating mild to moderate AD, was proven to affect neutrophil counts [145]. Baricitinib (LY3009104) is an oral JAK-1/2 inhibitor approved in Europe and Japan for treating moderate to severe AD in adult patients with positive results in many clinical trials (e.g., NCT03435081 phase 3) [144]. Combining baricitinib with corticosteroids could improve the signs and symptoms of moderate-to-severe AD in both pediatric and adult patients [149, 179]. To summarize, topical pan-JAK and oral JAK1 inhibitors may alleviate AD symptoms due to their anti-neutrophil-inflammatory effects.

Calcineurin Inhibitors

Calcineurin inhibitors can inhibit T cell activation and proliferation as well as the production and release of various proinflammatory cytokines [180]. Tacrolimus (FK506) [181] and pimecrolimus [148] are available topical calcineurin inhibitors that the FDA approved to treat moderate-to-severe AD. Studies have shown that tacrolimus has an impact on neutrophil activity in AD, leading to various effects such as reduced numbers of neutrophil granulocytes, decreased production of MPO, impaired chemotaxis of neutrophils, as well as dysregulation of CXCR4 and IL-4 [146]. Furthermore, tacrolimus attenuates NETs formation in allogeneic hematopoietic stem cell transplant recipients, thus impairing neutrophil function [182]. Numerous clinical trials have demonstrated the effectiveness of pimecrolimus (Table 1), an immunosuppressant drug of the calcineurin inhibitor in treating AD [148]. However, no direct evidence exists regarding its impact on neutrophils in AD models. Oral cyclosporine (Table 1), a systemic calcineurin inhibitor, effectively treats severe AD [183]. In addition to its effects on T cells, cyclosporine influences innate immune cells like neutrophils. It can inhibit various neutrophil processes, including the production of ROS and formation of NETs, and even demonstrated anti-fungal properties [184, 185]. However, there is currently no direct evidence available regarding the impact of cyclosporine on neutrophils in AD models. Methotrexate and azathioprine showed improvement in clinical trials with AD patients, whereas azathioprine showed better evidence [150]. In a case study involving an AD patient, treatment with azathioprine resulted in an improvement of AD symptoms accompanied by a reduction of 40% in both leucocyte and neutrophil counts. The reductions observed in leucocyte and neutrophil counts were reversed upon ceasing the treatment [152].

Thymodepressin is an immunosuppressive agent that is used to treat autoimmune and allergic conditions caused by lymphocyte-mediated hyperimmune reactions. In skin biopsy samples obtained from patients with AD, thymodepressin was observed to reduce the count of Ki-67-positive nuclei in keratinocytes and alleviate symptoms such as sleep disturbances, itching, erythema, and desquamation [104]. The mechanisms by which oral and topical calcineurin inhibitors treat AD might be related to their anti-neutrophilic effects and deserve more attention and research in the future.

Leukotriene Pathway Inhibitors

The LTs pathway is crucial in AD [186]. Montelukast, an oral cysteinyl-leukotriene-1 receptor antagonist, inhibits neutrophilic inflammation by regulating the cAMP pathway [187]. Montelukast has been approved by the FDA for treating asthma [188] but showed only a weak efficacy in children with AD in a Korean clinical trial (NCT00557284) and insufficient results in US and Australian clinical trials in AD (NCT00903357 NCT00903357) [189]. Other studies also supported the off-label use of montelukast in treating AD [157]. 5-Lipoxygenase (5-LOX), an enzyme that oxidizes arachidonic acid to LTs such as LTB4, is necessary for the chemotaxis and activation of neutrophils [190]. LTB4 from activated neutrophils is crucial for allergic skin inflammation [63, 155]. In addition, in an animal study, Q301 (zileuton) cream, a 5-LOX inhibitor, could ameliorate AD skin symptoms in phase 2 clinical trials for AD (NCT03571620, NCT02426359, both completed) [156, 191]. Treatment with docosahexaenoic acid and eicosapentaenoic acid plus tacrolimus decreased infiltration of T cells, B cells, eosinophils, neutrophils, and serum IgE level via inhibition of LTB4 [155]. Moreover, dihomo-γ-linolenic acid (DGLA, DS107), which can be easily obtained by endogenous conversion from γ-linolenic acid (GLA)-rich vegetable oils, completed phase 2 clinical trials on safety and efficacy in moderate to severe AD patients (NCT02211417, NCT02864498, NCT02925793). The unsaturated fatty acids are well known for their inhibitory effects on human neutrophil function [192, 193], emphasizing the need for balanced arachidonic and unsaturated fatty acid intake [194]. Thus, drugs and nutrients targeting the LTs pathways might exert an anti-neutrophilic inflammation effect and are worthy of further development for AD treatment.

Monoclonal Antibodies

Several monoclonal antibodies (mAbs) targeting the Th2 axis have been developed for treating AD, reflecting the critical role of this pathway. Notably, IL-4 and IL-13, key Th2 cytokines, inhibit neutrophil effector functions via type I and type II IL-4Rs on the neutrophil surface [195]. Clinical improvements have been observed with fezakinumab (anti-IL-22 mAb) in severe AD cases. Other mAbs, including nemolizumab (anti-IL-31R), dupilumab (anti-IL-4R/IL-13), tralokinumab (anti-IL-13), and lebrikizumab (anti-IL-13), have also shown promising efficacy. However, their impact on neutrophil functions and broader AD subtypes warrants further investigation. Dupilumab is the first monoclonal antibody approved by the FDA in 2017 for treating moderate to severe AD with acceptable side effects [196]. Dupilumab partially decreased neutrophil count in treated patients [154] and reduced the risk of skin infection in adults with moderate-to-severe AD [197]. In 2021, dupilumab was further approved for children (6–11 years). Also in 2021, tralokinumab was approved by the FDA for the treatment of moderate to severe AD in adults. Both dupilumab and tralokinumab suppress the immune responses mediated by type 2 cytokines [198].

Bristol-Myers’s Squibb-981164 (anti-IL-31 mAb), tezepelumab (anti-TSLP mAb), and MK-8226 (anti-TSLP receptor mAb) have shown promise as potential therapies for AD [199]. However, while tezepelumab, a humanized anti-TSLP antibody, demonstrated safety in a randomized phase 2 trial (NCT00757042), it failed to significantly reduce pruritus and clinical scores (SCORAD, IGA) in AD patients (NCT03809663) [200]. This raises questions about its efficacy in this context despite its success in asthma trials [201]. Further investigation is warranted to clarify the therapeutic outcome of these monoclonal antibodies for AD treatment.

N-Acetylcysteine

N-acetylcysteine (NAC) is an antidote to acetaminophen overdose, which exerts mucolytic, antioxidant, and anti-inflammatory effects [202]. NAC inhibits chemotaxis, adhesion, and oxidative burst in human neutrophils [203]. In addition, topical application of NAC solution increased skin hydration and ameliorated transepidermal water loss in patients with AD [204]. NAC treatment enhances the formation of the skin barrier by increasing the expression of epidermal growth factor receptor, E-cadherin, and occludin (regulator of neutrophils transepithelial migration) in the skin of Flaky tail mice [205]. Therefore, topical treatment with NAC may help AD patients via its antioxidant, anti-neutrophilic, and skin-protective effects.

Nutrients and Vitamins

Nutrients, vitamins, and body homeostasis, including adequate sleep, are vital for immune system function. While genetic factors contribute to AD and other atopic conditions, nutritional deficiencies can disrupt immune responses. Long-term deficits in iron, zinc, vitamin A, and vitamin D are associated with increased Th2 response, antigen presentation, and activation of immune cells, including neutrophils and mast cells, leading to higher IgE production [206].

Zinc [207] and iron [208] supplementation has shown potential in improving AD symptoms by modulating neutrophil function, including degranulation and formation of NETs and ROS in mice [209]. Vitamin C [210], collagen [211], and probiotics [211] enhance skin hydration in AD patients, while vitamin D (clinical studies, such as NCT00789880 [212] and NCT02058186) supplementation has been linked to symptom improvement through mechanisms such as enhanced neutrophilic antimicrobial peptide production, which promotes the Th17 axis [143]. Vitamin E may help manage AD by reducing neutrophil-driven inflammation [213] and skin barrier protection [214].

Atopic Dermatitis Drugs Meta-analyses

According to the meta-analyses from 50 randomized controlled trials on AD drugs, dupilumab, azathioprine, baricitinib, and cyclosporine A, all the drugs that were reported to affect neutrophilic function (Table 1) received robust trial evidence for the efficacy and safety in adults [150]. Among topical AD treatments, meta-analyses from 10 randomized controlled trials showed tofacitinib, ruxolitinib, and delgocitinib with superior efficacy over other JAK and PDE4 inhibitors and much better than corticosteroids [215]. Another 5 randomized controlled trials showed tofacitinib with the best effect on pruritus [216]. All these drugs affected neutrophil counts in patients or suppressed the pathological effects of neutrophils in AD models (Table 1), highlighting their impact on the treatment of AD and the role of neutrophilic inflammation in AD, worth further development.

Study Limitations

Literature is scarce on where AD treatment drugs interfere with neutrophilic activity in AD samples. Nevertheless, the authors have identified several drugs in Table 1 that have been correlated with neutrophil function in AD. By highlighting these drugs, the aim is to inspire future researchers to explore the possibilities of these drugs in managing neutrophilic activities in AD.

Conclusion

Classically, AD is a chronic immune-mediated inflammatory skin disease that is mainly orchestrated by Th2 cells. The strong correlation between type 2 inflammation and AD has led to a scarcity of information regarding the potential involvement of neutrophils in the immunopathogenesis of AD. However, most of the presented data are from animal models, and only a small amount of data is derived from human AD patients. The emerging evidence offers insights into the potential roles of neutrophils in AD. Several key points have been identified: first, NLR in patients with AD is higher than that in normal individuals. Second, repeated skin scratching can promote neutrophil infiltration in AD. Third, high CXCL1 chemoattractant levels were detected in the lesional skin of patients with AD. Fourth, AD patients demonstrate increased expression of MPO, predominantly found in neutrophils and associated with oxidative stress. Fifth, NE, a serine protease released by neutrophils, activates PAR2 in keratinocytes, contributing to immune effector infiltration in AD. Sixth, neutrophil-derived oxidative stress and NETs are involved in the flare-up of AD caused by S. aureus. Lastly, certain drugs used to treat AD could affect neutrophilic inflammation. Enhancing our understanding of the pathogenic role of neutrophils in AD could facilitate the development of novel therapeutic strategies for the condition.

Materials and Methods

The PubMed and SciFinder databases were used as search engines for this literature review. Authors used the keywords “atopic dermatitis,” “atopic eczema,” ‘Th1,” and “Th2,” “neutrophils,” “neutrophil chemoattractant,” “neutrophilic degranulation,” “pruritus,” “NETs,” “oxidative stress,” and “skin inflammation” to find the articles that could potentially associate neutrophils and atopic dermatitis. There was no discrimination when selecting an article, whether the samples were human or animal models or the experiment was done in-vivo or ex-vivo. Figures 1 and 2 were created with BioRender.com.

The authors thoroughly read the abstract, methodology, results, discussion, and the article’s conclusion to gather information. EndNote ver. 20 was the citation app used to generate and manage the library reference.

Acknowledgements

We are grateful to Professor Mohamed El-Shazly for language support.

Abbreviations

AD

Atopic dermatitis

Th cells

T helper cells

COVID-2

Coronavirus disease 2019

FDA

Food and Drug Administration

LT

Leukotriene

IL

Interleukin

MPO

Myeloperoxidase

JAK

Janus kinase

LOX

Lipoxygenase

NE

Neutrophil elastase

NETs

Neutrophil extracellular traps

PDE

Phosphodiesterase

Authors’ Contributions

CCC and JRMSDC wrote the paper. CWJ drew the figures. NLW, TR and MK consulted and revised the treatment schemes and the manuscript. HTL initiated the concept and supervised the writing. All authors read and approved the final manuscript.

Funding

This review was supported by grants from the National Science and Technology Council (NSTC), Taiwan (NSTC 113-2320-B-037-023; 113-2321-B-255-001; 112-2320-B-037-012; 111-2320-B-255-006-MY3), and in part by a grant from the Kaohsiung Medical University Research Foundation (KMU-Q113011). The funders had no role in the study design, data collection, data analysis, manuscript preparation, and/or publication decisions.

Data Availability

No datasets were generated or analysed during the current study.

Declarations

Conflict of Interest

The authors declare no competing interests.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Chih-Chao Chiang, Wei-Jen Cheng, and Joseph Renz Marion Santiago Dela Cruz these authors contributed equally to this work.

Contributor Information

Nan-Lin Wu, Email: t222@mmc.edu.tw.

Michal Korinek, Email: michalk@kmu.edu.tw.

Tsong-Long Hwang, Email: htl@mail.cgust.edu.tw.

References

  • 1.Chiang CC, Cheng WJ, Korinek M, Lin CY, Hwang TL (2019) Neutrophils in psoriasis. Front Immunol 10:2376. 10.3389/fimmu.2019.02376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Burn GL, Foti A, Marsman G, Patel DF, Zychlinsky A (2021) The neutrophil. Immunity 54:1377–1391. 10.1016/j.immuni.2021.06.006 [DOI] [PubMed] [Google Scholar]
  • 3.Mihlan M, Glaser KM, Epple MW, Lämmermann T (2022) Neutrophils: amoeboid migration and swarming dynamics in tissues. Frontiers in Cell and Developmental Biology. 10.3389/fcell.2022.871789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Yang G et al (2020) Skin barrier abnormalities and immune dysfunction in atopic dermatitis. Int J Mol Sci. 10.3390/ijms21082867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yang SC, Tsai YF, Pan YL, Hwang TL (2021) Understanding the role of neutrophils in acute respiratory distress syndrome. Biomed J 44:439–446. 10.1016/j.bj.2020.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Herrero-Cervera A, Soehnlein O, Kenne E (2022) Neutrophils in chronic inflammatory diseases. Cell Mol Immunol 19:177–191. 10.1038/s41423-021-00832-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hosoki K, Itazawa T, Boldogh I, Sur S (2016) Neutrophil recruitment by allergens contribute to allergic sensitization and allergic inflammation. Curr Opin Allergy Clin Immunol 16:45–50. 10.1097/ACI.0000000000000231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dong X, Dong X (2018) Peripheral and central mechanisms of itch. Neuron 98:482–494. 10.1016/j.neuron.2018.03.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.(2023) A study about how many people around the world have atopic dermatitis. Br J Dermatol 190(1):e6. 10.1093/bjd/ljad462 [DOI] [PubMed]
  • 10.Tian J, Zhang D, Yang Y, Huang Y, Wang L, Yao X, Lu Q (2023) Global epidemiology of atopic dermatitis: a comprehensive systematic analysis and modelling study. Br J Dermatol 190:55–61. 10.1093/bjd/ljad339 [DOI] [PubMed] [Google Scholar]
  • 11.Bonamonte D et al (2019) The role of the environmental risk factors in the pathogenesis and clinical outcome of atopic dermatitis. Biomed Res Int 2019:2450605. 10.1155/2019/2450605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Murota H, Katayama I (2017) Exacerbating factors of itch in atopic dermatitis. Allergol Int 66:8–13. 10.1016/j.alit.2016.10.005 [DOI] [PubMed] [Google Scholar]
  • 13.Batmaz SB (2018) Simple markers for systemic inflammation in pediatric atopic dermatitis patients. Indian J Dermatol 63:305–310. 10.4103/ijd.IJD_427_17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Inokuchi-Sakata S et al (2021) Role of eosinophil relative count and neutrophil-to-lymphocyte ratio in the assessment of severity of atopic dermatitis. Acta Derm Venereol 101:adv00491. 10.2340/00015555-3838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dogru M, Citli R (2017) The neutrophil-lymphocyte ratio in children with atopic dermatitis: a case-control study. Clin Ter 168:e262–e265. 10.7417/T.2017.2017 [DOI] [PubMed] [Google Scholar]
  • 16.Chan TC et al (2018) Atopic dermatitis in Chinese patients shows T(H)2/T(H)17 skewing with psoriasiform features. J Allergy Clin Immunol 142:1013–1017. 10.1016/j.jaci.2018.06.016 [DOI] [PubMed] [Google Scholar]
  • 17.Suarez-Farinas M et al (2013) Intrinsic atopic dermatitis shows similar TH2 and higher TH17 immune activation compared with extrinsic atopic dermatitis. J Allergy Clin Immunol 132:361–370. 10.1016/j.jaci.2013.04.046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bertino L et al (2020). Oxidative stress and atopic dermatitis Antioxidants (Basel). 10.3390/antiox9030196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Agrawal R, Wisniewski JA, Woodfolk JA (2011) The role of regulatory T cells in atopic dermatitis. Curr Probl Dermatol 41:112–124. 10.1159/000323305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Salimian J et al (2022) Atopic dermatitis: molecular, cellular, and clinical aspects. Mol Biol Rep 49:3333–3348. 10.1007/s11033-021-07081-7 [DOI] [PubMed] [Google Scholar]
  • 21.Ng C et al (2016) Hyper IgE in childhood eczema and risk of asthma in Chinese children. Molecules. 10.3390/molecules21060753 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ellis CN, Mancini AJ, Paller AS, Simpson EL, Eichenfield LF (2012) Understanding and managing atopic dermatitis in adult patients. Semin Cutan Med Surg 31:S18-22. 10.1016/j.sder.2012.07.006 [DOI] [PubMed] [Google Scholar]
  • 23.Weidinger S, Novak N (2016) Atopic dermatitis. Lancet 387:1109–1122. 10.1016/S0140-6736(15)00149-X [DOI] [PubMed] [Google Scholar]
  • 24.Esaki H et al (2016) Early-onset pediatric atopic dermatitis is T(H)2 but also T(H)17 polarized in skin. J Allergy Clin Immunol 138:1639–1651. 10.1016/j.jaci.2016.07.013 [DOI] [PubMed] [Google Scholar]
  • 25.Bozek A, Zajac M, Krupka M (2020) Atopic dermatitis and psoriasis as overlapping syndromes. Mediators Inflamm 2020:7527859. 10.1155/2020/7527859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tokura Y (2010) Extrinsic and intrinsic types of atopic dermatitis. J Dermatol Sci 58:1–7. 10.1016/j.jdermsci.2010.02.008 [DOI] [PubMed] [Google Scholar]
  • 27.Fuiano N, Incorvaia C (2012) Dissecting the causes of atopic dermatitis in children: less foods, more mites. Allergol Int 61:231–243. 10.2332/allergolint.11-RA-0371 [DOI] [PubMed] [Google Scholar]
  • 28.Illi S et al (2004) The natural course of atopic dermatitis from birth to age 7 years and the association with asthma. J Allergy Clin Immunol 113:925–931. 10.1016/j.jaci.2004.01.778 [DOI] [PubMed] [Google Scholar]
  • 29.Liu FT, Goodarzi H, Chen HY (2011) IgE, mast cells, and eosinophils in atopic dermatitis. Clin Rev Allergy Immunol 41:298–310. 10.1007/s12016-011-8252-4 [DOI] [PubMed] [Google Scholar]
  • 30.Tsoi LC et al (2020) Progression of acute-to-chronic atopic dermatitis is associated with quantitative rather than qualitative changes in cytokine responses. J Allergy Clin Immunol 145:1406–1415. 10.1016/j.jaci.2019.11.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Scibiorek M et al (2023) IL-4Rα signalling in B cells and T cells play differential roles in acute and chronic atopic dermatitis. Sci Rep 13:144. 10.1038/s41598-022-26637-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gittler JK et al (2012) Progressive activation of TH2/TH22 cytokines and selective epidermal proteins characterizes acute and chronic atopic dermatitis. Journal of Allergy and Clinical Immunology 130:1344–1354. 10.1016/j.jaci.2012.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cesare AD, Meglio PD, Nestle FO (2008) A role for Th17 cells in the immunopathogenesis of atopic dermatitis? J Investig Dermatol 128:2569–2571. 10.1038/jid.2008.283 [DOI] [PubMed] [Google Scholar]
  • 34.Chen Y et al (2024) Atopic dermatitis and psoriasis: similarities and differences in metabolism and microbiome. Clin Rev Allergy Immunol. 10.1007/s12016-024-08995-3 [DOI] [PubMed] [Google Scholar]
  • 35.Chen Y-L et al (2019) Proof-of-concept clinical trial of etokimab shows a key role for IL-33 in atopic dermatitis pathogenesis. Science Translational Medicine 11:eaax2945. 10.1126/scitranslmed.aax2945 [DOI] [PubMed] [Google Scholar]
  • 36.Choy DF et al (2012) Comparative transcriptomic analyses of atopic dermatitis and psoriasis reveal shared neutrophilic inflammation. J Allergy Clin Immunol 130:1335-1343.e1335. 10.1016/j.jaci.2012.06.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zheng C, Cao T, Ye C, Zou Y (2023) Neutrophil recruitment by CD4 tissue-resident memory T cells induces chronic recurrent inflammation in atopic dermatitis. Clin Immunol 256:109805. 10.1016/j.clim.2023.109805 [DOI] [PubMed] [Google Scholar]
  • 38.Li CH et al (2021) Immunological map in COVID-19. J Microbiol Immunol Infect. 10.1016/j.jmii.2021.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Huang Z, Fu Z, Huang W, Huang K (2020) Prognostic value of neutrophil-to-lymphocyte ratio in sepsis: A meta-analysis. Am J Emerg Med 38:641–647. 10.1016/j.ajem.2019.10.023 [DOI] [PubMed] [Google Scholar]
  • 40.El-Gazzar AG, Kamel MH, Elbahnasy OKM, El-Naggar ME (2020) Prognostic value of platelet and neutrophil to lymphocyte ratio in COPD patients. Expert Rev Respir Med 14:111–116. 10.1080/17476348.2019.1675517 [DOI] [PubMed] [Google Scholar]
  • 41.Jiang Y, Ma W (2017) Assessment of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in atopic dermatitis patients. Med Sci Monit 23:1340–1346. 10.12659/msm.900212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Böhm I, Bauer R (1997) Th1 cells, Th2 cells and atopic dermatitis. Hautarzt 48:223–227. 10.1007/s001050050573 [DOI] [PubMed] [Google Scholar]
  • 43.Wynn TA (2015) Type 2 cytokines: mechanisms and therapeutic strategies. Nat Rev Immunol 15:271–282. 10.1038/nri3831 [DOI] [PubMed] [Google Scholar]
  • 44.Zheng Z et al (2021) Lung mesenchymal stromal cells influenced by Th2 cytokines mobilize neutrophils and facilitate metastasis by producing complement C3. Nat Commun 12:6202. 10.1038/s41467-021-26460-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rukwied R, Lischetzki G, Mcglone F, Heyer G, Schmelz M (2000) Mast cell mediators other than histamine induce pruritus in atopic dermatitis patients: a dermal microdialysis study. Br J Dermatol 142:1114–1120. 10.1046/j.1365-2133.2000.03535.x [DOI] [PubMed] [Google Scholar]
  • 46.Dudeck J et al (2021) Directional mast cell degranulation of tumor necrosis factor into blood vessels primes neutrophil extravasation. Immunity 54:468-483.e465. 10.1016/j.immuni.2020.12.017 [DOI] [PubMed] [Google Scholar]
  • 47.Nakamura Y et al (2013) Staphylococcus δ-toxin induces allergic skin disease by activating mast cells. Nature 503:397–401. 10.1038/nature12655 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Schramm R, Thorlacius H (2004) Neutrophil recruitment in mast cell-dependent inflammation: inhibitory mechanisms of glucocorticoids. Inflamm Res 53:644–652. 10.1007/s00011-004-1307-8 [DOI] [PubMed] [Google Scholar]
  • 49.Pejler G et al (2022) Mast cell tryptase potentiates neutrophil extracellular trap formation. J Innate Immun 14:433–446. 10.1159/000520972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Neuber K, Hilger RA, Konig W (1991) Interleukin-3, interleukin-8, FMLP and C5a enhance the release of leukotrienes from neutrophils of patients with atopic dermatitis. Immunology 73:83–87 [PMC free article] [PubMed] [Google Scholar]
  • 51.Kimata H, Lindley I (1994) Detection of plasma interleukin-8 in atopic dermatitis. Arch Dis Child 70:119–122. 10.1136/adc.70.2.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tokumasu R et al (2016) Dose-dependent role of claudin-1 in vivo in orchestrating features of atopic dermatitis. Proc Natl Acad Sci 113:E4061–E4068. 10.1073/pnas.1525474113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cuppari C et al (2016) HMGB1 levels in children with atopic eczema/dermatitis syndrome (AEDS). Pediatr Allergy Immunol 27:99–102. 10.1111/pai.12481 [DOI] [PubMed] [Google Scholar]
  • 54.Hoste E et al (2019) Epithelial HMGB1 delays skin wound healing and drives tumor initiation by priming neutrophils for NET formation. Cell Rep 29:2689-2701.e2684. 10.1016/j.celrep.2019.10.104 [DOI] [PubMed] [Google Scholar]
  • 55.Mu S et al (2024) SIRT1-Mediated HMGB1 deacetylation suppresses neutrophil extracellular traps related to blood–brain barrier impairment after cerebral venous thrombosis. Mol Neurobiol 61:6060–6076. 10.1007/s12035-024-03959-2 [DOI] [PubMed] [Google Scholar]
  • 56.Focken J et al (2023) Neutrophil extracellular traps enhance S aureus skin colonization by oxidative stress induction and downregulation of epidermal barrier genes. Cell Rep 42:113148. 10.1016/j.celrep.2023.113148 [DOI] [PubMed] [Google Scholar]
  • 57.Senda N et al (2021) HMGB1-mediated chromatin remodeling attenuates Il24 gene expression for the protection from allergic contact dermatitis. Proc Natl Acad Sci U S A. 10.1073/pnas.2022343118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Correa MP et al (2022) Expression pattern and immunoregulatory roles of galectin-1 and galectin-3 in atopic dermatitis and psoriasis. Inflammation 45:1133–1145. 10.1007/s10753-021-01608-7 [DOI] [PubMed] [Google Scholar]
  • 59.Corrêa MP, Andrade FEC, Gimenes AD, Gil CD (2017) Anti-inflammatory effect of galectin-1 in a murine model of atopic dermatitis. J Mol Med 95:1005–1015. 10.1007/s00109-017-1566-9 [DOI] [PubMed] [Google Scholar]
  • 60.Mali SS, Bautista DM (2021) Basophils add fuel to the flame of eczema itch. Cell 184:294–296. 10.1016/j.cell.2020.12.035 [DOI] [PubMed] [Google Scholar]
  • 61.Walsh CM et al (2019) Neutrophils promote CXCR3-dependent itch in the development of atopic dermatitis. Elife. 10.7554/eLife.48448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hou T et al (2021) IL-37 targets TSLP-primed basophils to alleviate atopic dermatitis. Int J Mol Sci 22:7393. 10.3390/ijms22147393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Oyoshi MK et al (2012) Leukotriene B4-driven neutrophil recruitment to the skin is essential for allergic skin inflammation. Immunity 37:747–758. 10.1016/j.immuni.2012.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yin C et al (2020) IL-33/ST2 induces neutrophil-dependent reactive oxygen species production and mediates gout pain. Theranostics 10:12189–12203. 10.7150/thno.48028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Moosbrugger-Martinz V et al (2022) Revisiting the roles of filaggrin in atopic dermatitis. Int J Mol Sci. 10.3390/ijms23105318 [DOI] [PMC free article] [PubMed]
  • 66.Hoff S, Oyoshi MK, Macpherson A, Geha RS (2015) The microbiota is important for IL-17A expression and neutrophil infiltration in lesional skin of Flg(ft/ft) mice. Clin Immunol 156:128–130. 10.1016/j.clim.2014.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cheung PF et al (2010) Activation of human eosinophils and epidermal keratinocytes by Th2 cytokine IL-31: implication for the immunopathogenesis of atopic dermatitis. Int Immunol 22:453–467. 10.1093/intimm/dxq027 [DOI] [PubMed] [Google Scholar]
  • 68.Ungar B et al (2017) An integrated model of atopic dermatitis biomarkers highlights the systemic nature of the disease. J Invest Dermatol 137:603–613. 10.1016/j.jid.2016.09.037 [DOI] [PubMed] [Google Scholar]
  • 69.Kalish H, Phillips TM (2012) Assessment of chemokine profiles in human skin biopsies by an immunoaffinity capillary electrophoresis chip. Methods 56:198–203. 10.1016/j.ymeth.2011.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sakai H et al (2019) Interference of skin scratching attenuates accumulation of neutrophils in murine allergic contact dermatitis model. Inflammation 42:2226–2235. 10.1007/s10753-019-01086-y [DOI] [PubMed] [Google Scholar]
  • 71.Qu L, Fu K, Yang J, Shimada SG, LaMotte RH (2015) CXCR3 chemokine receptor signaling mediates itch in experimental allergic contact dermatitis. Pain 156:1737–1746. 10.1097/j.pain.0000000000000208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vazirinejad R, Ahmadi Z, Kazemi Arababadi M, Hassanshahi G, Kennedy D (2014) The biological functions, structure and sources of CXCL10 and its outstanding part in the pathophysiology of multiple sclerosis. NeuroImmunoModulation 21:322–330. 10.1159/000357780 [DOI] [PubMed] [Google Scholar]
  • 73.Amarbayasgalan T, Takahashi H, Dekio I, Morita E (2013) Interleukin-8 content in the stratum corneum as an indicator of the severity of inflammation in the lesions of atopic dermatitis. Int Arch Allergy Immunol 160:63–74. 10.1159/000339666 [DOI] [PubMed] [Google Scholar]
  • 74.Mawhin MA et al (2018) Neutrophils recruited by leukotriene B4 induce features of plaque destabilization during endotoxaemia. Cardiovasc Res 114:1656–1666. 10.1093/cvr/cvy130 [DOI] [PubMed] [Google Scholar]
  • 75.Lammermann T et al (2013) Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature 498:371–375. 10.1038/nature12175 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Honda T, Kabashima K (2019) Prostanoids and leukotrienes in the pathophysiology of atopic dermatitis and psoriasis. Int Immunol 31:589–595. 10.1093/intimm/dxy087 [DOI] [PubMed] [Google Scholar]
  • 77.Henrickson SE (2021) Basophils getting on your nerves? Itching for clarity on flares in atopic dermatitis. Sci Immunol. 10.1126/sciimmunol.abg8047 [DOI] [PubMed] [Google Scholar]
  • 78.He SH, Zhang HY, Zeng XN, Chen D, Yang PC (2013) Mast cells and basophils are essential for allergies: mechanisms of allergic inflammation and a proposed procedure for diagnosis. Acta Pharmacol Sin 34:1270–1283. 10.1038/aps.2013.88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Voisin T et al (2021) The CysLT(2)R receptor mediates leukotriene C(4)-driven acute and chronic itch. Proc Natl Acad Sci U SA. 10.1073/pnas.2022087118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fogh K, Herlin T, Kragballe K (1989) Eicosanoids in skin of patients with atopic dermatitis: prostaglandin E2 and leukotriene B4 are present in biologically active concentrations. J Allergy Clin Immunol 83:450–455. 10.1016/0091-6749(89)90132-2 [DOI] [PubMed] [Google Scholar]
  • 81.Huang Y et al (2014) Serum metabolomics study and eicosanoid analysis of childhood atopic dermatitis based on liquid chromatography-mass spectrometry. J Proteome Res 13:5715–5723. 10.1021/pr5007069 [DOI] [PubMed] [Google Scholar]
  • 82.Kolmert J et al (2021) Urinary leukotriene E4 and prostaglandin D2 metabolites increase in adult and childhood severe asthma characterized by type 2 inflammation. A Clinical Observational Study. Am J Respir Crit Care Med 203:37–53. 10.1164/rccm.201909-1869OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.He H et al (2020) Increased cardiovascular and atherosclerosis markers in blood of older patients with atopic dermatitis. Ann Allergy Asthma Immunol 124:70–78. 10.1016/j.anai.2019.10.013 [DOI] [PubMed] [Google Scholar]
  • 84.Simonetti O et al (2021) Oxidative stress and alterations of paraoxonases in atopic dermatitis. Antioxidants (Basel). 10.3390/antiox10050697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wei Choo CY et al (2021) Oxidative stress is associated with atopic indices in relation to childhood rhinitis and asthma. J Microbiol Immunol Infect 54:466–473. 10.1016/j.jmii.2020.01.009 [DOI] [PubMed] [Google Scholar]
  • 86.Wiedow O, Wiese F, Streit V, Kalm C, Christophers E (1992) Lesional elastase activity in psoriasis, contact dermatitis, and atopic dermatitis. J Invest Dermatol 99:306–309. 10.1111/1523-1747.ep12616644 [DOI] [PubMed] [Google Scholar]
  • 87.Zhao P et al (2015) Neutrophil elastase activates protease-activated receptor-2 (PAR2) and transient receptor potential vanilloid 4 (TRPV4) to cause inflammation and pain. J Biol Chem 290:13875–13887. 10.1074/jbc.M115.642736 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Buhl T et al (2020) Protease-activated receptor-2 regulates neuro-epidermal communication in atopic dermatitis. Front Immunol 11:1740. 10.3389/fimmu.2020.01740 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Heuberger DM, Schuepbach RA (2019) Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 17:4. 10.1186/s12959-019-0194-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Barr TP et al (2019) PAR2 pepducin-based suppression of inflammation and itch in atopic dermatitis models. J Invest Dermatol 139:412–421. 10.1016/j.jid.2018.08.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Elistratova IV, Morozov SG, Zakharova IA (2016) Expression proteaseinhibitors receptor PAR-2 on neutrophils peripheral blood of patients with atopic dermatitis and their relation with heat shock proteins HSP90. RJSKD 19:53–58. 10.18821/1560-9588-2016-19-1-53-58 [Google Scholar]
  • 92.Briot A et al (2009) Kallikrein 5 induces atopic dermatitis-like lesions through PAR2-mediated thymic stromal lymphopoietin expression in Netherton syndrome. J Exp Med 206:1135–1147. 10.1084/jem.20082242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Elias PM, Wakefield JS (2014) Mechanisms of abnormal lamellar body secretion and the dysfunctional skin barrier in patients with atopic dermatitis. J Allergy Clin Immunol 134:781–791. 10.1016/j.jaci.2014.05.048. (e781) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ji H, Li XK (2016) Oxidative stress in atopic dermatitis. Oxid Med Cell Longev 2016:2721469. 10.1155/2016/2721469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.El-Benna J et al (2016) Priming of the neutrophil respiratory burst: role in host defense and inflammation. Immunol Rev 273:180–193. 10.1111/imr.12447 [DOI] [PubMed] [Google Scholar]
  • 96.Cavalcante-Silva LHA et al (2021) Neutrophils and COVID-19: the road so far. Int Immunopharmacol 90:107233. 10.1016/j.intimp.2020.107233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Martinod K, Deppermann C (2021) Immunothrombosis and thromboinflammation in host defense and disease. Platelets 32:314–324. 10.1080/09537104.2020.1817360 [DOI] [PubMed] [Google Scholar]
  • 98.Trivedi A, Khan MA, Bade G, Talwar A (2021) Orchestration of neutrophil extracellular traps (Nets), a unique innate immune function during chronic obstructive pulmonary disease (COPD) development. Biomedicines. 10.3390/biomedicines9010053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Bonaventura A, Vecchie A, Abbate A, Montecucco F (2020) Neutrophil extracellular traps and cardiovascular diseases: an update. Cells. 10.3390/cells9010231 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Teijeira A et al (2021) IL8, Neutrophils, and NETs in a collusion against cancer immunity and immunotherapy. Clin Cancer Res 27:2383–2393. 10.1158/1078-0432.CCR-20-1319 [DOI] [PubMed] [Google Scholar]
  • 101.O’Neil LJ, Kaplan MJ, Carmona-Rivera C (2019) The role of neutrophils and neutrophil extracellular traps in vascular damage in systemic lupus erythematosus. J Clin Med. 10.3390/jcm8091325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Song W, Ye J, Pan N, Tan C, Herrmann M (2020) Neutrophil extracellular traps tied to rheumatoid arthritis: points to ponder. Front Immunol 11:578129. 10.3389/fimmu.2020.578129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hu SC et al (2016) Neutrophil extracellular trap formation is increased in psoriasis and induces human beta-defensin-2 production in epidermal keratinocytes. Sci Rep 6:31119. 10.1038/srep31119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sapuntsova SG et al (2011) Status of free-radical oxidation and proliferation processes in patients with atopic dermatitis and lichen planus. Bull Exp Biol Med 150:690–692. 10.1007/s10517-011-1224-0 [DOI] [PubMed] [Google Scholar]
  • 105.Alexander H et al (2020) The role of bacterial skin infections in atopic dermatitis: expert statement and review from the International Eczema Council Skin Infection Group. Br J Dermatol 182:1331–1342. 10.1111/bjd.18643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hsu C-Y et al (2024) Antimicrobial peptides (AMPs): new perspectives on their function in dermatological diseases. Int J Pept Res Ther 30:33. 10.1007/s10989-024-10609-7 [Google Scholar]
  • 107.Sugaya M (2020) The role of Th17-related cytokines in atopic dermatitis. Int J Mol Sci 21:1314. 10.3390/ijms21041314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Dhingra N et al (2013) Attenuated neutrophil axis in atopic dermatitis compared to psoriasis reflects T<sub>H</sub>17 pathway differences between these diseases. Journal of Allergy and Clinical Immunology 132:498-501.e493. 10.1016/j.jaci.2013.04.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Iwamoto K, Moriwaki M, Miyake R, Hide M (2019) Staphylococcus aureus in atopic dermatitis: Strain-specific cell wall proteins and skin immunity. Allergol Int 68:309–315. 10.1016/j.alit.2019.02.006 [DOI] [PubMed] [Google Scholar]
  • 110.Forte WC, Guardian VC, Mantovani PA, Dionigi PC, Menezes MC (2009) Evaluation of phagocytes in atopic dermatitis. Allergol Immunopathol (Madr) 37:302–308. 10.1016/j.aller.2009.06.003 [DOI] [PubMed] [Google Scholar]
  • 111.Gonzalez T, Biagini Myers JM, Herr AB, Khurana Hershey GK (2017) Staphylococcal biofilms in atopic dermatitis. Curr Allergy Asthma Rep 17:81. 10.1007/s11882-017-0750-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Bier K, Schittek B (2021) Beneficial effects of coagulase-negative Staphylococci on Staphylococcus aureus skin colonization. Exp Dermatol 30:1442–1452. 10.1111/exd.14381 [DOI] [PubMed] [Google Scholar]
  • 113.De Donato DP et al (2024) Staphylococcus aureus Serine protease-like protein A (SplA) induces IL-8 by keratinocytes and synergizes with IL-17A. Cytokine 180:156634. 10.1016/j.cyto.2024.156634 [DOI] [PubMed] [Google Scholar]
  • 114.Beavers WN, Skaar EP (2016) Neutrophil-generated oxidative stress and protein damage in Staphylococcus aureus. Pathog Dis. 10.1093/femspd/ftw060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Bitschar K et al (2020) Staphylococcus aureus skin colonization is enhanced by the interaction of neutrophil extracellular traps with keratinocytes. J Invest Dermatol 140:1054–1065. 10.1016/j.jid.2019.10.017. (e1054) [DOI] [PubMed] [Google Scholar]
  • 116.Wong C-K, Chu IM-T, Hon K-L, Tsang MS-M, Lam CW-K (2016) Aberrant expression of bacterial pattern recognition receptor NOD2 of basophils and microbicidal peptides in atopic dermatitis. Molecules 21:471. 10.3390/molecules21040471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Guo X et al (2020) HP-NAP ameliorates OXA-induced atopic dermatitis symptoms in mice. Immunopharmacol Immunotoxicol 42:416–422. 10.1080/08923973.2020.1806869 [DOI] [PubMed] [Google Scholar]
  • 118.Cianferoni A, Spergel J (2014) The importance of TSLP in allergic disease and its role as a potential therapeutic target. Expert Rev Clin Immunol 10:1463–1474. 10.1586/1744666x.2014.967684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chieosilapatham P et al (2021) Keratinocytes: innate immune cells in atopic dermatitis. Clin Exp Immunol 204:296–309. 10.1111/cei.13575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.West EE et al (2016) A TSLP-complement axis mediates neutrophil killing of methicillin-resistant Staphylococcus aureus. Sci Immunol. 10.1126/sciimmunol.aaf8471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Eichenfield LF et al (2014) Guidelines of care for the management of atopic dermatitis: Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol 71:116–132. 10.1016/j.jaad.2014.03.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Stouffer J, Chang JG (2023) Strategies for topical corticosteroid use in children and adults with eczema. Am Fam Physician 107:133–134 [PubMed] [Google Scholar]
  • 123.Lax SJ et al (2022) Strategies for using topical corticosteroids in children and adults with eczema. The Cochrane Database of Systematic Reviews 3:CD013356. 10.1002/14651858.CD013356.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.van Halewijn KF et al (2019) Different potencies of topical corticosteroids for a better treatment strategy in children with atopic dermatitis (the Rotterdam Eczema study): protocol for an observational cohort study with an embedded randomised open-label controlled trial. BMJ Open 9:e027239. 10.1136/bmjopen-2018-027239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Sulzberger MB, Witten VH (1952) The effect of topically applied compound F in selected dermatoses. J Invest Dermatol 19:101–102. 10.1038/jid.1952.72 [DOI] [PubMed] [Google Scholar]
  • 126.Camisa C, Garofola C (2020) Topical corticosteroids. Comprehensive Dermatologic Drug Therapy, 4th edn. pp 511-527.e516. 10.1016/B978-0-323-61211-1.00045-0 [Google Scholar]
  • 127.Hauk PJ, Hamid QA, Chrousos GP, Leung DY (2000) Induction of corticosteroid insensitivity in human PBMCs by microbial superantigens. J Allergy Clin Immunol 105:782–787. 10.1067/mai.2000.105807 [DOI] [PubMed] [Google Scholar]
  • 128.Ramamoorthy S, Cidlowski JA (2016) Corticosteroids: Mechanisms of action in health and disease. Rheum Dis Clin North Am 42:15–31. 10.1016/j.rdc.2015.08.002. (vii) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Drucker AM et al (2018) Use of systemic corticosteroids for atopic dermatitis: International Eczema Council consensus statement. Br J Dermatol 178:768–775. 10.1111/bjd.15928 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Newsom M, Bashyam AM, Balogh EA, Feldman SR, Strowd LC (2020) New and emerging systemic treatments for atopic dermatitis. Drugs 80:1041–1052. 10.1007/s40265-020-01335-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Drucker AM et al (2022) Systemic immunomodulatory treatments for atopic dermatitis: update of a living systematic review and network meta-analysis. JAMA Dermatol 158:523–532. 10.1001/jamadermatol.2022.0455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.He H, Guttman-Yassky E (2019) JAK inhibitors for atopic dermatitis: an update. Am J Clin Dermatol 20:181–192. 10.1007/s40257-018-0413-2 [DOI] [PubMed] [Google Scholar]
  • 133.Facheris P, Jeffery J, Del Duca E, Guttman-Yassky E (2023) The translational revolution in atopic dermatitis: the paradigm shift from pathogenesis to treatment. Cell Mol Immunol 20:448–474. 10.1038/s41423-023-00992-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Sakai K, Sanders KM, Pavlenko D, Lozada T, Akiyama T (2021) Crisaborole prevents infiltration of neutrophils to suppress itch in a mouse model of atopic dermatitis. Itch 6:e53–e53. 10.1097/itx.0000000000000053 [Google Scholar]
  • 135.Dunne AE et al (2019) Direct inhibitory effect of the PDE4 inhibitor roflumilast on neutrophil migration in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 60:445–453. 10.1165/rcmb.2018-0065OC [DOI] [PubMed] [Google Scholar]
  • 136.Gooderham M et al (2023) The safety and efficacy of roflumilast cream 0.15% and 0.05% in patients with atopic dermatitis: randomized, double-blind, phase 2 proof of concept study. J Drugs Dermatol 22:139–1476. 10.36849/jdd.7295 [DOI] [PubMed] [Google Scholar]
  • 137.Simpson EL et al (2019) A phase 2 randomized trial of apremilast in patients with atopic dermatitis. J Invest Dermatol 139:1063–1072. 10.1016/j.jid.2018.10.043 [DOI] [PubMed] [Google Scholar]
  • 138.Perez-Aso M et al (2015) Apremilast, a novel phosphodiesterase 4 (PDE4) inhibitor, regulates inflammation through multiple cAMP downstream effectors. Arthritis Res Ther 17:249. 10.1186/s13075-015-0771-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Dhillon S (2020) Delgocitinib: first approval. Drugs 80:609–615. 10.1007/s40265-020-01291-2 [DOI] [PubMed] [Google Scholar]
  • 140.Kathuria H et al (2020) Proposome for transdermal delivery of tofacitinib. Int J Pharm 585:119558. 10.1016/j.ijpharm.2020.119558 [DOI] [PubMed] [Google Scholar]
  • 141.Perche PO, Cook MK, Feldman SR (2023) Abrocitinib: a new FDA-approved drug for moderate-to-severe atopic dermatitis. Ann Pharmacother 57:86–98. 10.1177/10600280221096713 [DOI] [PubMed] [Google Scholar]
  • 142.Schmieder GJ et al (2018) Efficacy and safety of the Janus kinase 1 inhibitor PF-04965842 in patients with moderate-to-severe psoriasis: phase II, randomized, double-blind, placebo-controlled study. Br J Dermatol 179:54–62. 10.1111/bjd.16004 [DOI] [PubMed] [Google Scholar]
  • 143.Muller S, Maintz L, Bieber T (2024) Treatment of atopic dermatitis: Recently approved drugs and advanced clinical development programs. Allergy 79:1501–1515. 10.1111/all.16009 [DOI] [PubMed] [Google Scholar]
  • 144.Hoy SM (2022) Baricitinib: a review in moderate to severe atopic dermatitis. Am J Clin Dermatol 23:409–420. 10.1007/s40257-022-00684-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Okamoto M et al (2023) The inhibition of glycolysis in T cells by a Jak inhibitor ameliorates the pathogenesis of allergic contact dermatitis in mice. J Invest Dermatol. 10.1016/j.jid.2023.03.1667 [DOI] [PubMed] [Google Scholar]
  • 146.Caproni M et al (2007) The comparative effects of tacrolimus and hydrocortisone in adult atopic dermatitis: an immunohistochemical study. Br J Dermatol 156:312–319. 10.1111/j.1365-2133.2006.07609.x [DOI] [PubMed] [Google Scholar]
  • 147.Chang K-T, Lin HY-H, Kuo C-H, Hung C-H (2016) Tacrolimus suppresses atopic dermatitis-associated cytokines and chemokines in monocytes. J Microbiol Immunol Infect 49:409–416. 10.1016/j.jmii.2014.07.006 [DOI] [PubMed] [Google Scholar]
  • 148.Prucha H et al (2013) Pimecrolimus, a topical calcineurin inhibitor used in the treatment of atopic eczema. Expert Opin Drug Metab Toxicol 9:1507–1516. 10.1517/17425255.2013.819343 [DOI] [PubMed] [Google Scholar]
  • 149.Bieber T et al (2022) Efficacy and safety of baricitinib in combination with topical corticosteroids in patients with moderate-to-severe atopic dermatitis with inadequate response, intolerance or contraindication to ciclosporin: results from a randomized, placebo-controlled, phase III clinical trial (BREEZE-AD4). Br J Dermatol 187:338–352. 10.1111/bjd.21630 [DOI] [PubMed] [Google Scholar]
  • 150.Siegels D et al (2021) Systemic treatments in the management of atopic dermatitis: A systematic review and meta-analysis. Allergy 76:1053–1076. 10.1111/all.14631 [DOI] [PubMed] [Google Scholar]
  • 151.Ina K et al (2002) Suppressive effects of cyclosporine A on neutrophils and T cells may be related to therapeutic benefits in patients with steroid-resistant ulcerative colitis. Inflamm Bowel Dis 8:1–9. 10.1097/00054725-200201000-00001 [DOI] [PubMed] [Google Scholar]
  • 152.Kham SK, Soh CK, Aw DC, Yeoh AE (2009) TPMT*26 (208F–>L), a novel mutation detected in a Chinese. Br J Clin Pharmacol 68:120–123. 10.1111/j.1365-2125.2009.03405.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Fuggle NR et al (2015) The adverse effect profile of oral azathioprine in pediatric atopic dermatitis, and recommendations for monitoring. J Am Acad Dermatol 72:108–114. 10.1016/j.jaad.2014.08.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Wollenberg A et al (2020) Laboratory safety of dupilumab in moderate-to-severe atopic dermatitis: results from three phase III trials (LIBERTY AD SOLO 1, LIBERTY AD SOLO 2, LIBERTY AD CHRONOS). Br J Dermatol 182:1120–1135. 10.1111/bjd.18434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Yoshida S, Yasutomo K, Watanabe T (2016) Treatment with DHA/EPA ameliorates atopic dermatitis-like skin disease by blocking LTB4 production. J Med Invest 63:187–191. 10.2152/jmi.63.187 [DOI] [PubMed] [Google Scholar]
  • 156.Fleischer A, Nam K, Kim J, Ahn K, Kang H, Choi J, Choi GJ, Lee S, Jung C (2019) Q301 (Zileuton) cream demonstrates superiority to vehicle in improving atopic dermatitis: Results from a phase 2A trial. JAAD 81:AB112. 10.1016/j.jaad.2019.06.427 [Google Scholar]
  • 157.Chin WK, Lee SWH (2018) A systematic review on the off-label use of montelukast in atopic dermatitis treatment. Int J Clin Pharm 40:963–976. 10.1007/s11096-018-0655-3 [DOI] [PubMed] [Google Scholar]
  • 158.Fahrbach K et al (2020) Crisaborole ointment, 2%, for treatment of patients with mild-to-moderate atopic dermatitis: systematic literature review and network meta-analysis. Dermatol Ther (Heidelb) 10:681–694. 10.1007/s13555-020-00389-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Li H, Zuo J, Tang W (2018) Phosphodiesterase-4 inhibitors for the treatment of inflammatory diseases. Front Pharmacol 9:1048. 10.3389/fphar.2018.01048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Totani L et al (2021) Type-4 phosphodiesterase (PDE4) blockade reduces NETosis in cystic fibrosis. Front Pharmacol 12:702677. 10.3389/fphar.2021.702677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Hon KLE, Chan VPY, Leung AKC (2021) Experimental drugs with the potential to treat atopic eczema. J Exp Pharmacol 13:487–498. 10.2147/JEP.S259299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Eichenfield LF et al (2017) Long-term safety of crisaborole ointment 2% in children and adults with mild to moderate atopic dermatitis. J Am Acad Dermatol 77:641–649. 10.1016/j.jaad.2017.06.010. (e645) [DOI] [PubMed] [Google Scholar]
  • 163.Zane LT et al (2016) Crisaborole topical ointment, 2% in patients ages 2 to 17 years with atopic dermatitis: a phase 1b, open-label, maximal-use systemic exposure study. Pediatr Dermatol 33:380–387. 10.1111/pde.12872 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Paller AS et al (2016) Efficacy and safety of crisaborole ointment, a novel, nonsteroidal phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in children and adults. J Am Acad Dermatol 75:494–503. 10.1016/j.jaad.2016.05.046. (e496) [DOI] [PubMed] [Google Scholar]
  • 165.Yosipovitch G et al (2018) Early relief of pruritus in atopic dermatitis with crisaborole ointment, a non-steroidal, phosphodiesterase 4 inhibitor. Acta Derm Venereol 98:484–489. 10.2340/00015555-2893 [DOI] [PubMed] [Google Scholar]
  • 166.Andoh T, Yoshida T, Kuraishi Y (2014) Topical E6005, a novel phosphodiesterase 4 inhibitor, attenuates spontaneous itch-related responses in mice with chronic atopy-like dermatitis. Exp Dermatol 23:359–361. 10.1111/exd.12377 [DOI] [PubMed] [Google Scholar]
  • 167.Zane LT et al (2016) Crisaborole and its potential role in treating atopic dermatitis: overview of early clinical studies. Immunotherapy 8:853–866. 10.2217/imt-2016-0023 [DOI] [PubMed] [Google Scholar]
  • 168.Tobin D et al (1992) Increased number of immunoreactive nerve fibers in atopic dermatitis. J Allergy Clin Immunol 90:613–622. 10.1016/0091-6749(92)90134-n [DOI] [PubMed] [Google Scholar]
  • 169.Lin ZC et al (2018) Topical application of anthranilate derivatives ameliorates psoriatic inflammation in a mouse model by inhibiting keratinocyte-derived chemokine expression and neutrophil infiltration. FASEB J. 10.1096/fj.201800354 [DOI] [PubMed] [Google Scholar]
  • 170.Le Joncour A et al (2023) Type-4 phosphodiesterase (PDE4) blockade reduces neutrophil activation in Behcet’s disease. Arthritis Rheumatol. 10.1002/art.42486 [DOI] [PubMed] [Google Scholar]
  • 171.Fragoulis GE, McInnes IB, Siebert S, JAK-inhibitors (2019) New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford) 58:43–54. 10.1093/rheumatology/key276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Dowty ME et al (2019) Janus kinase inhibitors for the treatment of rheumatoid arthritis demonstrate similar profiles of in vitro cytokine receptor inhibition. Pharmacol Res Perspect 7:e00537. 10.1002/prp2.537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Goswami R et al (2012) STAT6-dependent regulation of Th9 development. J Immunol 188:968–975. 10.4049/jimmunol.1102840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Oetjen LK et al (2017) Sensory neurons co-opt classical immune signaling pathways to mediate chronic itch. Cell 171:217–228. 10.1016/j.cell.2017.08.006. (e213) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Chokesuwattanaskul S et al (2018) 250 The effect of JAK inhibition on neutrophil killing, netosis and metabolism in rheumatoid arthritis. Rheumatology. 10.1093/rheumatology/key075.474 [Google Scholar]
  • 176.Mikhaylov D, Ungar B, Renert-Yuval Y, Guttman-Yassky E (2023) Oral Janus kinase inhibitors for atopic dermatitis. Ann Allergy Asthma Immunol. 10.1016/j.anai.2023.01.020 [DOI] [PubMed] [Google Scholar]
  • 177.Ferreira S, Guttman-Yassky E, Torres T (2020) Selective JAK1 inhibitors for the treatment of atopic dermatitis: focus on upadacitinib and abrocitinib. Am J Clin Dermatol 21:783–798. 10.1007/s40257-020-00548-6 [DOI] [PubMed] [Google Scholar]
  • 178.Guttman-Yassky E et al (2020) Upadacitinib in adults with moderate to severe atopic dermatitis: 16-week results from a randomized, placebo-controlled trial. J Allergy Clin Immunol 145:877–884. 10.1016/j.jaci.2019.11.025 [DOI] [PubMed] [Google Scholar]
  • 179.Torrelo A et al (2023) Efficacy and safety of baricitinib in combination with topical corticosteroids in pediatric patients with moderate-to-severe atopic dermatitis with inadequate response to topical corticosteroids: results from a phase 3, randomized, double-blind, placebo-controlled study (BREEZE-AD PEDS). Br J Dermatol. 10.1093/bjd/ljad096 [DOI] [PubMed] [Google Scholar]
  • 180.Carr WW (2013) Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Paediatr Drugs 15:303–310. 10.1007/s40272-013-0013-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Cury Martins J et al (2015) Topical tacrolimus for atopic dermatitis. The Cochrane Database of Systematic Reviews 2015:CD009864. 10.1002/14651858.CD009864.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Imbert S et al (2016) Calcineurin inhibitors impair neutrophil activity against Aspergillus fumigatus in allogeneic hematopoietic stem cell transplant recipients. J Allergy Clin Immunol 138:860–868. 10.1016/j.jaci.2016.02.026 [DOI] [PubMed] [Google Scholar]
  • 183.Miyano K, Tsunemi Y (2021) Current treatments for atopic dermatitis in Japan. J Dermatol 48:140–151. 10.1111/1346-8138.15730 [DOI] [PubMed] [Google Scholar]
  • 184.Gupta AK, Giaglis S, Hasler P, Hahn S (2014) Efficient neutrophil extracellular trap induction requires mobilization of both intracellular and extracellular calcium pools and is modulated by cyclosporine A. PLoS ONE 9:e97088. 10.1371/journal.pone.0097088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Liddicoat AM, Lavelle EC (2019) Modulation of innate immunity by cyclosporine A. Biochem Pharmacol 163:472–480. 10.1016/j.bcp.2019.03.022 [DOI] [PubMed] [Google Scholar]
  • 186.Yanes DA, Mosser-Goldfarb JL (2018) Emerging therapies for atopic dermatitis: The prostaglandin/leukotriene pathway. J Am Acad Dermatol 78:S71–S75. 10.1016/j.jaad.2017.12.021 [DOI] [PubMed] [Google Scholar]
  • 187.Anderson R et al (2009) Montelukast inhibits neutrophil pro-inflammatory activity by a cyclic AMP-dependent mechanism. Br J Pharmacol 156:105–115. 10.1111/j.1476-5381.2008.00012.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Trinh HKT, Lee SH, Cao TBT, Park HS (2019) Asthma pharmacotherapy: an update on leukotriene treatments. Expert Rev Respir Med 13:1169–1178. 10.1080/17476348.2019.1670640 [DOI] [PubMed] [Google Scholar]
  • 189.Jeon YH, Min TK, Yang HJ, Pyun BY (2016) A double-blind, randomized, crossover study to compare the effectiveness of montelukast on atopic dermatitis in Korean children. Allergy Asthma Immunol Res 8:305–311. 10.4168/aair.2016.8.4.305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Zschaler J, Arnhold J (2016) Impact of simultaneous stimulation of 5-lipoxygenase and myeloperoxidase in human neutrophils. Prostaglandins Leukot Essent Fatty Acids 107:12–21. 10.1016/j.plefa.2016.02.001 [DOI] [PubMed] [Google Scholar]
  • 191.Crow DW, Marsella R, Nicklin CF (2001) Double-blinded, placebo-controlled, cross-over pilot study on the efficacy of zileuton for canine atopic dermatitis. Vet Dermatol 12:189–195. 10.1046/j.0959-4493.2001.00254.x [DOI] [PubMed] [Google Scholar]
  • 192.Ziboh VA et al (2004) Suppression of leukotriene B4 generation by ex-vivo neutrophils isolated from asthma patients on dietary supplementation with gammalinolenic acid-containing borage oil: possible implication in asthma. Clin Dev Immunol 11:13–21. 10.1080/10446670410001670445 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Hwang TL et al (2009) Suppression of superoxide anion and elastase release by C18 unsaturated fatty acids in human neutrophils. J Lipid Res 50:1395–1408. 10.1194/jlr.M800574-JLR200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Suzuki N et al (2020) Association between polyunsaturated fatty acid and reactive oxygen species production of neutrophils in the general population. Nutrients. 10.3390/nu12113222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Egholm C, Heeb LEM, Impellizzieri D, Boyman O (2019) The regulatory effects of interleukin-4 receptor signaling on neutrophils in type 2 immune responses. Front Immunol 10:2507. 10.3389/fimmu.2019.02507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Tameez Ud Din A, Malik I, Arshad D, Tameez Ud Din A (2020) Dupilumab for atopic dermatitis: the silver bullet we have been searching for? Cureus 12:7565. 10.7759/cureus.7565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Fleming P, Drucker AM (2018) Risk of infection in patients with atopic dermatitis treated with dupilumab: a meta-analysis of randomized controlled trials. J Am Acad Dermatol 78:62–69. 10.1016/j.jaad.2017.09.052. (e61) [DOI] [PubMed] [Google Scholar]
  • 198.Tubau C, Puig L (2021) Therapeutic targeting of the IL-13 pathway in skin inflammation. Expert Rev Clin Immunol 17:15–25. 10.1080/1744666X.2020.1858802 [DOI] [PubMed] [Google Scholar]
  • 199.Kim J, Kim BE, Leung DYM (2019) Pathophysiology of atopic dermatitis: Clinical implications. Allergy Asthma Proc 40:84–92. 10.2500/aap.2019.40.4202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Simpson EL et al (2019) Tezepelumab, an anti-thymic stromal lymphopoietin monoclonal antibody, in the treatment of moderate to severe atopic dermatitis: a randomized phase 2a clinical trial. J Am Acad Dermatol 80:1013–1021. 10.1016/j.jaad.2018.11.059 [DOI] [PubMed] [Google Scholar]
  • 201.Pelaia C et al (2021) Tezepelumab: a potential new biological therapy for severe refractory asthma. Int J Mol Sci. 10.3390/ijms22094369 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Bavarsad Shahripour R, Harrigan MR, Alexandrov AV (2014) N-acetylcysteine (NAC) in neurological disorders: mechanisms of action and therapeutic opportunities. Brain Behav 4:108–122. 10.1002/brb3.208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Kharazmi A, Nielsen H, Schiotz PO (1988) N-acetylcysteine inhibits human neutrophil and monocyte chemotaxis and oxidative metabolism. Int J Immunopharmacol 10:39–46. 10.1016/0192-0561(88)90148-8 [DOI] [PubMed] [Google Scholar]
  • 204.Nakai K et al (2015) Effects of topical N-acetylcysteine on skin hydration/transepidermal water loss in healthy volunteers and atopic dermatitis patients. Ann Dermatol 27:450–451. 10.5021/ad.2015.27.4.450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Nakai K et al (2012) Reduced expression of epidermal growth factor receptor, E-cadherin, and occludin in the skin of flaky tail mice is due to filaggrin and loricrin deficiencies. Am J Pathol 181:969–977. 10.1016/j.ajpath.2012.06.005 [DOI] [PubMed] [Google Scholar]
  • 206.Peroni DG, Hufnagl K, Comberiati P, Roth-Walter F (2022) Lack of iron, zinc, and vitamins as a contributor to the etiology of atopic diseases. Front Nutr 9:1032481. 10.3389/fnut.2022.1032481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Gray NA, Dhana A, Stein DJ, Khumalo NP (2019) Zinc and atopic dermatitis: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol 33:1042–1050. 10.1111/jdv.15524 [DOI] [PubMed] [Google Scholar]
  • 208.Kuzmicka W et al (2022) Iron excess affects release of neutrophil extracellular traps and reactive oxygen species but does not influence other functions of neutrophils. Immunol Cell Biol 100:87–100. 10.1111/imcb.12509 [DOI] [PubMed] [Google Scholar]
  • 209.Kuzmicka W et al (2020) Zinc supplementation modulates NETs release and neutrophils’ degranulation. Nutrients. 10.3390/nu13010051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Holt S et al (2009) A pilot study of the efficacy of a vitamin C-containing showerhead on symptoms of eczema. N Z Med J 122:91–92 [PubMed] [Google Scholar]
  • 211.Camilion JV, Khanna S, Anasseri S, Laney C, Mayrovitz HN (2022) Physiological, pathological, and circadian factors impacting skin hydration. Cureus 14:e27666. 10.7759/cureus.27666 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Hata TR et al (2014) A randomized controlled double-blind investigation of the effects of vitamin D dietary supplementation in subjects with atopic dermatitis. J Eur Acad Dermatol Venereol 28:781–789. 10.1111/jdv.12176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Luostarinen R, Siegbahn A, Saldeen T (1991) Effects of dietary supplementation with vitamin E on human neutrophil chemotaxis and generation of LTB4. Ups J Med Sci 96:103–111. 10.3109/03009739109179263 [DOI] [PubMed] [Google Scholar]
  • 214.Teo CWL, Tay SHY, Tey HL, Ung YW, Yap WN (2021) Vitamin E in atopic dermatitis: from preclinical to clinical studies. Dermatology 237:553–564. 10.1159/000510653 [DOI] [PubMed] [Google Scholar]
  • 215.Zhang L, Du D, Wang L, Guo L, Jiang X (2021) Efficacy and safety of topical Janus kinase and phosphodiesterase inhibitor-4 inhibitors for the treatment of atopic dermatitis: a network meta-analysis. J Dermatol 48:1877–1883. 10.1111/1346-8138.16126 [DOI] [PubMed] [Google Scholar]
  • 216.Rodriguez-Le Roy Y, Ficheux AS, Misery L, Brenaut E (2022) Efficacy of topical and systemic treatments for atopic dermatitis on pruritus: a systematic literature review and meta-analysis. Front Med (Lausanne) 9:1079323. 10.3389/fmed.2022.1079323 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Clinical Reviews in Allergy & Immunology are provided here courtesy of Springer

RESOURCES