Abstract
Allergic diseases are acute and chronic inflammatory conditions resulting from disproportionate responses to environmental stimuli. Affecting approximately 40% of the global population, these diseases significantly contribute to morbidity and increasing healthcare costs. Allergic reactions are triggered by pollen, house dust mites, animal dander, mold, food antigens, venoms, toxins, and drugs. This review explores the pivotal role of the epithelium in the skin, lungs, and gastrointestinal tract in regulating the allergic response, and delves into the mechanisms of tissue-specific epithelial-immune interactions in this context, with recent advances highlighting their roles in the initiation, elicitation, and resolution phases of allergy. Understanding these intricate interactions at epithelial barriers is essential for developing targeted therapies to manage and treat allergic diseases.
Introduction
Allergic diseases such as allergic asthma, food allergy, allergic rhinitis, and atopic dermatitis (AD) are caused by disproportionate responses to environmental stimuli. Allergic diseases affect ~40% of the human population, causing morbidity and increased healthcare costs worldwide [1]. Allergic reactions occur when pollen, house-dust mite (HDM) products, animal dander, mold, pollen, food antigens, venoms, toxins, or drugs are inhaled, ingested, or contact an individual’s skin. Epithelial cells that line body surfaces including the skin, respiratory tract, and gastrointestinal (GI) tract are the first point of contact with allergens. Across tissues, epithelial cells sense allergens and/or are damaged by allergen proteases and produce interleukin (IL)-25, IL-33, and/or thymic stromal lymphopoietin (TSLP). These alarmins activate group 2 innate lymphoid cells (ILC2s) and CD4+ T helper Type 2 (Th2) cells that produce the Type 2 cytokines IL-4, −5, −9, and −13, which elicit an array of immune and epithelial effector functions that underlie the signs and symptoms of allergy, including epithelial hyperplasia and increased barrier permeability and mucin production [2–7]. While many of these processes are conserved across tissues, different tissue sites also have unique epithelial features, tied to their specific functions, that impact the regulation of the allergic response [2,5,8]. This review will focus on the central role that skin, lung, and gut epithelial cells play in initiating, perpetuating, and resolving allergic inflammation and the tissue-specific cues and environmental factors that regulate these processes.
Initiation of allergic responses at barrier sites
Epithelial cells as first responders
Epithelial barrier structure and function differ across tissues, which influences the initiation of the Type 2 response. The skin epidermis has a stratified squamous epithelium, composed of multiple layers of keratinized epithelial cells that create a strong barrier. The lung and GI tract both have a single columnar epithelial cell layer to facilitate gas exchange and nutrient absorption, respectively. However, the lung epithelium is composed of ciliated cells that line airways, while GI epithelial cells are organized into crypt-villus units (small intestine only) and have microvilli. The epithelial cell lineages that compose the barrier can also be tissue-specific; for example, the GI and respiratory tracts contain chemosensory tuft cells, while the skin apparently lacks this lineage [9]. However, regardless of tissue site, cytokines such as IL-25, IL-33, and TSLP, and other chemical mediators prompt the first events that underlie allergen detection and initiation of the Type 2 allergic response (Figure 1).
Figure 1. Tissue-specific epithelial structure in Type 2 Immune response.
Diverse epithelial landscapes drive Type 2 immunity across barrier tissues, including the skin (top left), lung (top right), and the gastrointestinal (GI) tract (bottom left). In the skin epithelium, atopic dermatitis (AD) is characterized by barrier disruption, allowing allergen penetration, triggering release of interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP) by keratinocytes. This results in activation of T helper type 2 (Th2) and group 2 innate lymphoid (ILC2) cells and secretion of IL-4 and IL-13, contributing to amplification of the Type 2 immune response. In the lung epithelium, allergen exposure can induce IL-33 and IL-25 release from epithelial cells, promoting activation of Th2s and ILC2s. IL-5 signaling results in eosinophilia while IL-13 acts on goblet cells to promote mucus secretion. Additionally, lipoxin A4 can inhibit mucus secretion and dampen inflammation in the airway epithelium. Lastly, in the GI epithelium, IL-33 and TSLP are also secreted by epithelial cells, while IL-25 is secreted specifically by tuft cells, all of which ultimately amplify Th2 and ILC2 responses and Type 2 inflammation in the gut.
For example, in a murine model of food-induced anaphylaxis following skin sensitization, intestinal epithelial cell-derived IL-25, most likely from tuft cells, the sole source of intestinal epithelial IL-25 [10], was required for ILC2 IL-4/IL-13 production that expanded mast cells to cause anaphylaxis [11]. Similarly, during murine intestinal helminth infection that also elicits Type 2 inflammation, tuft cells released IL-25 that activated ILC2s to produce IL-13, which acted back on the epithelium to promote goblet and tuft cell hyperplasia and increased mucin production that supports helminth expulsion [8,10,12,13]. These data underscore the importance of tuft cell-derived IL-25 in the intestine, though IL-25 also activated pro-allergic ILC2s in the murine lung and induced allergic airway inflammation [14], and keratinocyte-derived IL-25 prompted ILC2 IL-13 expression associated with epithelial hyperplasia in a mouse model of AD [15].
TSLP and IL-33 play key roles in promoting the allergic cascade across tissues. In a murine model of skin sensitization to food allergens, epithelial-derived TSLP and/or IL-33 promoted ILC2 responses to drive anaphylaxis or upper GI inflammation similar to human eosinophilic esophagitis (EoE), an allergic disease of the esophagus [11,16,17]. Following early-life colonization with the skin commensal Staphylococcus lentus, keratinocyte-derived TSLP mobilized ILC2s that drove AD in adult mice [18]. In the murine and human lung, IL-33 has been shown to promote the migration, proliferation, and cytokine producing capacity of pro-allergic lung ILC2s in allergy [19,20]. Together, these studies and others underscore cross-tissue and tissue-specific modules of inflammation that underlie allergic disease mediated by epithelial-derived cytokines (Figure 1).
Epithelial cells also express pattern recognition receptors that allow them to directly respond to allergens [21]. Protease allergens in fungi and HDM degrade epithelial junction proteins to elicit alarmin release and cleave fibrinogen, reviewed in detail in ref [22]. Cleaved fibrinogen and other allergens such as HDM could also signal directly through Toll-like receptor 4 (TLR4) to induce allergic airway inflammation in mice [23,24]. Furthermore, IL-33 was directly cleaved by protease allergens, activating ILC2s and causing eosinophilia in the airway, promoting murine allergic inflammation [25]. However, future studies in humans and murine models will be required to fully understand how direct interactions between epithelial cells and allergens activate the barrier to promote cytokine release in a tissue-specific manner.
Antigen presentation
Antigen uptake, processing, and presentation to allergen-specific CD4+ T cells by dendritic cells (DCs) is highly dependent on crosstalk with epithelial cells and underlies the sensitization phase as the adaptive immune response is trained to inappropriately respond to environmental allergens [3]. In murine allergic asthma models, epithelial cell-derived IL-33, TSLP, IL-1a, colony-stimulating factor 1, and granulocyte-monocyte-colony stimulating factor (GM-CSF) acted on classical DC2s [26–28] that have been shown to preferentially drive Th2 polarization [3,29,30]. TSLP induced expression of OX40 ligand on human DCs that promoted Th2 polarization [31] that could drive allergic responses. Similarly, in human skin from AD patients, keratinocytes released TSLP, IL-1, tumor necrosis factor α, GM-CSF, and IL-18 to regulate the maturation of antigen-loaded DCs [32].
While DCs play the preeminent role in Th2 polarization in allergy [29] (Figure 2), some epithelial cells can also express major histocompatibility complex class II (MHC-II) and act as non-professional antigen presenting cells [8,33]. Recent studies have identified functional roles for MHC-II+ epithelial cells in interactions with the microbiota, during graft-versus-host disease, preserving lung epithelial barrier integrity, and in cancer [34–37]. In allergy, a human esophageal epithelial cell line could process antigen and activate T cells in vitro under conditions similar to those found in the esophagus of patients with EoE [38], and human nasal epithelial cells from patients with allergic rhinitis were able to activate autologous T cells when stimulated with birch pollen antigen [39]. The study of the role of MHC-II+ epithelial cells in allergy remains an active area of investigation, with the potential to reveal novel mechanisms by which the epithelium regulates the initiation or promulgation of adaptive allergic responses.
Figure 2. Effector mechanisms of allergic inflammation in the gastrointestinal tract.
Epithelial cells are initiators and drivers of allergic inflammation upon exposure to allergens. Epithelial-derived cytokines such as interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP) play critical roles in recruiting and activating various immune cells that propagate the Type 2 response. IL-4, IL-5, IL-9, and IL-13 further promote inflammation, and IL-13 in particular acts back on the epithelium to drive a whole-tissue remodeling response. In the gastrointestinal (GI) tract, this remodeling is often associated with increased mucus production, goblet and tuft cell hyperplasia, and increased barrier permeability. Nerve growth factor (NGF); Neuromedin U (NMU).
Allergic effector mechanisms
Epithelial support of immune responses
Following sensitization, epithelial cells again take center stage as critical effectors in allergic inflammation, coordinating an interplay with immune and neural cells. In response to allergen challenge, epithelial cells again release IL-25, IL-33, and TSLP that activate ILC2s, Ag-specific CD4+ T cells, and innate granulocytes such as eosinophils, mast cells, and basophils [8,10,16,40,41]. Epithelial-derived factors can also provide support for immunoglobulin E (IgE) and memory Th2 responses. For example, TSLP could program human DCs to drive emergence of T follicular helper cell responses in vitro that promoted IgE production by B cells [42]. Such support of IgE production is critical, as IgE binds to the high-affinity IgE receptor, FcεRIα, on mast cells and basophils, with subsequent allergen exposure and crosslinking prompting degranulation [43] that releases histamine, neurotrophic factors, bioactive lipids, Type 2 cytokines, and proteases that further propagate the immune response [3,8]. However, some allergic responses are largely IgE-independent, including EoE [16,44]. In IgE-dependent and -independent responses, epithelial-derived cytokines act on Th2 memory cells to drive IL-4 and IL-5 production, which contributes to the recruitment and activation of eosinophils [16,45]. TSLP has pleotropic effects on Th2 memory cells (reviewed in ref [8]) and can promote basophil activities in AD and EoE [8,16]. Thus, epithelial-derived cytokines are critical in promoting immune responses in the allergic cascade when allergen challenge occurs, in the GI tract and other tissues (Figure 2).
In addition to their role as cytokine-producers, epithelial cells, particularly tuft cells, are an emerging source of bioactive lipids called eicosanoids that can tune inflammation. Eicosanoids include the prostanoids, leukotrienes, lipoxins, and other species that derive from arachidonic acid and are synthesized via species-specific enzymatic synthesis cascades [46–48]. Cysteinyl leukotrienes (cysLTs) in particular have a pathogenic role in allergic airway inflammation [49]. Loss of the transcription factor FoxA2 in the airway epithelium drove increased cysLT production and allergic airway disease [50]. A recent study showed that in mice, tracheal tuft cells, termed brush cells, produced cysLTs, dependent upon the ATP sensor P2Y2, that drove ILC2 activation and allergic responses to Alternaria allergen [14,51]. Similarly, prostaglandin D2 (PGD2) could also promote allergic airway inflammation, synergizing with airway epithelium-derived IL-25 and IL-33 to activate ILC2s [52] and recruit them to the inflamed lung [19]. However, the role of tuft cell-derived eicosanoids in allergic immune responses remains unclear in many contexts; tuft cell-derived cysLTs contributed to intestinal Type 2 inflammation in helminth infection [53], and small intestinal tuft cells have been shown to produce PGD2 [54], but whether tuft cells across tissues are a key eicosanoid source that activate or recruit immune cells in allergy requires further study.
Epithelial cells as effectors in remodeled allergic tissues
Allergic disease involves drastic tissue remodeling mediated by epithelial changes driven by Type 2 cytokines [3,8] (Figure 1). In the GI tract, IL-13 acts on intestinal epithelial stem cells to skew differentiation towards tuft and goblet cells [8,33,54]. Single cell RNA-sequencing has shown that that IL-13 drove stem cells away from self-renewal and towards tuft and goblet cell gene signatures in mouse intestinal epithelial cells [55] or small intestinal organoid cultures [56]. Accumulation of tuft and goblet cells is a critical driver of intestinal Type 2 inflammation. Tuft cells produce IL-25, leukotrienes, and acetylcholine (Ach) that can alter endothelial, epithelial, and immune function and increase intestinal permeability to facilitate allergen translocation across the barrier [9,10,12–14,53,57]. Goblet cells produce mucus that elicits allergic pathology, and in naive wild-type mice, goblet cells formed goblet cell-associated antigen passages (GAPs) that allowed luminal antigens to access lamina propria immune cells [58], potentially contributing to sensitization and tolerance (Figures 1 and 2).
In the lung, IL-13-mediated epithelial remodeling in allergic asthma is particularly well-characterized. Allergic asthma features airway hyperresponsiveness, mucus hypersecretion, and remodeling characterized by changes in the composition of epithelial cells in the airways and epithelial cell apoptosis (smooth muscle cell proliferation and fibroblast activation also occur), contributing to airflow limitation and respiratory symptoms, reviewed in ref [21]. In this context, granulocytes and other immune cells release factors, such as IL-13, that trigger goblet cell mucus hyperproduction, and human eosinophils produced Charcot-Leyden crystals composed of galectin 10 that embedded in mucus and acted as a Type 2 adjuvant [59]. In addition, using human nasal polyp epithelial cells and murine models, tuft cells have been shown to be a source of PGE2 that prompted increased mucociliary transport function [60]. IL-13 and other cytokines, like IL-31, also drive epithelial remodeling and functional changes in the skin during allergy, supporting the development of pruritic acute or chronic lesions with distinct epithelial features and a breach in the skin barrier [61] that can predispose to other allergic diseases (a process termed “the allergic march,” reviewed in detail in ref [62]). The pruritis that leads to scratching and related pathology in AD speaks to the importance of the neuro-immune-epithelial link in the skin (discussed below). Thus, epithelial cells serve as end-point effectors that execute a suite of pro-inflammatory allergic responses in the GI tract (Figure 2), lung, and skin (Figure 1).
Importantly, tissue remodeling that occurs during allergy is not an acute event. Epithelial cells in allergy and other contexts “remember” past inflammatory responses, elevating baseline inflammation even in periods of reduced symptoms and increasing sensitivity to subsequent exposures. Epigenetic modifications in epithelial stem cells may play a critical role in these processes, altering gene expression in response to past exposures [63–66]. In the respiratory tract, airway epithelial basal stem cells from chronic rhinosinusitis patients generated less diverse progeny compared to controls, associated with distinct alterations in the chromatin landscape, and retained a Type 2-skewed signature and elevated Wnt pathway activity after exposure to Type 2 cytokines [66]. Whether and how epithelial inflammatory memory functions in the allergic GI tract and skin remains unknown, but a better understanding of the processes that underlie lasting changes in epithelial function and structure during allergy may offer novel avenues for intervention in chronic disease.
Neural-immune link
The rapid response to allergens is believed to be an evolutionary mechanism to expel toxins and establish avoidance behavior, suggesting a direct link between immune-epithelial function and physiology controlled by the nervous system – itch, sneezing, gut motility, and pain. Immune and epithelial cells express receptors for neural factors and secrete neurotropic factor/neurotransmitters. In turn, neurons express cytokine receptors, suggesting bi-directional communication [8,67,68]. In addition, neurons can directly sense a diverse array of enzymatically active allergens. Recent work has highlighted the critical role for this sensory mechanism in initiating allergic inflammation in the skin and DC migration via Substance P [69].
Perhaps the most well characterized neural-immune connection during allergy is between neurons and mast cells [8,68,70]. Broadly, murine and human mast cell-derived factors such as histamine, PGE2, and leukotriene C4 signal to neurons, and mast cells produce neurotrophic factors such as serotonin and nerve growth factor (NGF) that induce itch in the skin and increase gut motility. Mast cell degranulation is regulated by neurotransmitters including Substance P, calcitonin gene-related peptide (CGRP), Ach, and vasoactive intestine peptide [68]. In vivo, recent studies have shown that mast cells loaded with dietary-specific IgE antibodies led to meal-induced abdominal pain and subsequent avoidance behavior in mice [71,72]. In addition to this important role for mast cells, in the murine intestine, eosinophils responded to neuron-derived neuromedin U (NMU) to initiate goblet cell differentiation [73] (Figure 2), and NMU could also activate pro-allergic ILC2s from humans and mice to drive lung allergy [74–77]. Finally, a very recent study has shown that γδ T cells produced IL-3 that acted on sensory neurons to increase their sensitivity to the protease allergen papain and promote itch and allergic skin inflammation in mice [78].
There is also mounting evidence to support epithelial-neural connections during Type 2 responses [8,68]. In the GI tract, enteric neurons and epithelial tuft cells released Ach to stimulate mucus and epithelial fluid secretion in helminth-induced Type 2 inflammation [57], though whether this axis functions in intestinal allergy remains unclear. During allergic lung inflammation, epithelial-derived IL-25 synergized with NMU that activated ILC2s [77]. In addition, innervated pulmonary neuroendocrine cells, a rare epithelial cell type in the lung, produced CGRP and γ-aminobutyric acid that drove pulmonary ILC2 and goblet cell responses, respectively [79]. In AD, sensory neurons expressed receptors for Th2-derived IL-31 that promotes itch and epithelial cell-derived IL-33, TSLP, and NGF [80–82]. Also in the skin, neural-derived CGRP and Substance P drove Langerhans cells towards a Type 2 phenotype and induced keratinocyte release of tumor necrosis factor α (TNFα), IL-1b, and NGF [83,84]. Thus, evidence is emerging that neuro-epithelial functions are highly intertwined in allergic tissues. However, further studies are required to dissect the complex interplay of these interactions, particularly in humans, and whether they could be targeted therapeutically to treat allergy.
The environment shapes epithelial cell-dependent initiation and promotion of allergic disease
Environmental factors such as climate, pollution, and nutrition affect epithelial responses and are key contributors to allergic disease, driving pro-allergic pathways and, through climate change, prolonging exposure to seasonal allergens [1,85–88]. For example, exposing human airway epithelial cells to pollutant fine particulate matter resulted in inflammatory cytokine and mucin gene expression [89] and enhanced MUC5AC production, downstream of ER stress [90]. Diesel exhaust particles activated the aryl hydrocarbon receptor (AhR) on primary asthmatic human airway epithelial cells, associated with enhanced AhR binding to the IL25, IL33, and TSLP promoters [91]. In human skin, particulate matter exposure resulted in decreased filaggrin expression, dependent upon TNFa and the AhR, associated with epithelial barrier defects that could predispose to allergic sensitization [92]. While additional research is required, these studies suggest a key role for pollutants in shaping pro-allergic epithelial initiation and effector responses (Figure 3).
Figure 3. Environmental and nutritional factors mediating allergic disease.
The link between environmental factors and allergic disease has become apparent in recent years. Airborne pollutants such as particulate matter and greenhouse gases contribute to allergic disease exacerbation by altering the climate and promoting inflammatory epithelial responses. Additionally, the “Western lifestyle” can influence nutrition, limit beneficial breastfeeding behaviors, and alter commensal microbial communities, promoting the occurrence of allergy and exacerbating existing allergic responses. Levels of iron, zinc, vitamin D and other nutrients have been linked to inflammatory diseases like atopic dermatitis and asthma. The mechanisms by which environmental factors drive pro-allergic epithelial activities requires further study. House dust mite (HDM).
Shifting commensal microbial communities, in part attributable to a “Western” lifestyle that lacks microbial exposure and often features early life antibiotic use, also contribute to epithelial allergic inflammation. While this topic has been reviewed extensively elsewhere [87], a few examples related to the epithelium are notable. TLR4 on lung structural cells, most likely epithelial cells, was required for the HDM response in mice and promoted production of IL-25, IL-33, TSLP, and GM-CSF [24], though chronic exposure to farm dust or low-dose endotoxin protected mice against HDM-elicited allergic airway inflammation, dependent upon lung epithelial-intrinsic A20, a ubiquitin-modifying enzyme that regulates cytokine signaling [93]. Certain microbial species are associated with protection against allergy. Clostridia sp. colonization of germ-free mice elicited epithelial barrier-protective IL-22 that limited food allergy [94], and Anaerostipes caccae from the feces of healthy infants was protective against anaphylaxis in response to cow’s milk in colonized mice, associated with unique metabolic gene expression profiles in ileal epithelial cells [95]. Some species associated with allergy can promulgate allergic inflammation; monolayers of A549 airway epithelial cells exposed to Streptococcus salivarius from allergic rhinitis patients increased expression of IL33 and TSLP, and S. salivarius treatment exacerbated inflammation in a mouse model of rhinitis [96].
Gut microbes also release products such as short-chain fatty acids that tune allergic inflammation. The metabolites butyrate and propionate restored IL-4-, IL-13-, and HDM-elicited loss of barrier function in human airway epithelial cells [97]. In the skin, mice given butyrate had improved skin barrier integrity, decreased Il33 expression, and enhanced keratinocyte differentiation driven by increased mitochondrial fatty acid oxidation in a model of AD-like disease [98], and topical application of propionate in AD and AD-like disease in humans and mice led to improved symptoms and decreased IL-33 levels, with propionate inhibiting IL-33 production, dependent on increased AhR expression and/or function [99]. Together, these studies show that microbial signals can control epithelial initiation and effector responses that underlie allergy, providing exciting opportunities to modulate allergic disease using microbes or their products (Figure 3).
Finally, deficiencies in essential micronutrients and vitamins can be associated with allergy [88]. For example, zinc deficiency resulted in increased mucus-secreting goblet cells and caspase-3-driven epithelial apoptosis in murine allergic airway disease [100]. Vitamin D receptor levels were decreased in nasal epithelial cells from rhinosinusitis patients, leading to an increase in the epithelial-derived, inflammatory cytokine IL-36γ [101]. Similarly, individuals with EoE were more likely to have low vitamin D levels, correlated with an increased risk of sensitization through dysregulated epithelial barriers; in vitro, vitamin D-deprived esophageal epithelial cells in air-liquid interface cultures treated with IL-13 displayed exaggerated epithelial thickness and decreased expression of epithelial structural markers [102]. However, overall, while there are known associations between allergy and other nutritional deficiencies, including iron deficiency [103], we are just beginning to explore the role of nutrition in allergic disease. Mechanistic studies in murine models coupled with translational human studies will be required to fully dissect how nutrient status alters epithelial function in allergy (Figure 3).
Resolution of allergic responses
Immune cells and cytokines
Coordinated resolution of Type 2 inflammation is critical to restore tissue homeostasis, repair damage, and allow for responses to subsequent stimuli [104,105]. T regulatory cells (Tregs) play a key role in initiating tolerance to potential allergens (reviewed in refs [106,107]). During Type 2 responses, Tregs, as well as M2-like macrophages, produce anti-inflammatory cytokines such as transforming growth factor-β (TGF-β) and IL-10 that are critical in restraining ongoing responses [4,104–108]. Treg depletion in mice resulted in a loss of barrier integrity in the lung, with increased airway permeability and a loss of tight junction protein expression; these changes were partially reversed by anti-IL-4 treatment, suggesting a direct connection between the presence of Tregs and epithelial Type 2 inflammation [109]. More recently, the transcription factor RORα in Tregs was shown to drive expression of the death receptor DR3 that sequestered TNF ligand–related molecule 1, limiting ILC2 activation and downstream epithelial inflammation in allergic skin disease [110]. However, while Tregs clearly restrain the immune responses that promote allergy, the precise role that Tregs play in directly suppressing epithelial-intrinsic inflammation modules in the intestine (Figure 4), lung, and skin is still being studied [106,107].
Figure 4. Epithelial resolution pathways in allergic inflammation in the gastrointestinal tract.
Coordinated resolution of effector responses in Type 2 inflammation is vital for restoring tissue homeostasis and preventing chronic pathology. Recent work has highlighted a role for cytokines such as interleukin(IL)-10 and transforming growth factor-β (TGF-β) that can promote regeneration, derived from T regulatory cells (Tregs) and other immune cells; prostaglandins; and specialized pro-resolving mediators such as lipoxins, resolvins, protectins, and maresins in acting on and regulating epithelial responses. Prostaglandin D2 (PGD2) and PGE2 act directly on the epithelium, with PGD2 and its receptor chemoattractant receptor homologous molecule expressed by Th2 cells (CRTH2) counteracting the IL-13-driven inflammatory program in the epithelium. Damage-associated molecular pattern molecules (DAMPs), extracellular matrix (ECM).
Likewise, while the effects of anti-inflammatory cytokines such as IL-10 and TGF-β on the epithelium have been investigated in other contexts [4,108], less is known regarding their effects on the epithelium during allergy. Recent work has highlighted that culture of nasal epithelial cells with IL-10 or IL-10-producing ILC2s reversed grass pollen-induced loss of expression of tight junction protein zonula occludens-1 (and increased expression of IL-6 and IL-8, supporting barrier integrity [111]. In the lung, TGF-β plays roles in the epithelial-to-mesenchymal transition, mucin production, and cell survival, which can shape epithelial barrier integrity and contribute to allergy, reviewed in ref [112] (Figure 4). However, specific effects of TGF-β on the epithelium during the resolution of allergic inflammation across tissues remain to be fully described. Future studies that selectively delete IL-10 or TGF-β responsiveness in the epithelium in murine and human in vitro models of allergic inflammation will be required to determine how immunomodulatory cytokines act on the epithelium to resolve or suppress allergic inflammation.
Eicosanoids and lipid mediators
Eicosanoids can have potent pro-inflammatory effects, as described above, but they are pleiotropic factors and also have anti-inflammatory properties associated with Type 2 inflammation. However, their role in resolving allergic epithelial responses is only just emerging [46–48]. During intestinal helminth-elicited Type 2 inflammation, we have demonstrated that the eicosanoid PGD2, potentially derived from tuft cells, and its receptor chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) counteract IL-13-driven epithelial reprogramming, establishing an epithelial-intrinsic negative regulatory mechanism in intestinal Type 2 inflammation [54] (Figure 4). However, it remains unclear whether this circuit functions in allergy. In addition, PGD2 elicited a pro-remodeling response in CRTH2-expressing airway epithelial cells from human asthmatic donors and increased differentiation of goblet cells in air-liquid interface cultures from human samples, though decreased CRTH2 expression was observed in allergic compared to healthy epithelium in this context [113]. Thus, given the known pro-inflammatory functions of this axis on immune cells [19,52], how PGD2 and CRTH2 regulate epithelial responses in various allergic tissues must be studied further.
In addition to PGD2, other prostanoids can act on epithelial cells to resolve inflammation. In contact hypersensitivity, PGE2 suppressed skin inflammation via inhibition of keratinocyte activation, and the PGE2 receptor EP3 on conjunctival epithelium promoted resolution of murine experimental allergic conjunctivitis through inhibition of CXCL1 [114]. However, in human bronchial epithelial air-liquid interface cultures, PGE2 induced mucus secretion, potentially exacerbating asthma [115], demonstrating that the role of this prostanoid, like PGD2, can be context-dependent. Other lipid mediators such as lipoxins impact epithelial cell biology; for example, lipoxin B4 promoted resolution of allergic lung inflammation through inhibition of goblet cell hyperplasia [116], and in human airway epithelial cells, lipoxin A4 suppressed mucus secretion via inhibition of p38 and ERK phosphorylation [117]. Overall, the eicosanoid family is complex, with questions remaining regarding its function in vivo in humans, but as many eicosanoid pathways are highly druggable [46], an increased understanding of how eicosanoids promote resolution of inflammation may inform the development of new drugs to treat allergy (Figure 4).
Current therapeutics that target epithelial activities in allergy
As our understanding of the mechanisms that control Type 2 epithelial-driven inflammation has increased, novel monoclonal antibodies (mAbs) that block or neutralize epithelial-derived cytokines have been developed to treat allergic diseases [118]. Tezepelumab, an anti-TSLP mAb, was the first allergy treatment to directly target an epithelial-derived cytokine. Tezepelumab reduced asthma severity and is approved for use in moderate-severe asthma [119]. Therapies that target IL-25 and IL-33 are not available yet, though a number of anti-IL-33/IL-33R compounds are in clinical trials. Itepekimab (anti-IL-33) reduced asthma-related events in a clinical trial [120], and tozorakimab (anti-IL-33) blocked IL-33R-dependent effects in human primary cells [121]. Etokimab (anti-IL-33) was effective in improving symptoms of AD [122] and reduced reactivity to peanut challenge in allergic subjects [123]. Astegolimab (anti-IL-33R/ST2) was effective in reducing asthma exacerbation rates [124] but was not effective in AD [125], suggesting that targeting IL-33 to treat allergic disease must be context-specific. These studies highlight that understanding epithelial function can underpin successful efforts to treat allergic disease in the clinic.
Future work that unravels how other epithelial products contribute to allergy will hopefully lead to the development of new therapies that target a diverse array of epithelial functions. The eicosanoids represent a particularly attractive target [46–48]; the leukotriene inhibitor montelukast is already commonly used in the clinic to treat allergic asthma [126]. However, major unknowns related to epithelial cell-derived eicosanoids remain. For example, inhibition of the PGD2 receptor CRTH2 to treat allergic airway inflammation did not offer substantial benefit over placebo [127]. While this approach aimed to dampen CRTH2-dependent, pro-allergic immune responses, data showing that the PGD2-CRTH2 pathway suppressed Type 2 epithelial inflammation [54] suggests that a more complete understanding of the role of epithelial-derived eicosanoids is needed before we can successfully target these pathways therapeutically. However, developing therapies that enhance epithelial-intrinsic, pro-resolution pathways could be an attractive alternate route to treat allergic disease.
Conclusion
Epithelial barriers are central to the pathogenesis of allergic diseases, serving as critical interfaces for the initiation, propagation, and resolution of Type 2 inflammatory responses. Recent advances have highlighted how environmental factors, including pollutants, microbiota, and nutrient deficiencies, profoundly influence epithelial integrity and function in allergy, in a tissue-specific manner. Areas of particular interest in the field center around how these environmental factors and genetics intersect to mediate susceptibility to the development of allergic disease, how the neuro-immune-epithelial interplay influences allergic effector mechanisms, and how we can leverage our growing knowledge of the tissue-dependent mechanisms that control allergy to develop next-generation therapeutics to prevent and treat allergy. A deeper understanding of the epithelial-intrinsic mechanisms that promote allergy will be pivotal in developing targeted strategies to mitigate allergic disease burden and improve patient outcomes.
Acknowledgements
The authors thank members of the Tait Wojno laboratory. This work was supported by the NIH NIDDK (R01 AI1853796) to E.D.T.W. and University of Washington start-up funds to E.D.T.W. The content of this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Figures created with Biorender.com.
Abbreviations
- Ach
acetylcholine
- AD
atopic dermatitis
- AhR
aryl hydrocarbon receptor
- CGRP
calcitonin gene-related peptide
- CRTH2
chemoattractant receptor-homologous molecule expressed on Th2 cells
- cysLT
cysteinyl leukotriene
- DC
dendritic cell
- EoE
eosinophilic esophagitis
- GAP
goblet cell-associated antigen passages
- GM-CSF
granulocyte-monocyte colony-stimulating factor
- GI
gastrointestinal
- HDM
house dust mite
- IgE
immunoglobulin E
- IL
interleukin
- ILC2
group 2 innate lymphoid cells
- mAb
monoclonal antibody
- MHC-II
major Histocompatibility Complex II
- NGF
nerve growth factor
- NMU
neuromedin U
- PG
prostaglandin
- SCFA
short chain fatty acid
- TGF-β
transforming growth factor-β
- Th2 - CD4+
T helper Type 2
- TLR4
Toll-like receptor 4
- TNFα
tumor necrosis factor α
- TSLP
thymic stromal lymphopoietin
Footnotes
Declaration of Competing Interest
The authors have no interests to declare.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.AAAAI: Allergy Statistics. Edited by. Milwaukee, WI; 2024. vol 2024. [Google Scholar]
- 2.Molofsky AB, Locksley RM: The ins and outs of innate and adaptive type 2 immunity. Immunity 2023, 56:704–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hammad H, Debeuf N, Aegerter H, Brown AS, Lambrecht BN: Emerging Paradigms in Type 2 Immunity. Annu Rev Immunol 2022, 40:443–467. [DOI] [PubMed] [Google Scholar]
- 4.Branchett WJ, Lloyd CM: Regulatory cytokine function in the respiratory tract. Mucosal Immunol 2019, 12:589–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kopp EB, Agaronyan K, Licona-Limón I, Nish SA, Medzhitov R: Modes of type 2 immune response initiation. Immunity 2023, 56:687–694. [DOI] [PubMed] [Google Scholar]
- 6.Kim BS, Wojno ED, Artis D: Innate lymphoid cells and allergic inflammation. Curr Opin Immunol 2013, 25:738–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mukai K, Tsai M, Starkl P, Marichal T, Galli SJ: IgE and mast cells in host defense against parasites and venoms. Semin Immunopathol 2016, 38:581–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stanbery AG, Shuchi Smita, Jakob von Moltke, Tait Wojno ED, Ziegler SF: TSLP, IL-33, and IL-25: Not just for allergy and helminth infection. J Allergy Clin Immunol 2022, 150:1302–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kotas ME, O’Leary CE, Locksley RM: Tuft Cells: Context- and Tissue-Specific Programming for a Conserved Cell Lineage. Annu Rev Pathol 2023, 18:311–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jakob Von Moltke and Ming Ji and Hong Erh Liang and Richard ML: Tuft-cell-derived IL-25 regulates an intestinal ILC2–epithelial response circuit. Nature 2016, 529:221–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Leyva-Castillo JM, Galand C, Kam C, Burton O, Gurish M, Musser MA, Goldsmith JD, Hait E, Nurko S, Brombacher F, et al. : Mechanical Skin Injury Promotes Food Anaphylaxis by Driving Intestinal Mast Cell Expansion. Immunity 2019, 50:1262–1275.e1264. *This study revealed a new connection between allergy in the skin and GI tract. After skin tape-stripping in mice, keratinocytes produced IL-33 and intestinal tuft cells made IL-25 that activated IL-4-producing ILC2s, which in turn expanded small intestinal mast cells that cause anaphylaxis.*
- 12.Gerbe F, Sidot E, Smyth DJ, Ohmoto M, Matsumoto I, Dardalhon V, Cesses P, Garnier L, Pouzolles M, Brulin B, et al. : Intestinal epithelial tuft cells regulate type 2 mucosal responses required for expulsion of helminth parasites. Nature 2016, 529:226–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Howitt MR, Lavoie S, Michaud M, Blum AM, Tran SV, Weinstock JV, Gallini CA, Redding K, Margolskee RF, Osborne LC, et al. : Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 2016, 351:1329–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ualiyeva S, Lemire E, Aviles EC, Wong C, Boyd AA, Lai J, Liu T, Matsumoto I, Barrett NA, Boyce JA, et al. : Tuft cell-produced cysteinyl leukotrienes and IL-25 synergistically initiate lung type 2 inflammation. Sci Immunol 2021, 6:eabj0474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Leyva-Castillo JM, Galand C, Mashiko S, Bissonnette R, McGurk A, Ziegler SF, Dong C, McKenzie ANJ, Sarfati M, Geha RS: ILC2 activation by keratinocyte-derived IL-25 drives IL-13 production at sites of allergic skin inflammation. J Allergy Clin Immunol 2020, 145:1606–1614.e1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Noti M, Wojno ED, Kim BS, Siracusa MC, Giacomin PR, Nair MG, Benitez AJ, Ruymann KR, Muir AB, Hill DA, et al. : Thymic stromal lymphopoietin-elicited basophil responses promote eosinophilic esophagitis. Nat Med 2013, 19:1005–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Han H, Roan F, Johnston LK, Smith DE, Bryce PJ, Ziegler SF: IL-33 promotes gastrointestinal allergy in a TSLP-independent manner. Mucosal Immunol 2018, 11:394–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Cha J, Kim TG, Bhae E, Gwak HJ, Ju Y, Choe YH, Jang IH, Jung Y, Moon S, Kim T, et al. : Skin microbe-dependent TSLP-ILC2 priming axis in early life is co-opted in allergic inflammation. Cell Host Microbe 2024, 32:244–260.e211. *This work established a role for early-life skin colonization with particular microbes in the development of AD during adulthood. Colonization with S. lentus in young mice elicited keratinocyte TSLP production that established an ILC2 population in the skin that drove AD-like disease in adult mice.*
- 19.Oyesola OO, Duque C, Huang LC, Larson EM, Früh SP, Webb LM, Peng SA, Tait Wojno ED: The Prostaglandin D2 Receptor CRTH2 Promotes IL-33–Induced ILC2 Accumulation in the Lung. The Journal of Immunology 2020, 204:1001–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cayrol C, Girard JP: Interleukin-33 (IL-33): A critical review of its biology and the mechanisms involved in its release as a potent extracellular cytokine. Cytokine 2022, 156:155891. [DOI] [PubMed] [Google Scholar]
- 21.Hammad H, Lambrecht BN: The basic immunology of asthma. Cell 2021, 184:1469–1485. [DOI] [PubMed] [Google Scholar]
- 22.Soh WT, Zhang J, Hollenberg MD, Vliagoftis H, Rothenberg ME, Sokol CL, Robinson C, Jacquet A: Protease allergens as initiators-regulators of allergic inflammation. Allergy 2023, 78:1148–1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Millien VO, Lu W, Shaw J, Yuan X, Mak G, Roberts L, Song LZ, Knight JM, Creighton CJ, Luong A, et al. : Cleavage of fibrinogen by proteinases elicits allergic responses through Toll-like receptor 4. Science 2013, 341:792–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN: House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med 2009, 15:410–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cayrol C, Duval A, Schmitt P, Roga S, Camus M, Stella A, Burlet-Schiltz O, Gonzalez-de-Peredo A, Girard JP: Environmental allergens induce allergic inflammation through proteolytic maturation of IL-33. Nat Immunol 2018, 19:375–385. [DOI] [PubMed] [Google Scholar]
- 26.Moon HG, Kim SJ, Jeong JJ, Han SS, Jarjour NN, Lee H, Abboud-Werner SL, Chung S, Choi HS, Natarajan V, et al. : Airway Epithelial Cell-Derived Colony Stimulating Factor-1 Promotes Allergen Sensitization. Immunity 2018, 49:275–287.e275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Willart MA, Deswarte K, Pouliot P, Braun H, Beyaert R, Lambrecht BN, Hammad H: Interleukin-1α controls allergic sensitization to inhaled house dust mite via the epithelial release of GM-CSF and IL-33. J Exp Med 2012, 209:1505–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kabata H, Flamar AL, Mahlakõiv T, Moriyama S, Rodewald HR, Ziegler SF, Artis D: Targeted deletion of the TSLP receptor reveals cellular mechanisms that promote type 2 airway inflammation. Mucosal Immunol 2020, 13:626–636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Hammad H, Plantinga M, Deswarte K, Pouliot P, Willart MA, Kool M, Muskens F, Lambrecht BN: Inflammatory dendritic cells--not basophils--are necessary and sufficient for induction of Th2 immunity to inhaled house dust mite allergen. J Exp Med 2010, 207:2097–2111. *This landmark study identified the critical antigen presenting cells that drive Type 2 allergic inflammation in the lung. Dendritic cells were necessary and suffiicient to prime Th2 cells in a murine model of HDM-elicited allergic airway inflammation.*
- 30.Plantinga M, Guilliams M, Vanheerswynghels M, Deswarte K, Branco-Madeira F, Toussaint W, Vanhoutte L, Neyt K, Killeen N, Malissen B, et al. : Conventional and monocyte-derived CD11b(+) dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 2013, 38:322–335. [DOI] [PubMed] [Google Scholar]
- 31.Ito T, Wang YH, Duramad O, Hori T, Delespesse GJ, Watanabe N, Qin FX, Yao Z, Cao W, Liu YJ: TSLP-activated dendritic cells induce an inflammatory T helper type 2 cell response through OX40 ligand. J Exp Med 2005, 202:1213–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Pastore S, Fanales-Belasio E, Albanesi C, Chinni LM, Giannetti A, Girolomoni G: Granulocyte macrophage colony-stimulating factor is overproduced by keratinocytes in atopic dermatitis. Implications for sustained dendritic cell activation in the skin. J Clin Invest 1997, 99:3009–3017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Coakley G, Harris NL: The Intestinal Epithelium at the Forefront of Host–Helminth Interactions. In Trends in Parasitology 2020, 36:761–772. [DOI] [PubMed] [Google Scholar]
- 34.Heuberger CE, Janney A, Ilott N, Bertocchi A, Pott S, Gu Y, Pohin M, Friedrich M, Mann EH, Pearson C, et al. : MHC class II antigen presentation by intestinal epithelial cells fine-tunes bacteria-reactive CD4 T cell responses. Mucosal Immunol 2023, 3:416–430. [DOI] [PubMed] [Google Scholar]
- 35.Koyama M, Mukhopadhyay P, Schuster IS, Henden AS, Hülsdünker J, Varelias A, Vetizou M, Kuns RD, Robb RJ, Zhang P, et al. : MHC Class II Antigen Presentation by the Intestinal Epithelium Initiates Graft-versus-Host Disease and Is Influenced by the Microbiota. Immunity 2019, 51:885–898.e887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Beyaz S, Chung C, Mou H, Bauer-Rowe KE, Xifaras ME, Ergin I, Dohnalova L, Biton M, Shekhar K, Eskiocak O, et al. : Dietary suppression of MHC class II expression in intestinal epithelial cells enhances intestinal tumorigenesis. Cell Stem Cell 2021, 28:1922–1935.e1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shenoy AT, Lyon De Ana C, Arafa EI, Salwig I, Barker KA, Korkmaz FT, Ramanujan A, Etesami NS, Soucy AM, Martin IMC, et al. : Antigen presentation by lung epithelial cells directs CD4. Nat Commun 2021, 12:5834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mulder DJ, Pooni A, Mak N, Hurlbut DJ, Basta S, Justinich CJ: Antigen presentation and MHC class II expression by human esophageal epithelial cells: role in eosinophilic esophagitis. Am J Pathol 2011, 178:744–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Arebro J, Tengroth L, Razavi R, Kumlien Georén S, Winqvist O, Cardell LO: Antigen-presenting epithelial cells can play a pivotal role in airway allergy. J Allergy Clin Immunol 2016, 137:957–960.e957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gurram RK, Wei D, Yu Q, Butcher MJ, Chen X, Cui K, Hu G, Zheng M, Zhu X, Oh J, et al. : Crosstalk between ILC2s and Th2 cells varies among mouse models. Cell Rep 2023, 42:112073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Schuijs MJ, Brenis Gomez CM, Bick F, Van Moorleghem J, Vanheerswynghels M, van Loo G, Beyaert R, Voehringer D, Locksley RM, Hammad H, et al. : Interleukin-33-activated basophils promote asthma by regulating Th2 cell entry into lung tissue. J Exp Med 2024, 221:e20240103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pattarini L, Trichot C, Bogiatzi S, Grandclaudon M, Meller S, Keuylian Z, Durand M, Volpe E, Madonna S, Cavani A, et al. : TSLP-activated dendritic cells induce human T follicular helper cell differentiation through OX40-ligand. J Exp Med 2017, 214:1529–1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Velez TE, Bryce PJ, Hulse KE: Mast Cell Interactions and Crosstalk in Regulating Allergic Inflammation. Curr Allergy Asthma Rep 2018, 18:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Simon D, Cianferoni A, Spergel JM, Aceves S, Holbreich M, Venter C, Rothenberg ME, Terreehorst I, Muraro A, Lucendo AJ, et al. : Eosinophilic esophagitis is characterized by a non-IgE-mediated food hypersensitivity. Allergy 2016, 71:611–620. [DOI] [PubMed] [Google Scholar]
- 45.Morimoto Y, Hirahara K, Kiuchi M, Wada T, Ichikawa T, Kanno T, Okano M, Kokubo K, Onodera A, Sakurai D, et al. : Amphiregulin-Producing Pathogenic Memory T Helper 2 Cells Instruct Eosinophils to Secrete Osteopontin and Facilitate Airway Fibrosis. Immunity 2018, 49:134–150.e136. [DOI] [PubMed] [Google Scholar]
- 46.Oyesola OO, Tait Wojno ED: Prostaglandin regulation of type 2 inflammation: From basic biology to therapeutic interventions. Eur J Immunol 2021, 51:2399–2416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dennis EA, Norris PC: Eicosanoid storm in infection and inflammation. Nat Rev Immunol 2015, 15:511–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sokolowska M, Rovati GE, Diamant Z, Untersmayr E, Schwarze J, Lukasik Z, Sava F, Angelina A, Palomares O, Akdis CA, et al. : Current perspective on eicosanoids in asthma and allergic diseases: EAACI Task Force consensus report, part I. Allergy 2021, 76:114–130. [DOI] [PubMed] [Google Scholar]
- 49.Peters-Golden M, Henderson WR: Leukotrienes. N Engl J Med 2007, 357:1841–1854. [DOI] [PubMed] [Google Scholar]
- 50.Tang X, Liu XJ, Tian C, Su Q, Lei Y, Wu Q, He Y, Whitsett JA, Luo F: Foxa2 regulates leukotrienes to inhibit Th2-mediated pulmonary inflammation. Am J Respir Cell Mol Biol 2013, 49:960–970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ualiyeva S, Hallen N, Kanaoka Y, Ledderose C, Matsumoto I, Junger WG, Barrett NA, Bankova LG: Airway brush cells generate cysteinyl leukotrienes through the ATP sensor P2Y2. Sci Immunol 2020, 5:eaax7224. *This work established a key role for airway tuft cells in producing cysLTs during allergic airway inflammation. Murine airway tuft cells, compared to other airway epithelial cells, preferentially produced cysLTs in response to ionophore, dependent upon expression of the purinergic receptor P2Y2 that detects the cellular damage signal ATP.
- 52.Barnig C, Cernadas M, Dutile S, Liu X, Perrella MA, Kazani S, Wechsler ME, Israel E, Levy BD: Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma. Sci Transl Med 2013, 5:174ra126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.McGinty JW, Ting HA, Billipp TE, Nadjsombati MS, Khan DM, Barrett NA, Liang HE, Matsumoto I, von Moltke J: Tuft-Cell-Derived Leukotrienes Drive Rapid Anti-helminth Immunity in the Small Intestine but Are Dispensable for Anti-protist Immunity. Immunity 2020, 52:528–541.e527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Oyesola OO, Shanahan MT, Kanke M, Mooney BM, Webb LM, Smita S, Matheson MK, Campioli P, Pham D, Früh SP, et al. : PGD2 and CRTH2 counteract Type 2 cytokine–elicited intestinal epithelial responses during helminth infection. The Journal of Experimental Medicine 2021, 218:e20202178. *This study identified a novel mechanism by which tuft cell-derived prostaglandin restrained Type 2 inflammation in the intestinal epithelium. Murine small intestinal tuft cells produced PGD2 that counteracted IL-13-induced inflammation in IECs that expressed the PGD2 receptor CRTH2 during helminth infection.*
- 55.Biton M, Haber AL, Rogel N, Burgin G, Beyaz S, Schnell A, Ashenberg O, Su CW, Smillie C, Shekhar K, et al. : T Helper Cell Cytokines Modulate Intestinal Stem Cell Renewal and Differentiation. Cell 2018, 175:1307–1320.e1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lindholm HT, Parmar N, Drurey C, Campillo Poveda M, Vornewald PM, Ostrop J, Díez-Sanchez A, Maizels RM, Oudhoff MJ: BMP signaling in the intestinal epithelium drives a critical feedback loop to restrain IL-13-driven tuft cell hyperplasia. Sci Immunol 2022, 7:eabl6543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, et al. : Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024, 57:1243–1259.e8. *This hallmark study provided a direct connection between intestinal epithelial tuft cells and regulation of epithelial fluid secretion in Type 2 inflammation. Intestinal tuft cell-derived Ach acted on neighboring IECs to prompt chloride secretion that was critical in expeling worms during intestinal helminth infection.*
- 58.Noah TK, Knoop KA, McDonald KG, Gustafsson JK, Waggoner L, Vanoni S, Batie M, Arora K, Naren AP, Wang YH, et al. : IL-13-induced intestinal secretory epithelial cell antigen passages are required for IgE-mediated food-induced anaphylaxis. J Allergy Clin Immunol 2019, 144:1058–1073.e1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Persson EK, Verstraete K, Heyndrickx I, Gevaert E, Aegerter H, Percier JM, Deswarte K, Verschueren KHG, Dansercoer A, Gras D, et al. : Protein crystallization promotes type 2 immunity and is reversible by antibody treatment. Science 2019, 364:eaaw4295. *This work identified a novel mechanism by which protein crystals embed in epithelial-derived mucus to cause pathology associated with allergic airway disease, in humans and mice. Charcot-Leyden crystals composed of galectin-10 from human eosinophils acted as a Type 2 adjuvant, embedding in mucus to drive Type 2 inflammation in humanized mice.*
- 60.Kotas ME, Moore CM, Gurrola JG, Pletcher SD, Goldberg AN, Alvarez R, Yamato S, Bratcher PE, Shaughnessy CA, Zeitlin PL, et al. : IL-13-programmed airway tuft cells produce PGE2, which promotes CFTR-dependent mucociliary function. JCI Insight 2022, 7:e159832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Leung DY: Atopic dermatitis: the skin as a window into the pathogenesis of chronic allergic diseases. J Allergy Clin Immunol 1995, 96:302–318; quiz 319. [DOI] [PubMed] [Google Scholar]
- 62.Paller AS, Spergel JM, Mina-Osorio P, Irvine AD: The atopic march and atopic multimorbidity: Many trajectories, many pathways. J Allergy Clin Immunol 2019, 143:46–55. [DOI] [PubMed] [Google Scholar]
- 63.Ostuni R, Piccolo V, Barozzi I, Polletti S, Termanini A, Bonifacio S, Curina A, Prosperini E, Ghisletti S, Natoli G: Latent enhancers activated by stimulation in differentiated cells. Cell 2013, 152:157–171. [DOI] [PubMed] [Google Scholar]
- 64.Naik S, Larsen SB, Gomez NC, Alaverdyan K, Sendoel A, Yuan S, Polak L, Kulukian A, Chai S, Fuchs E: Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 2017, 550:475–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jakwerth CA, Ordovas-Montanes J, Blank S, Schmidt-Weber CB, Zissler UM: Role of Respiratory Epithelial Cells in Allergic Diseases. Cells 2022, 11:1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Ordovas-Montanes J, Dwyer DF, Nyquist SK, Buchheit KM, Vukovic M, Deb C, Wadsworth MH, Hughes TK, Kazer SW, Yoshimoto E, et al. : Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature 2018, 560:649–654. *This study was the first to identify a “trained” inflammatory response in the respriatory epithelium during allergic airway disease that may contribute to chronicity. Human respiratory epithelial stem cells from chronic rhinosinusitis patients retained a skewed Type 2 inflammatory gene signature, epigenetic alterations, and a persistent inability to produce diverse progeny after IL-4/IL-13 stimulation.*
- 67.Emanuel E, Arifuzzaman M, Artis D: Epithelial-neuronal-immune cell interactions: Implications for immunity, inflammation, and tissue homeostasis at mucosal sites. J Allergy Clin Immunol 2024, 153:1169–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Voisin T, Bouvier A, Chiu IM: Neuro-immune interactions in allergic diseases: novel targets for therapeutics. Int Immunol 2017, 29:247–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Perner C, Flayer CH, Zhu X, Aderhold PA, Dewan ZNA, Voisin T, Camire RB, Chow OA, Chiu IM, Sokol CL: Substance P Release by Sensory Neurons Triggers Dendritic Cell Migration and Initiates the Type-2 Immune Response to Allergens. Immunity 2020, 53:1063–1077.e1067. *This work provided a novel mechanism by which a sensory neuron-derived factor communicated with innate immune cells to drive allergy in the skin. Sensory neurons directly sensed allergen and produced Substance P that elicited DC migration to promote skin allergy.*
- 70.Plum T, Binzberger R, Thiele R, Shang F, Postrach D, Fung C, Fortea M, Stakenborg N, Wang Z, Tappe-Theodor A, et al. : Mast cells link immune sensing to antigen-avoidance behaviour. Nature 2023, 620:634–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Aguilera-Lizarraga J, Florens MV, Viola MF, Jain P, Decraecker L, Appeltans I, Cuende-Estevez M, Fabre N, Van Beek K, Perna E, et al. : Local immune response to food antigens drives meal-induced abdominal pain. Nature 2021, 590:151–156. *This hallmark study provided the first mechanistic link between immune responses to food and mechanisms that cause food avoidance, highlighting that the allergic response may have evolved to protect from exposure to potentially dangerous foods. Murine mast cells loaded with dietary-specific IgE antibodies released histamine that sensitized visceral afferent nerves to cause meal-induced abdominal pain.*
- 72.Florsheim EB, Bachtel ND, Cullen JL, Lima BGC, Godazgar M, Carvalho F, Chatain CP, Zimmer MR, Zhang C, Gautier G, et al. : Immune sensing of food allergens promotes avoidance behaviour. Nature 2023, 620:643–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Y, Liu S, Zhou K, Wang Y, Chen Y, Hu W, Li S, Li H, Wang Q, He D, et al. : Neuromedin U programs eosinophils to promote mucosal immunity of the small intestine. Science 2023, 381:1189–1196. [DOI] [PubMed] [Google Scholar]
- 74.Ye Y, Luo J, Zeng N, Jiang S, Chen W, Hoyle RD, Klenerman P, Pavord ID, Xue L: Neuromedin U promotes human type 2 immune responses. Mucosal Immunol 2022, 15:990–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Jarick KJ, Topczewska PM, Jakob MO, Yano H, Arifuzzaman M, Gao X, Boulekou S, Stokic-Trtica V, Leclère PS, Preußer A, et al. : Non-redundant functions of group 2 innate lymphoid cells. Nature 2022, 611:794–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ju X, Nagashima A, Dvorkin-Gheva A, Wattie J, Howie K, Whetstone C, Ranjbar M, Cusack R, Ditta R, Paré G, et al. : Neuromedin-U Mediates Rapid Activation of Airway Group 2 Innate Lymphoid Cells in Mild Asthma. Am J Respir Crit Care Med 2024, 210:755–765. [DOI] [PubMed] [Google Scholar]
- 77.Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour RE, Nyman J, Dionne D, Hofree M, Cuoco MS, Rodman C, Farouq D, et al. : The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 2017, 549:351–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Flayer CH, Kernin IJ, Matatia PR, Zeng X, Yarmolinsky DA, Han C, Naik PR, Buttaci DR, Aderhold PA, Camire RB, et al. : A γδ T cell-IL-3 axis controls allergic responses through sensory neurons. Nature 2024, 634:440–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Sui P, Wiesner DL, Xu J, Zhang Y, Lee J, Van Dyken S, Lashua A, Yu C, Klein BS, Locksley RM, et al. : Pulmonary neuroendocrine cells amplify allergic asthma responses. Science 2018, 360:eaan8546. *This work linked innervated respiratory epithelial function with immune and epithelial responses to drive allergic airway inflammation. In mice, innervated pulmonary neuroendocrine cells in the lung released CGRP and γ-aminobutyric acid, which activated pulmonary ILC2 and goblet cell responses, respectively, to support allergic pathology in the lung*
- 80.Trier AM, Mack MR, Fredman A, Tamari M, Ver Heul AM, Zhao Y, Guo CJ, Avraham O, Ford ZK, Oetjen LK, et al. : IL-33 signaling in sensory neurons promotes dry skin itch. J Allergy Clin Immunol 2022, 149:1473–1480.e1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Cevikbas F, Wang X, Akiyama T, Kempkes C, Savinko T, Antal A, Kukova G, Buhl T, Ikoma A, Buddenkotte J, et al. : A sensory neuron-expressed IL-31 receptor mediates T helper cell-dependent itch: Involvement of TRPV1 and TRPA1. J Allergy Clin Immunol 2014, 133:448–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Meng J, Li Y, Fischer MJM, Steinhoff M, Chen W, Wang J: Th2 Modulation of Transient Receptor Potential Channels: An Unmet Therapeutic Intervention for Atopic Dermatitis. Front Immunol 2021, 12:696784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Shi X, Wang L, Clark JD, Kingery WS: Keratinocytes express cytokines and nerve growth factor in response to neuropeptide activation of the ERK1/2 and JNK MAPK transcription pathways. Regul Pept 2013, 186:92–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ding W, Stohl LL, Wagner JA, Granstein RD: Calcitonin gene-related peptide biases Langerhans cells toward Th2-type immunity. J Immunol 2008, 181:6020–6026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Faraz K, Seely M, Marano AL: The role of the environment in allergic skin disease. Curr Allergy Asthma Rep 2024, 24:323–330. [DOI] [PubMed] [Google Scholar]
- 86.Kim BE, Hui-Beckman JW, Nevid MZ, Goleva E, Leung DYM: Air pollutants contribute to epithelial barrier dysfunction and allergic diseases. Ann Allergy Asthma Immunol 2024, 132:433–439. [DOI] [PubMed] [Google Scholar]
- 87.Iweala OI, Nagler CR: The Microbiome and Food Allergy. Annu Rev Immunol 2019, 37:377–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhang P: The Role of Diet and Nutrition in Allergic Diseases. Nutrients 2023, 15:3583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Grytting VS, Skuland T, Ballangby J, Refsnes M, Låg M, Øvrevik J, Mariussen E: The effects of fine particulate matter (SRM 2786) on three different 3D lung models exposed at the air-liquid interface - A comparative study. Toxicol In Vitro 2024, 98:105841. [DOI] [PubMed] [Google Scholar]
- 90.Hu L, Xu C, Tang X, Yu S, Wang L, Li Q, Zhou X: Fine particulate matter promotes airway inflammation and mucin production by activating endoplasmic reticulum stress and the IRE1α/NOD1/NF‑κB pathway. Int J Mol Med 2023, 52:96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Weng CM, Wang CH, Lee MJ, He JR, Huang HY, Chao MW, Chung KF, Kuo HP: Aryl hydrocarbon receptor activation by diesel exhaust particles mediates epithelium-derived cytokines expression in severe allergic asthma. Allergy 2018, 73:2192–2204. [DOI] [PubMed] [Google Scholar]
- 92.Kim BE, Kim J, Goleva E, Berdyshev E, Lee J, Vang KA, Lee UH, Han S, Leung S, Hall CF, et al. : Particulate matter causes skin barrier dysfunction. JCI Insight 2021, 6:e145185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Schuijs MJ, Willart MA, Vergote K, Gras D, Deswarte K, Ege MJ, Madeira FB, Beyaert R, van Loo G, Bracher F, et al. : Farm dust and endotoxin protect against allergy through A20 induction in lung epithelial cells. Science 2015, 349:1106–1110. [DOI] [PubMed] [Google Scholar]
- 94.Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, Tjota MY, Seo GY, Cao S, Theriault BR, et al. : Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A 2014, 111:13145–13150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Feehley T, Plunkett CH, Bao R, Choi Hong SM, Culleen E, Belda-Ferre P, Campbell E, Aitoro R, Nocerino R, Paparo L, et al. : Healthy infants harbor intestinal bacteria that protect against food allergy. Nat Med 2019, 25:448–453. *This work identified a novel link between a commenseal bacterium and protection against food allergy. Using a mouse model of cow’s milk allergy, colonization of mice with Anaerostipes caccae from the feces of healthy infants protected against milk-induced anaphylaxis.*
- 96.Miao P, Jiang Y, Jian Y, Shi J, Liu Y, Piewngam P, Zheng Y, Cheung GYC, Liu Q, Otto M, et al. : Exacerbation of allergic rhinitis by the commensal bacterium Streptococcus salivarius. Nat Microbiol 2023, 8:218–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Richards LB, Li M, Folkerts G, Henricks PAJ, Garssen J, van Esch BCAM: Butyrate and Propionate Restore the Cytokine and House Dust Mite Compromised Barrier Function of Human Bronchial Airway Epithelial Cells. Int J Mol Sci 2020, 22:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Trompette A, Pernot J, Perdijk O, Alqahtani RAA, Domingo JS, Camacho-Muñoz D, Wong NC, Kendall AC, Wiederkehr A, Nicod LP, et al. : Gut-derived short-chain fatty acids modulate skin barrier integrity by promoting keratinocyte metabolism and differentiation. Mucosal Immunol 2022, 15:908–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Qiu Z, Zhu Z, Liu X, Chen B, Yin H, Gu C, Fang X, Zhu R, Yu T, Mi W, et al. : A dysregulated sebum-microbial metabolite-IL-33 axis initiates skin inflammation in atopic dermatitis. J Exp Med 2022, 219:e20212397. *This study highlighted a novel mechanism by which commensal bacteria of the skin and their metabolites can protect against AD-like disease. In a mouse model, the sebum metabolite proprionate limited skin epithelial IL-33 production and controlled AD-like disease.*
- 100.Truong-Tran AQ, Ruffin RE, Foster PS, Koskinen AM, Coyle P, Philcox JC, Rofe AM, Zalewski PD: Altered zinc homeostasis and caspase-3 activity in murine allergic airway inflammation. Am J Respir Cell Mol Biol 2002, 27:286–296. [DOI] [PubMed] [Google Scholar]
- 101.Xiao Q, Wang H, Song J, Qin ZY, Pan L, Liao B, Deng YK, Ma J, Liu JX, Hu J, et al. : Impaired local Vitamin D3 metabolism contributes to IL-36g overproduction in epithelial cells in chronic rhinosinusitis with nasal polyps. Rhinology 2024, 62:236–249. [DOI] [PubMed] [Google Scholar]
- 102.Brusilovsky M, Rochman M, Shoda T, Kotliar M, Caldwell JM, Mack LE, Besse JA, Chen X, Weirauch MT, Barski A, et al. : Vitamin D receptor and STAT6 interactome governs oesophageal epithelial barrier responses to IL-13 signalling. Gut 2023, 72:834–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Peroni DG, Hufnagl K, Comberiati P, Roth-Walter F: Lack of iron, zinc, and vitamins as a contributor to the etiology of atopic diseases. Front Nutr 2022, 9:1032481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Onali S, Favale A, Fantini MC: The Resolution of Intestinal Inflammation: The Peace-Keeper’s Perspective. Cells 2019, 8:344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gieseck RL, Wilson MS, Wynn TA: Type 2 immunity in tissue repair and fibrosis. Nat Rev Immunol 2018, 18:62–76. [DOI] [PubMed] [Google Scholar]
- 106.Martinez-Blanco M, Mukhatayev Z, Chatila TA: Pathogenic mechanisms in the evolution of food allergy. Immunol Rev 2024, 326:219–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Martín-Orozco E, Norte-Muñoz M, Martínez-García J: Regulatory T Cells in Allergy and Asthma. Front Pediatr 2017, 5:117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Maizels RM: The multi-faceted roles of TGF-β in regulation of immunity to infection. Adv Immunol 2021, 150:1–42. [DOI] [PubMed] [Google Scholar]
- 109.Jonckheere AC, Steelant B, Seys SF, Cremer J, Dilissen E, Boon L, Liston A, Schrijvers R, Breynaert C, Vanoirbeek JAJ, et al. : Peribronchial Inflammation Resulting from Regulatory T Cell Deficiency Damages the Respiratory Epithelium and Disturbs Barrier Function. J Immunol 2022, 209:1595–1605. [DOI] [PubMed] [Google Scholar]
- 110.Malhotra N, Leyva-Castillo JM, Jadhav U, Barreiro O, Kam C, O’Neill NK, Meylan F, Chambon P, von Andrian UH, Siegel RM, et al. : RORα-expressing T regulatory cells restrain allergic skin inflammation. Sci Immunol 2018, 3:eaao6923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Golebski K, Layhadi JA, Sahiner U, Steveling-Klein EH, Lenormand MM, Li RCY, Bal SM, Heesters BA, Vilà-Nadal G, Hunewald O, et al. : Induction of IL-10-producing type 2 innate lymphoid cells by allergen immunotherapy is associated with clinical response. Immunity 2021, 54:291–307.e297. *This work identified a novel population of suppressive ILC2s that acted directly to limit epithelial Type 2 inflammation. IL-10-producing ILC2s increased tight junction protein expression and barrier integrity in human nasal epithelial cells exposed to grass pollen, and sublingual immunotherapy that decreased grass pollen allergy symptoms in patients was associated with increased IL-10-producing ILC2s.*
- 112.Kraik K, Tota M, Laska J, Łacwik J, Paździerz Ł, Sędek Ł, Gomułka K: The Role of Transforming Growth Factor-β (TGF-β) in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Cells 2024, 13:1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Stinson SE, Amrani Y, Brightling CE: D prostanoid receptor 2 (chemoattractant receptor-homologous molecule expressed on TH2 cells) protein expression in asthmatic patients and its effects on bronchial epithelial cells. J Allergy Clin Immunol 2015, 135:395–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Honda T, Matsuoka T, Ueta M, Kabashima K, Miyachi Y, Narumiya S: Prostaglandin E(2)-EP(3) signaling suppresses skin inflammation in murine contact hypersensitivity. J Allergy Clin Immunol 2009, 124:809–818.e802. [DOI] [PubMed] [Google Scholar]
- 115.Akaba T, Komiya K, Suzaki I, Kozaki Y, Tamaoki J, Rubin BK: Activating prostaglandin E2 receptor subtype EP4 increases secreted mucin from airway goblet cells. Pulm Pharmacol Ther 2018, 48:117–123. [DOI] [PubMed] [Google Scholar]
- 116.Karra L, Haworth O, Priluck R, Levy BD, Levi-Schaffer F: Lipoxin B₄ promotes the resolution of allergic inflammation in the upper and lower airways of mice. Mucosal Immunol 2015, 8:852–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Seo EH, Lee SH, Choi BY, Oh CS, Kim JK: Exogenous Lipoxin A4 attenuates IL4-induced Mucin Expression in Human Airway Epithelial Cells. Int J Med Sci 2023, 20:406–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Eggel A, Pennington LF, Jardetzky TS: Therapeutic monoclonal antibodies in allergy: Targeting IgE, cytokine, and alarmin pathways. Immunol Rev 2024, online ahead of print, doi: 10.1111/imr.13380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Corren J, Parnes JR, Wang L, Mo M, Roseti SL, Griffiths JM, van der Merwe R: Tezepelumab in Adults with Uncontrolled Asthma. N Engl J Med 2017, 377:936–946. *This landmark study was the first to show that an mAb directed at an epithelial-derived cytokine could successfully ameliorate allergic asthma in humans. Anti-TSLP treatment resulted in clinically significant symptom improvement over placebo in patients with uncontrolled asthma.*
- 120.Wechsler ME, Ruddy MK, Pavord ID, Israel E, Rabe KF, Ford LB, Maspero JF, Abdulai RM, Hu CC, Martincova R, et al. : Efficacy and Safety of Itepekimab in Patients with Moderate-to-Severe Asthma. N Engl J Med 2021, 385:1656–1668. [DOI] [PubMed] [Google Scholar]
- 121.England E, Rees DG, Scott IC, Carmen S, Chan DTY, Chaillan Huntington CE, Houslay KF, Erngren T, Penney M, Majithiya JB, et al. : Tozorakimab (MEDI3506): an anti-IL-33 antibody that inhibits IL-33 signalling via ST2 and RAGE/EGFR to reduce inflammation and epithelial dysfunction. Sci Rep 2023, 13:9825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Chen YL, Gutowska-Owsiak D, Hardman CS, Westmoreland M, MacKenzie T, Cifuentes L, Waithe D, Lloyd-Lavery A, Marquette A, Londei M, et al. : Proof-of-concept clinical trial of etokimab shows a key role for IL-33 in atopic dermatitis pathogenesis. Sci Transl Med 2019, 11:eaax2945. [DOI] [PubMed] [Google Scholar]
- 123.Chinthrajah S, Cao S, Liu C, Lyu SC, Sindher SB, Long A, Sampath V, Petroni D, Londei M, Nadeau KC: Phase 2a randomized, placebo-controlled study of anti-IL-33 in peanut allergy. JCI Insight 2019, 4:e131347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kelsen SG, Agache IO, Soong W, Israel E, Chupp GL, Cheung DS, Theess W, Yang X, Staton TL, Choy DF, et al. : Astegolimab (anti-ST2) efficacy and safety in adults with severe asthma: A randomized clinical trial. J Allergy Clin Immunol 2021, 148:790–798. [DOI] [PubMed] [Google Scholar]
- 125.Maurer M, Cheung DS, Theess W, Yang X, Dolton M, Guttman A, Choy DF, Dash A, Grimbaldeston MA, Soong W: Phase 2 randomized clinical trial of astegolimab in patients with moderate to severe atopic dermatitis. J Allergy Clin Immunol 2022, 150:1517–1524. [DOI] [PubMed] [Google Scholar]
- 126.Powell WS: Eicosanoid receptors as therapeutic targets for asthma. Clin Sci (Lond) 2021, 135:1945–1980. [DOI] [PubMed] [Google Scholar]
- 127.Yang D, Guo X, Liu T, Li Y, Du Z, Liu C: Efficacy and Safety of Prostaglandin D2 Receptor 2 Antagonism with Fevipiprant for Patients with Asthma: a Systematic Review and Meta-analysis of Randomized Controlled Trials. Curr Allergy Asthma Rep 2021, 21:39. [DOI] [PubMed] [Google Scholar]




