Skip to main content

Biological mechanisms and therapeutic prospects of interleukin-33 in pathogenesis and treatment of allergic disease

Abstract

Allergic diseases significantly impact the quality of life of people around the world. Cytokines play a crucial role in regulating the immune system. Due to their importance in pro-inflammatory mechanisms, cytokines are used to understand pathogenesis and serve as biomarkers in many diseases. One such cytokine is interleukin-33, a member of the IL-1 family, including IL- 1α, IL-1β, and IL-18. The IL-33 receptor is a heterodimer of IL-1 receptor-like 1 and IL-1 receptor accessory protein. IL-33 plays a critical role in regulating innate and adaptive immune responses. The primary targets of IL-33 in vivo are tissue-resident immune cells, including mast cells, group 2 innate lymphoid cells, regulatory T cells, T helper 2 cells, eosinophils, basophils, dendritic cells, Th1 cells, CD8 + T cells, NK cells, iNKT cells, B cells, neutrophils, and macrophages. However, IL-33 appears to act as an alarm signal that is promptly released by producing cells under cellular damage or stress conditions. IL-33 regulates signaling and various biological functions, including induction of pro-inflammatory cytokines, regulation of cell proliferation, and involvement in tissue remodeling. IL-33 is fundamental in immune-related diseases and plays a critical role in the control of inflammation. Recently, IL-33 has been shown to significantly impact allergic diseases, primarily by inducing Th2 immune responses. IL-33 is a key regulator of mast cell function and a promising therapeutic target for treating allergic diseases. This review provides an overview of the current understanding of the role of IL-33 in allergy pathogenesis and potential clinical approaches.

Introduction

Allergic disorders are widespread in developed countries and have an essential socio-economic impact [1]. Allergic rhinitis (AR), asthma, and atopic dermatitis (AD) are examples of chronic allergic diseases that persist throughout life [2, 3]. Recent clinical and experimental studies have helped to understand the underlying mechanisms and to develop targeted therapies. Ultimately, allergen exposure triggers mast cell stimulation and degranulation, eosinophil employment and penetration, T helper (Th2) cell activation and differentiation, and antigen-specific immunoglobulin E (IgE) creation. There is now a bias towards a type 2 cytokine response, categorized by excessive manufacture of IL-4, IL-5, and IL-13 cytokines in the upper and lower airways [4, 5]. IL-4 initiates and encourages Th2 replies and also triggers these effector cells, whereas IL-13 donates to the advancement of allergic answers by stimulating mucus creation and eosinophilic penetration by making eotaxin [6, 7]. Upon FcεRI binding, basophils and mast cells manufacture numerous chemical intermediaries, including histamine and lipid metabolites [8]. The airway epithelium is the primary protection contour, defending the airway from pathogens and potential threats [9, 10]. Nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) and Toll-like receptors (TLRs) are taking place in the outside of epithelial cells and enable them to identify structurally preserved pathogen-associated molecular patterns (PAMPs). Upon encountering these external danger signs, epithelial cells initiate immunity replies [11, 12]. The primary epithelial response to PAMP-TLR/NLR communication is the creation of numerous cytokines that can stimulate specific immune responses, bias the type 2 immune response, and reduce inflammation [12, 13]. Cytokines perform a vital role in the instruction of adaptive and innate systems and have been used to comprehend pathogenesis and as biomarkers in different illnesses, including allergic disorders, autoimmune disorders, and cancer [14, 15]. Recent studies suggest that IL-33 plays a significant role in allergic disorders [16, 17]. This review focuses on the structure, biology, and currently reported role of the IL-33 cytokine and its potential clinical implications in allergic inflammatory disorders.

IL-33: sources, structure, functions

IL-33 (IL-1F11) was first recognized as high endothelial venule nuclear protein (NF-HEV) in endothelial cells of human lymphoid tissues [18, 19]. In 2005, IL-33 was reclassified as a participant in the IL-1 superfamily [19]. IL-33 has a double job as an extracellular cytokine of the IL-1 superfamily (pro-inflammatory) and intracellularly as a nuclear factor regulating gene appearance [19, 20]. Intracellularly, IL-33 has transcriptional repressive properties, while nuclear IL-33 attenuates pro-inflammatory signaling via appropriating nuclear NF-kB, reducing NF-kB-driven gene expression [21]. Conversely, IL-33 encourages inflammation in endothelial cells by upregulating the p65 subunit of the NF-kB compound in the nucleus. In the excite of cells, IL-33 functions as a pro-inflammatory cytokine, and its biological properties are initiated by connecting to their receptor [22]. The IL-33 gene is situated on chromosome 9 at 9p24.1 [23]. The human IL-33 gene contains eight exons and produces a 270 amino acid protein (30 kDa) with two evolutionarily conserved domains [24, 25]. The cytokine area is structured like IL-1 and has folding possessions analogous to IL-1α, IL-1β, and IL-18 [26, 27]. Under normal conditions, complete IL-33 is continuously present in numerous cell kinds throughout human tissues and is situated in their nuclei [19, 23].

The main foundations of activity in humans include human endothelial cells, epithelial cells in barrier tissues, and reticular fibroblast cells in lymphoid tissues. The nervous system comprises glial cells, neurons and astrocytes, smooth muscle cells, osteoblasts, adipocytes, and various immune cells such as macrophages, mast cells, monocytes, and dendritic cells (DCs). This suggests a significant character in defending the body against damage and contamination in standard and recurrently irritated tissues and tumor tissue [23, 28]. In the inactive situation, IL-33 is located in the nucleus and bound to chromatin via a chromatin-connecting domain, a critical component of cellular homeostasis that acts as a transcriptional repressor [29, 30]. IL-33 shares similarities with alarmins or damage-associated molecular patterns (DAMPs), pro-inflammatory factors released through injured cells [31, 32]. Alarmins perform as intercellular indications via binding to chemotactic and pattern recognition receptors (PRRs) to enhance immunity cell responses in host protection [18]. IL-33 has multiple effects on the immune response. It is a component of the host’s immunity to pathogens or immune-related inflammatory disorders. The innate immune system can produce IL-33 to respond to infectious agents and irritants [33, 34]. PAMP-generated IL-33 is vital in encouraging local airway inflammation related to Th2-dependent antigens and augmenting innate immunity [35, 36]. It can straightly cause airway inflammation and is frequently not required to initiate and distinguish antigen-specific Th2 cells [37, 38]. The IL-33 receptor (IL-33R) is a heterodimer containing a flexible ST2 (T1, IL-1RL1, IL-1R4) and a familiar, generally stated coreceptor known as IL-1 receptor accessory protein (IL-1RAcP) (IL-1R3) [39, 40]. ST2, a toll-like receptor/interleukin-1 receptor (TIR) superfamily participant, constitutes the IL-33 receptor [41, 42].

ST2 is the sole receptor for IL-33, although it has four splice isoforms from a single transcript according to promoter complicated [33, 43]. The two main products of ST2 genes, which result from substitute splicing, are transmembrane shape ST2 (ST2 or ST2L) and soluble shape ST2 (sST2) [44, 45]. ST2L is a functional element of IL-33R that is selectively and persistently stated on a diversity of immune cells such as eosinophils, mast cells, ILC2, basophils, DCs, NKs, Th2 cells, macrophages, Tregs, B cells, airway smooth muscle cells, also on endothelial cells, epithelial cells and fibroblasts [23, 46, 47]. Conversely, sST2 is considered to be an IL-33 decoy receptor [20, 48]. The heterodimer formed by the binding of IL-33, ST2, and IL-1RAP results in dimerization of the TIR domain, which stimulates downstream signaling through the heterodimeric complex. IARK-1, IARK-4, and mitogen-activated protein kinase (MAPK) kinases activate the TNF receptor-associated factor 6 (TRAF6) signaling pathway, which in turn activates activator protein 1 (AP-1) through c-Jun N-terminal kinases (JNKs). TRAF6 also activates the inhibitor of nuclear factor-κB (NF-κB) kinase (IKK) complex, releasing active NF-κB from the complex and activating nuclear transcription signals necessary for creating other inflammatory cytokines. Upon receptor stimulation, Jun kinase (JNK) and extracellular signal-regulated kinase (ERK) 1/2 promote IRF1 activation, resulting in the inhibition of Foxp3 and GATA3 expression [49], thereby inducing inflammatory cytokine production [50, 51](Fig. 1).

Fig. 1
figure 1

IL-33 impact in different cells. IL-33 is primarily expressed by different types of structural cells. IL-33 through linking to ST2 + immune cells cause different effect on cells, such as Induce production of various cytokines and chemokines by mast cells or Induces eosinophilia. transmembrane shape (ST2)

Altering the subcellular localization of IL-33 may significantly impact immune homeostasis, suggesting that nuclear sequestration of IL-33 may limit its pro-inflammatory potential [52]. IL-33 may also perform as a link among non-specific and specific immune cells, promoting Th2-mediated inflammation [53, 54]. In reply to IL-33, ILC2 can multiply and secrete IL-5 and IL-13 in mouse models [55, 56]. Also, IL-33 can activate further non-specific immune cells such as basophils, macrophages, mast cells, and eosinophils, leading to a more significant contribution to airway inflammation [55, 56]. In addition, IL-33 can affect specific immunity by stimulating ILC2 and DCs and adaptive immune cells that express the ST2 receptor. Therefore, IL-33 induces DC to make a variety of cytokines and chemokines [57, 58].

Exposure to IL-33 can induce DCs to support the extension and specialization of CD4 + T cells by promoting the manufacture of IL-5 and IL-13 [59, 60]. IL-33 plays a critical character in the immune response associated with Th2 [63, 64], with ST2-expressing Th2 cell’s ability to generate IL-5 and IL-13 in response to IL-33 [61]. When combined with an antigen, IL-33 can polarize human naive CD4 + T cells towards IL-5 manufacture. In addition, differentiation of IL-5 + T cells was induced by the classical IL-33 ways, including MyD88, MAPKs, and NF-κB only in the attendance of the IL-1R addition protein [62, 63]. IL-33 has been rolled into numerous non-allergic disorders such as infectious diseases, musculoskeletal diseases, inflammatory bowel disease, cancer, obesity, and diabetes [33, 51]. IL-33 strongly stimulates the manufacture of Th2 cytokines and thus exacerbates the progression of Th2-associated diseases such as asthma and allergic rhinitis [64]. In addition, IL-33 has been revealed to play an essential character in immune regulatory responses [65, 66]. The genes programming IL-33 and ST2/IL1RL1 have been recognized as significant vulnerability loci for human asthma in numerous genome-wide association educations [67, 68]. Manufacture of pro-inflammatory cytokines and activation of Th2-type immune cells can exacerbate allergic diseases [39, 69]. Several studies have shown that IL-33 is a potent activator of mast cells and basophils, inducing cell migration, maturation, adhesion, and survival while producing several pro-inflammatory cytokines [70, 71]. Examination of patient samples and mouse models supports the critical character of the IL-33/ILC2 axis in allergic inflammation in numerous tissues and disorders [69, 72]. The essential function of endogenous IL-33 in allergic inflammation was primarily established in IL-33-deficient mice [73]. In addition, IL-33 expression is increased in bronchial epithelial cells of samples with asthma and epidermal keratinocytes of patients with atopic dermatitis [74, 75]. Increased expression of IL-33 and ST2 has been reported in lung tissue from both asthmatic and allergic airways, with ST2 playing a critical character in antigen-induced airway inflammation [76].

IL-33 in different allergic diseases

Asthma

The incidence of allergic asthma has amplified significantly in industrialized countries over the last century (ranging from 5.2 to 16.8%) [77]. Asthma is highly heterogeneous, and all endotypes are typically characterized via airway hyperresponsiveness (AHR), tissue renovation, and extreme Th2 inflammation [78, 79]. Asthma is now understood to be a multifaceted disease with different entities and some variation in pathogenesis [45, 79]. Previous research has revealed that a principal Th2 lymphocyte answer may contribute to the development of asthma symptoms, primarily through the secretion of cytokines such as IL-4, IL-5, and IL-13 [45, 68].

In calculation, nonspecific immune cells such as mast cells, basophils, neutrophils, EOSs, DC, and ILC2s have been occupied in the beginning or development of the disorders [80, 81]. Recent studies have shown that the motivation of cells of the nonspecific immune system triggers the stimulation of NF-kB, which results in the creation of pro-Th2 cytokines such as IL-33 [59, 82]. Airway epithelial cells and APCs enhance allergic airway inflammation and thus the pathophysiological symbols of asthma through discharging cytokines such as IL-1β and IL-6. IL-33 derived from Epithelium encourages Th2 and ILC2 distinction, which straight allergic immune responses via the release of IL-4, IL-5, and IL-13 [6, 63, 82].

IL-33, an important allergy cytokine, participates in nonspecific and specific immunity in asthmatic immune stimulation [79, 83]. This cytokine directly stimulates macrophages, osteoclasts, Th2 cells, mast cells, DCs, and other subsets that express T1/ST2, the IL-33 receptor [84]. Downstream IL-33 provokes high levels of IL-5 and IL-13 manufactured by IL-33-irritated ILC2s, resulting in a strong pro-allergic response [85]. A genetic polymorphism in IL-33 or ST2 has been recognized in patients with asthma [86]. Two single nucleotide polymorphisms (SNPs) in ST2 - rs1420101 and rs11685480 - have been suggested to control plasma sST2 levels in airway epithelial cells and the distal lung parenchyma, correspondingly, subsequent in an amplified risk of classical airway inflammation in asthmatics [87]. Significant correlations were found between the plasma parts of the IL-33/ST2 axis in patients with allergic rhinitis and those with associated allergic asthma [88]. Remarkably, IL-33 appearance is upregulated in the bronchial mucosa of asthmatic patients. This is associated with disease harshness [89]. In a mouse model, airway inflammation and AHR were decreased by knocking out ILC2s or blocking IL-33-ST2 signaling [90]. in asthma, When basophils stimulated by IL-33, basophils express ST2 and create Th2 cytokines [91]. Another study showed that in vitro motivation with IL-33 amplified ST2L mRNA expression in eosinophils, leading to improved eosinophil adhesiveness and CD11b expression, also feasibility, which was reduced by a deactivating antibody to ST2 [44](Fig. 2).

Fig. 2
figure 2

IL-33 in allergic diseases pathology. IL-33 produce by different cell such as epithelial cells and bind to its receptor, then activates various ST2 + immune cell types implicated in the allergic responses for example, Th2, ILC, Mast cell. These cells produce cytokine such as IL-4, IL-5 and IL-13, that impact on EOS for mediator releases. Innate lymphoid cells (ILC2s); Eosinophil (EOS)

Given the significance of IL-33 signaling in allergic asthma, numerous biologics have been advanced and tried as potential treatments. Inhibition of the AHR reduced IL-5 levels in BALF, and reduced eosinophil numbers were observed in an OVA challenge mouse model preserved with anti-IL-33 or sST2 [92]. Monoclonal antibodies such as RG6149 [93], and CNTO7160 [94], either against ST2 or IL-33, such as ANB020 [93], REGN3500 [93], and MEDI3506, have undergone or completed clinical trials. A high-affinity monoclonal antibody against dokimab (LY3375880) is in preclinical development to inhibit IL-33-dependent inflammatory signaling [95].

Allergic rhinitis

Allergic rhinitis (AR) is a public allergic inflammatory disease in 10–40% of the population [96, 97]. AR is an IgE-mediated type 1 allergic disorder of the nasal mucosa triggered via exposure to nasal allergens. Nasal responses in AR consist of IgE-related early-stage responses and Th2 cytokine-related late-stage responses [98]. Due to early-stage responses, medical signs, including sneezing and rhinorrhea, happen within 5–30 min. Late-stage responses occur 6–24 h after allergen exposure, and Eosinophilic accumulation in the nasal mucosa is the primary pathological change associated with the late-stage response [98, 99]. A previous study established an essential rise in serum IL-33 levels in Japanese patients suffering from seasonal AR and also found a significant correlation between vulnerability to AR and polymorphisms in the IL-33 gene [100]. Notably, the amount of IL-33-responsive ILCs was also found to be amplified in nasal polyps from patients with rhinosinusitis [101]. After exposure to ragweed, mice show exacerbated symptoms of AR, activation of nasal Th2, increased serum levels of ragweed-specific IgE, and penetration of the nasal mucosa by eosinophils and basophils, which are not detected in IL-33- and ST2-deficient mice [102]. IL-33 protein is consistently stated in the nucleus of nasal epithelial cells and is rapidly released into the nasal fluid in response to ragweed pollen exposure [103]. Collected with the involvement of IL-33 in activating eosinophils, basophils, and mast cells to manufacture allergic inflammatory mediators, this mechanism may result in the persistent attacks and permanent mucosal hypertrophy seen in AR [104, 105]. However, during the pollen season, IL-33 mRNA appearance is significantly upregulated in nasal epithelial cells of AR patients, and this increased extracellular statement is related to decreased IL-33 protein appearance in inflamed nasal epithelial cells [103]. Furthermore, the IL-33 receptor ST2 is significantly stated in the nasal mucosa of AR patients, proposing that IL-33/ST2 may be critical in developing allergic nasal disease [52, 106]. In addition, the IL-33-ST2 encouraged the Th2 response to cooperate with the Th17 immune response, demonstrating numerous T cell responses in developing allergic nasal disease [103, 107]. Targeting IL-33 and its receptor represents a new therapeutic method for handling allergic rhinitis [108]. Anti-IL-33 treatment decreases nasal scratching, improves skin stripping, and decreases eosinophilic infiltration and serum total and OVA-specific IgE levels [109, 110] (Table 1).

Food allergy (FA)

A food allergy is an abnormal immune response to an antigen ingested through food [111]. The medical features of FA are varied and random, influenced by many factors—symptoms of FA vary from oral allergy syndrome to life-threatening anaphylaxis [111, 112]. The prevalence of food allergy is growing worldwide, affecting both industrialized and developing countries, with about 8% of children and 11% of adults in the United States affected [113]. Symptoms of food allergy can differ from somebody to somebody, with some experiencing mild abdominal discomfort and others experiencing severe, dangerous anaphylaxis [114, 115]. Consequently, food allergy has public health and economic implications [116]. In the sensitivity stages, food allergens first come into contact with protective surfaces on the skin, gastrointestinal tract, or respiratory tract. Skin barrier surfaces with defects and a decrease in their honesty increase the permeability of allergens, producing proinflammatory cytokines derived from epithelial tissue, particularly IL-33 and IL-25 [46, 117]. Together, these cytokines begin an allergic inflammatory process that leads to allergic responses in individuals with food allergies [8, 115]. Cytokines activate ILC2s, induce distinction of naive CD4 + T cells into Th2 cells, create proinflammatory cytokines such as IL-4, IL-5, IL-9, and IL-13, induce isotype switching from IgG to IgE in B cells, trigger linking of IgE to FcεRI receptors on mast cells or basophils followed by degranulation, and result in the liberate of histamine and other proinflammatory allergic mediators [8, 81, 111, 115]. Animal surveys found that epithelial cell-derived IL-33 is vital in advancing food allergy [118]. Also, another study showed that skin exposure to peanut induced cytokine appearance in T cells and dendritic cells that was reliant on IL-33-ST2 signaling, signifying that IL-33 may perform a role in mediating food allergy through skin exposure early in life [118, 119]. At the sensitization stage, IL-33 induced intestinal anaphylaxis through cross-linking of IgE and degranulation of mast cells [118, 120]. In calculation to mast cells, IL-33 can stimulate ILCs and overwhelm the Treg role, which promotes FA immunity [121]. The study proposes that IL-33 plays a significant role in the association between barrier defects and FA [120, 121]. Recently, a study reported that injection of an mAb against the IL-33 receptor effectively prevents the advancement of FA [122]. Thus, IL-33 and its receptor ST2 may serve as potential therapeutic goals to attenuate the mast cell-associated pathogenesis of FA [119]. Some suggest that IL-33 inhibitors may be of healing worth in FA patients (Table 2) [123].

Atopic dermatitis

Atopic dermatitis (AD) is a primary inflammatory skin condition branded by long-lasting or recurrent pruritus and may be associated with other atopic situations such as asthma and food allergy [124]. AD has a higher occurrence in children but can also disturb adults [125]. It is more likely to occur in people with allergies [126, 127]. AD is associated with over-reactivity of the skin, disruption of the epidermal barrier role, dry skin, and increased vulnerability to skin contaminations. The cause of inflammation has been ascribed to the stimulation of various immune cells. Mast cells are critical in initiating the disease, while Tregs performance is essential in controlling the inflammatory response [128]. AD is characterized by Th2 cytokines in the acute phase and raised levels of Th1 cytokines in the lasting phase [126, 128]. A dysfunction in the nonspecific and specific immune response could initiate skin injury, provoking an elevated Th2 response that exacerbates and progresses AD [129]. However, CD4 + T lymphocytes respond to IL-33 and stimulate interferon-gamma production. This promotes chronic skin inflammation [130]. In addition, IL-33, as tissue-derived cytokines, has recently been implicated in atopic dermatitis [131, 132]. Based on current explanations, IL-33 is identified as a prominent threat signal and pathogenic driving force in atopic dermatitis [131].

Imai et al. established that a transgenic mouse expressing IL-33 impulsively advances AD by stimulating ILC2s and basophils [132]. Additionally, IL-33 was found to be a significant regulator of eosinophil role [132]. Shimizu et al. investigated the correlation between AD and a polymorphism of the ST2 gene and proposed that the IL-33-ST2 axis plays a crucial character in AD [133]. Other studies showed the upregulation of IL-33 in the epidermis and the penetration of ST2-positive cells in the dermis of skin lesions in AD patients [75, 134]. An SNP located in the distal promoter of the ST2 gene on chromosome 2q12 shows a significant association with AD, and there is a robust association between tall soluble ST2 and total IgE stages in the sera of AD patients [135].

Still, mRNA levels of IL-33 and ST2 were significantly higher in AD patients’ affected skin than in non-affected skin and healthy people [75]. Various treatments have been advanced to heal AD, including topical steroids, calcineurin inhibitors, and immunosuppressive agents [133]. Blocking IL-33/ST2 signaling may be a potential healing goal for alleviating the pruritus and skin inflammation of dermatitis related to IL-33/ST2 signaling [75, 136].

Allergic conjunctivitis

Th2 responses on the ocular surface are responsible for causing inflammation to various allergens, which can lead to tissue damage and infection, ultimately resulting in the advancement and aggravation of atopic keratoconjunctivitis [137]. IL-33 protein has been reported to be highly stated in human vascular endothelial cells and conjunctival epithelium in giant papillae gained from patients with atopic keratoconjunctivitis [138]. After administration of ragweed pollen, IL-33 is released, leading to penetration of eosinophils and CD4 + T cells with stimulated ST2 on the surface of the conjunctival stroma [139]. These activations are significantly reduced in IL-33 knockout mice [140].

Chronic spontaneous urticaria (CSU) and anaphylaxis

CSU is a common skin condition considered by the presence of itchy wheals that last less than 24 h and persist for at least six weeks and often for several decades [141]. CSU is an autoimmune disorder with hive formation instruments reliant on IgG and IgE [142]. Despite a favorable prognosis, CSU is a debilitating disease as its primary symptom, pruritus, can cause sleep trouble and harmfully affect quality of life and work performance [143]. There are signs of a role for IL-33 in the pathogenesis of CSU [144]. In particular, studies of skin biopsies from patients with CSU have shown significantly elevated levels of IL-33 in contrast to healthy controls, proposing that the appearance of Th2-promoting cytokines may play an essential role in healing [145]. Another study proposes that IL-33 signaling may be a critical orientation of histaminergic itch in mast cell-associated pruritic situations such as CSU [146]. This suggests that IL-33 plays a character in causing and expressing CSU.

IL-33 levels in the serum of atopic patients increase significantly during anaphylaxis [147, 148], suggesting that IL-33 may play a character in advancing anaphylaxis. Similarly, IL-33 can induce mast cell degranulation via ST2, leading to systemic and passive cutaneous anaphylaxis in IgE-sensitized mice, even in the nonappearance of a specific IgE antigen [147, 149]. This suggests that the IL-33-ST2 pathway plays a vital role in inflammation during the later stages of passive cutaneous anaphylaxis in mice.

Table 1 Animal model and in vitro surveys of IL-33 allergic diseases
Table 2 Human studies of IL-33 in allergic diseases

Concluding remark

IL-33 is a tissue-derived nuclear cytokine from the IL-1 family, expressed in endothelial cells, epithelial cells, and fibroblast-like cells under both homeostasis and inflammation. It has been identified as an alarmin cytokine from the IL-1 family; IL-33 has appeared as a central modulator of tissue Tregs and ILC2s, with significant characteristics in type-2, type-1, and regulatory immune responses. Current studies suggest that IL-33 promotes the manufacture of substantial quantities of IL-5 and IL-13 by ILC2s, potentially contributing to the initiation of allergic inflammation. The intense activity of IL-33 on ILC2s, and the pivotal role of these cells in the early stages of allergic airway inflammation, may help explain the involvement of the IL-33/ST2 pathway in the genetic susceptibility to human allergic diseases. Beyond its pathological role in allergic diseases, IL-33 also contributes to various immune regulatory processes. However, further studies are needed to clarify the precise therapeutic potential of targeting the IL-33/ST2 axis in different disease contexts and its physiological roles, as current evidence is still evolving.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Zuberbier T, Lötvall J, Simoens S, Subramanian S, Church MK. Economic burden of inadequate management of allergic diseases in the European Union: a GA2LEN review. Allergy. 2014;69:1275–9.

    Article  CAS  PubMed  Google Scholar 

  2. Samitas K, Carter A, Kariyawasam H, Xanthou G. Upper and lower airway remodelling mechanisms in asthma, allergic rhinitis and chronic rhinosinusitis: the one airway concept revisited. Allergy. 2018;73:993–1002.

    Article  CAS  PubMed  Google Scholar 

  3. Yii A, Tay TR, Choo X, Koh M, Tee A, Wang DY. Precision medicine in united airways disease: a treatable traits approach. Allergy. 2018;73:1964–78.

    Article  CAS  PubMed  Google Scholar 

  4. Agrawal K, Arora N. Serine protease allergen favours Th2 responses via PAR-2 and STAT 3 activation in murine model. Allergy. 2018;73:569–75.

    Article  CAS  PubMed  Google Scholar 

  5. Bousquet J, Akdis CA, Grattan C, Eigenmann PA, Hoffmann-Sommergruber K, Agache I, Jutel M. Highlights and recent developments in airway diseases in EAACI journals (2018). Allergy. 2019;74:2329–41.

    Article  PubMed  Google Scholar 

  6. Lloyd CM, Snelgrove RJ. Type 2 immunity: expanding our view. Sci Immunol. 2018;3:eaat1604.

    Article  PubMed  Google Scholar 

  7. Paul WE, Zhu J. How are TH2-type immune responses initiated and amplified? Nat Rev Immunol. 2010;10:225–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Crespo JF, Cabanillas B. (2023) Recent advances in cellular and molecular mechanisms of IgE-mediated food allergy. Food Chemistry: 135500.

  9. Persson C. Airways exudation of plasma macromolecules: innate defense, epithelial regeneration, and asthma. J Allergy Clin Immunol. 2019;143:1271–86.

    Article  CAS  PubMed  Google Scholar 

  10. Ogulur I, Pat Y, Ardicli O, Barletta E, Cevhertas L, Fernandez-Santamaria R, Huang M, Bel Imam M, Koch J, Ma S. Advances and highlights in biomarkers of allergic diseases. Allergy. 2021;76:3659–86.

    Article  PubMed  Google Scholar 

  11. Girkin JL, Maltby S, Bartlett NW. (2022) Toll-like receptor-agonist-based therapies for respiratory viral diseases: thinking outside the cell. Eur Respiratory Rev 31.

  12. Johnston SL, Goldblatt DL, Evans SE, Tuvim MJ, Dickey BF. Airway epithelial innate immunity. Front Physiol. 2021;12:749077.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Patel NN, Kohanski MA, Maina IW, Workman AD, Herbert DBR, Cohen NA. Sentinels at the wall: epithelial-derived cytokines serve as triggers of upper airway type 2 inflammationInternational. forum of allergy & rhinology Wiley Online Library; 2019. pp. 93–9.

  14. Eivary SHA, Kheder RK, Najmaldin SK, Kheradmand N, Esmaeili S-A, Hajavi J. Implications of IL-21 in solid tumor therapy. Med Oncol. 2023;40:191.

    Article  CAS  PubMed  Google Scholar 

  15. Liu C, Chu D, Kalantar-Zadeh K, George J, Young HA, Liu G. Cytokines: from clinical significance to quantification. Adv Sci. 2021;8:2004433.

    Article  CAS  Google Scholar 

  16. Mitchell PD, Salter BM, Oliveria JP, El-Gammal A, Tworek D, Smith SG, Sehmi R, Gauvreau GM, O’Byrne PM. IL-33 and its receptor ST2 after inhaled allergen challenge in allergic asthmatics. Int Arch Allergy Immunol. 2018;176:133–42.

    Article  CAS  PubMed  Google Scholar 

  17. Oshikawa K, Kuroiwa K, Tago K, Iwahana H, Yanagisawa K, Ohno S, Tominaga S-i, Sugiyama Y. Elevated soluble ST2 protein levels in sera of patients with asthma with an acute exacerbation. Am J Respir Crit Care Med. 2001;164:277–81.

    Article  CAS  PubMed  Google Scholar 

  18. Baekkevold ES, Roussigné M, Yamanaka T, Johansen F-E, Jahnsen FL, Amalric F, Brandtzaeg P, Erard M, Haraldsen G, Girard J-P. Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules. Am J Pathol. 2003;163:69–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cayrol C, Girard JP. Interleukin-33 (IL‐33): a nuclear cytokine from the IL‐1 family. Immunol Rev. 2018;281:154–68.

    Article  CAS  PubMed  Google Scholar 

  20. Bianchetti L, Marini MA, Isgrò M, Bellini A, Schmidt M, Mattoli S. IL-33 promotes the migration and proliferation of circulating fibrocytes from patients with allergen-exacerbated asthma. Biochem Biophys Res Commun. 2012;426:116–21.

    Article  CAS  PubMed  Google Scholar 

  21. Ali S, Mohs A, Thomas M, Klare J, Ross R, Schmitz ML, Martin MU. The dual function cytokine IL-33 interacts with the transcription factor NF-κB to dampen NF-κB–stimulated gene transcription. J Immunol. 2011;187:1609–16.

    Article  CAS  PubMed  Google Scholar 

  22. Choi Y-S, Park JA, Kim J, Rho S-S, Park H, Kim Y-M, Kwon Y-G. Nuclear IL-33 is a transcriptional regulator of NF-κB p65 and induces endothelial cell activation. Biochem Biophys Res Commun. 2012;421:305–11.

    Article  CAS  PubMed  Google Scholar 

  23. Liew FY, Girard J-P, Turnquist HR. Interleukin-33 in health and disease. Nat Rev Immunol. 2016;16:676–89.

    Article  CAS  PubMed  Google Scholar 

  24. Mehrabi Nasab E, Hassanzadeh Makoei R, Aghajani H, Athari SS. IL-33/ST2 pathway as upper‐hand of inflammation in allergic asthma contributes as predictive biomarker in heart failure. ESC Heart Fail. 2022;9:3785–90.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Trimarchi M, Lauritano D, Ronconi G, Caraffa A, Gallenga CE, Frydas I, Kritas SK, Calvisi V, Conti P. Mast cell cytokines in Acute and Chronic Gingival tissue inflammation: role of IL-33 and IL-37. Int J Mol Sci. 2022;23:13242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dwyer GK, D’Cruz LM, Turnquist HR. Emerging functions of IL-33 in homeostasis and immunity. Annu Rev Immunol. 2022;40:15–43.

    Article  CAS  PubMed  Google Scholar 

  27. Fields JK, Günther S, Sundberg EJ. Structural basis of IL-1 family cytokine signaling. Front Immunol. 2019;10:1412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nguyen PT, Dorman LC, Pan S, Vainchtein ID, Han RT, Nakao-Inoue H, Taloma SE, Barron JJ, Molofsky AB, Kheirbek MA. Microglial remodeling of the extracellular matrix promotes synapse plasticity. Cell. 2020;182:388–403. e315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gautier V, Cayrol C, Farache D, Roga S, Monsarrat B, Burlet-Schiltz O, Gonzalez de Peredo A, Girard J-P. Extracellular IL-33 cytokine, but not endogenous nuclear IL-33, regulates protein expression in endothelial cells. Sci Rep. 2016;6:34255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Thijs C, Müller A, Rist L, Kummeling I, Snijders B, Huber M, Van Ree R, Simões-Wüst A, Dagnelie P, Van Den Brandt P. Fatty acids in breast milk and development of atopic eczema and allergic sensitisation in infancy. Allergy. 2011;66:58–67.

    Article  CAS  PubMed  Google Scholar 

  31. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20:95–112.

    Article  CAS  PubMed  Google Scholar 

  32. Pandolfi F, Altamura S, Frosali S, Conti P. Key role of DAMP in inflammation, cancer, and tissue repair. Clin Ther. 2016;38:1017–28.

    Article  CAS  PubMed  Google Scholar 

  33. De la Fuente M, MacDonald TT, Hermoso MA. The IL-33/ST2 axis: role in health and disease. Cytokine Growth Factor Rev. 2015;26:615–23.

    Article  PubMed  Google Scholar 

  34. Di Salvo E, Ventura-Spagnolo E, Casciaro M, Navarra M, Gangemi S. (2018) IL-33/IL-31 axis: a potential inflammatory pathway. Mediators of inflammation 2018.

  35. Cayrol C, Girard J-P. IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr Opin Immunol. 2014;31:31–7.

    Article  CAS  PubMed  Google Scholar 

  36. Martin MU. (2013) Special aspects of interleukin-33 and the IL-33 receptor complexSeminars in immunology Elsevier, pp. 449–457.

  37. Endo Y, Hirahara K, Yagi R, Tumes DJ, Nakayama T. Pathogenic memory type Th2 cells in allergic inflammation. Trends Immunol. 2014;35:69–78.

    Article  CAS  PubMed  Google Scholar 

  38. Iinuma T, Okamoto Y, Morimoto Y, Arai T, Sakurai T, Yonekura S, Sakurai D, Hirahara K, Nakayama T. Pathogenicity of memory Th2 cells is linked to stage of allergic rhinitis. Allergy. 2018;73:479–89.

    Article  CAS  PubMed  Google Scholar 

  39. Aneas I, Decker DC, Howard CL, Sobreira DR, Sakabe NJ, Blaine KM, Stein MM, Hrusch CL, Montefiori LE, Tena J. Asthma-associated genetic variants induce IL33 differential expression through an enhancer-blocking regulatory region. Nat Commun. 2021;12:6115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Latiano A, Palmieri O, Pastorelli L, Vecchi M, Pizarro TT, Bossa F, Merla G, Augello B, Latiano T, Corritore G. Associations between genetic polymorphisms in IL-33, IL1R1 and risk for inflammatory bowel disease. PLoS ONE. 2013;8:e62144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Suzukawa M, Iikura M, Koketsu R, Nagase H, Tamura C, Komiya A, Nakae S, Matsushima K, Ohta K, Yamamoto K. An IL-1 cytokine member, IL-33, induces human basophil activation via its ST2 receptor. J Immunol. 2008;181:5981–9.

    Article  CAS  PubMed  Google Scholar 

  42. Zeng S, Wu J, Liu J, Qi F, Liu B. IL-33 receptor (ST2) signalling is important for regulation of Th2‐mediated airway inflammation in a murine model of acute respiratory syncytial virus infection. Scand J Immunol. 2015;81:494–501.

    Article  CAS  PubMed  Google Scholar 

  43. Pinto SM, Subbannayya Y, Rex D, Raju R, Chatterjee O, Advani J, Radhakrishnan A, Keshava Prasad T, Wani MR, Pandey A. A network map of IL-33 signaling pathway. J cell Communication Signal. 2018;12:615–24.

    Article  Google Scholar 

  44. Lee HY, Rhee CK, Kang JY, Byun JH, Choi JY, Kim SJ, Kim YK, Kwon SS, Lee SY. Blockade of IL-33/ST2 ameliorates airway inflammation in a murine model of allergic asthma. Exp Lung Res. 2014;40:66–76.

    Article  CAS  PubMed  Google Scholar 

  45. Lloyd CM. IL-33 family members and asthma–bridging innate and adaptive immune responses. Curr Opin Immunol. 2010;22:800–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dourado LPA, Noviello MLM, Alvarenga DM, Menezes Z, Perez DA, Batista NV, Menezes GB, Ferreira AVM, de Souza DG, Cara DC. Experimental food allergy leads to adipose tissue inflammation, systemic metabolic alterations and weight loss in mice. Cell Immunol. 2011;270:198–206.

    Article  CAS  PubMed  Google Scholar 

  47. Lee GR, Shin MK, Yoon DJ, Kim AR, Yu R, Park NH, Han IS. Topical application of capsaicin reduces visceral adipose fat by affecting adipokine levels in high-fat diet‐induced obese mice. Obesity. 2013;21:115–22.

    Article  PubMed  Google Scholar 

  48. Oshio T, Komine M, Tsuda H, Tominaga S-i, Saito H, Nakae S, Ohtsuki M. Nuclear expression of IL-33 in epidermal keratinocytes promotes wound healing in mice. J Dermatol Sci. 2017;85:106–14.

    Article  CAS  PubMed  Google Scholar 

  49. Siede J, Fröhlich A, Datsi A, Hegazy AN, Varga DV, Holecska V, Saito H, Nakae S, Löhning M. IL-33 receptor-expressing regulatory T cells are highly activated, Th2 biased and suppress CD4 T cell proliferation through IL-10 and TGFβ release. PLoS ONE. 2016;11:e0161507.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Lefrançais E, Duval A, Mirey E, Roga S, Espinosa E, Cayrol C, Girard J-P. Central domain of IL-33 is cleaved by mast cell proteases for potent activation of group-2 innate lymphoid cells. Proc Natl Acad Sci. 2014;111:15502–7.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Rostan O, Arshad MI, Piquet-Pellorce C, Robert-Gangneux F, Gangneux J-P, Samson M. Crucial and diverse role of the interleukin-33/ST2 axis in infectious diseases. Infect Immun. 2015;83:1738–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Soyka MB, Holzmann D, Basinski TM, Wawrzyniak M, Bannert C, Bürgler S, Akkoc T, Treis A, Rückert B, Akdis M. The induction of IL-33 in the sinus epithelium and its influence on T-helper cell responses. PLoS ONE. 2015;10:e0123163.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Jang TY, Kim YH. Interleukin-33 and mast cells bridge innate and adaptive immunity: from the allergologist’s perspective. Int Neurourol J. 2015;19:142.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Martin NT, Martin MU. Interleukin 33 is a guardian of barriers and a local alarmin. Nat Immunol. 2016;17:122–31.

    Article  CAS  PubMed  Google Scholar 

  55. Guo H, Bossila EA, Ma X, Zhao C, Zhao Y. Dual immune regulatory roles of interleukin-33 in pathological conditions. Cells. 2022;11:3237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Miller JE, Lingegowda H, Symons LK, Bougie O, Young SL, Lessey BA, Koti M, Tayade C. IL-33 activates group 2 innate lymphoid cell expansion and modulates endometriosis. JCI insight 6. 2021.

  57. Velayutham M, Roessing AS, Shiva S, Turnquist HR. IL-33 induces a metabolic programming supporting dendritic cells tolerogenicity. J Immunol 202: 2019;125.112-125.112.

  58. Enoksson M, Möller-Westerberg C, Wicher G, Fallon PG, Forsberg-Nilsson K, Lunderius-Andersson C, Nilsson G. Intraperitoneal influx of neutrophils in response to IL-33 is mast cell–dependent. Blood J Am Soc Hematol. 2013;121:530–6.

    CAS  Google Scholar 

  59. Drake LY, Kita H. IL-33: biological properties, functions, and roles in airway disease. Immunol Rev. 2017;278:173–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Rank MA, Kobayashi T, Kozaki H, Bartemes KR, Squillace DL, Kita H. IL-33–activated dendritic cells induce an atypical TH2-type response. J Allergy Clin Immunol. 2009;123:1047–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pastorelli L, Garg RR, Hoang SB, Spina L, Mattioli B, Scarpa M, Fiocchi C, Vecchi M, Pizarro TT. (2010) Epithelial-derived IL-33 and its receptor ST2 are dysregulated in ulcerative colitis and in experimental Th1/Th2 driven enteritis. Proceedings of the National Academy of Sciences 107: 8017–8022.

  62. Alvarez F, Fritz JH, Piccirillo CA. Pleiotropic effects of IL-33 on CD4 + T cell differentiation and effector functions. Front Immunol. 2019;10:522.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lu J, Kang J, Zhang C, Zhang X. The role of IL-33/ST2L signals in the immune cells. Immunol Lett. 2015;164:11–7.

    Article  CAS  PubMed  Google Scholar 

  64. Louten J, Rankin AL, Li Y, Murphy EE, Beaumont M, Moon C, Bourne P, McClanahan TK, Pflanz S, de Waal Malefyt R. Endogenous IL-33 enhances Th2 cytokine production and T-cell responses during allergic airway inflammation. Int Immunol. 2011;23:307–15.

    Article  CAS  PubMed  Google Scholar 

  65. Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in tissue homeostasis, injury, and inflammation. Immunity. 2015;42:1005–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Peine M, Marek RM, Löhning M. IL-33 in T cell differentiation, function, and immune homeostasis. Trends Immunol. 2016;37:321–33.

    Article  CAS  PubMed  Google Scholar 

  67. Falahi S, Mortazavi SHR, Salari F, Koohyanizadeh F, Rezaeimanesh A, Gorgin Karaji A. Association between IL-33 gene polymorphism (Rs7044343) and risk of allergic rhinitis. Immunol Investig. 2022;51:29–39.

    Article  CAS  Google Scholar 

  68. Savenije OE, John JMM, Granell R, Kerkhof M, Dijk FN, de Jongste JC, Smit HA, Brunekreef B, Postma DS, Van Steen K. Association of IL33–IL-1 receptor–like 1 (IL1RL1) pathway polymorphisms with wheezing phenotypes and asthma in childhood. J Allergy Clin Immunol. 2014;134:170–7.

    Article  CAS  PubMed  Google Scholar 

  69. Kondo Y, Yoshimoto T, Yasuda K, Futatsugi-Yumikura S, Morimoto M, Hayashi N, Hoshino T, Fujimoto J, Nakanishi K. Administration of IL-33 induces airway hyperresponsiveness and goblet cell hyperplasia in the lungs in the absence of adaptive immune system. Int Immunol. 2008;20:791–800.

    Article  CAS  PubMed  Google Scholar 

  70. Hardman C, Ogg G. Interleukin-33, friend and foe in type-2 immune responses. Curr Opin Immunol. 2016;42:16–24.

    Article  CAS  PubMed  Google Scholar 

  71. Schneider E, Petit-Bertron A-F, Bricard R, Levasseur M, Ramadan A, Girard J-P, Herbelin A, Dy M. IL-33 activates unprimed murine basophils directly in vitro and induces their in vivo expansion indirectly by promoting hematopoietic growth factor production. J Immunol. 2009;183:3591–7.

    Article  CAS  PubMed  Google Scholar 

  72. Bleck B, Kazeros A, Bakal K, Garcia-Medina L, Adams A, Liu M, Lee RA, Doris BT, Chiu A, Grunig G. Coexpression of type 2 immune targets in sputum-derived epithelial and dendritic cells from asthmatic subjects. J Allergy Clin Immunol. 2015;136:619–27. e615.

    Article  CAS  PubMed  Google Scholar 

  73. Oboki K, Ohno T, Kajiwara N, Arae K, Morita H, Ishii A, Nambu A, Abe T, Kiyonari H, Matsumoto K. IL-33 is a crucial amplifier of innate rather than acquired immunity. Proc Natl Acad Sci. 2010;107:18581–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Préfontaine D, Nadigel J, Chouiali F, Audusseau S, Semlali A, Chakir J, Martin JG, Hamid Q. Increased IL-33 expression by epithelial cells in bronchial asthma. J Allergy Clin Immunol. 2010;125:752–4.

    Article  PubMed  Google Scholar 

  75. Savinko T, Matikainen S, Saarialho-Kere U, Lehto M, Wang G, Lehtimäki S, Karisola P, Reunala T, Wolff H, Lauerma A. IL-33 and ST2 in atopic dermatitis: expression profiles and modulation by triggering factors. J Invest Dermatology. 2012;132:1392–400.

    Article  CAS  Google Scholar 

  76. Kakkar R, Lee RT. The IL-33/ST2 pathway: therapeutic target and novel biomarker. Nat Rev Drug Discovery. 2008;7:827–40.

    Article  CAS  PubMed  Google Scholar 

  77. Agache I, Lau S, Akdis CA, Smolinska S, Bonini M, Cavkaytar O, Flood B, Gajdanowicz P, Izuhara K, Kalayci O. EAACI guidelines on Allergen Immunotherapy: House dust mite-driven allergic asthma. Allergy. 2019;74:855–73.

    Article  CAS  PubMed  Google Scholar 

  78. Fahy JV. Type 2 inflammation in asthma—present in most, absent in many. Nat Rev Immunol. 2015;15:57–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lotvall J, Akdis CA, Bacharier LB, Bjermer L, Casale TB, Custovic A, Lemanske RF Jr, Wardlaw AJ, Wenzel SE, Greenberger PA. Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. J Allergy Clin Immunol. 2011;127:355–60.

    Article  PubMed  Google Scholar 

  80. Huang Y, Li X, Zhu L, Huang C, Chen W, Ling Z, Zhu S, Feng X, Yi C, Gu W. Thrombin cleaves IL-33 and modulates IL‐33‐activated allergic lung inflammation. Allergy. 2022;77:2104–20.

    Article  CAS  PubMed  Google Scholar 

  81. Pasha MA, Patel G, Hopp R, Yang Q. (2019) Role of innate lymphoid cells in allergic diseasesAllergy and asthma proceedings OceanSide Publications, p. 138.

  82. Lund S, Walford H, A Doherty H T. Type 2 innate lymphoid cells in allergic disease. Curr Immunol Rev. 2013;9:214–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Siroux V, Ballardini N, Soler M, Lupinek C, Boudier A, Pin I, Just J, Nadif R, Anto J, Melen E. The asthma-rhinitis multimorbidity is associated with IgE polysensitization in adolescents and adults. Allergy. 2018;73:1447–58.

    Article  CAS  PubMed  Google Scholar 

  84. Liew FY, Pitman NI, McInnes IB. Disease-associated functions of IL-33: the new kid in the IL-1 family. Nat Rev Immunol. 2010;10:103–10.

    Article  CAS  PubMed  Google Scholar 

  85. Schröder A, Lunding LP, Zissler UM, Vock C, Webering S, Ehlers JC, Orinska Z, Chaker A, Schmidt-Weber CB, Lang NJ. IL‐37 regulates allergic inflammation by counterbalancing pro‐inflammatory IL‐1 and IL‐33. Allergy. 2022;77:856–69.

    Article  PubMed  Google Scholar 

  86. Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P, Jonsdottir GM, Thorleifsson G, Helgadottir H, Steinthorsdottir V, Stefansson H. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009;41:342–7.

    Article  CAS  PubMed  Google Scholar 

  87. Gordon ED, Palandra J, Wesolowska-Andersen A, Ringel L, Rios CL, Lachowicz-Scroggins ME, Sharp LZ, Everman JL, MacLeod HJ, Lee JW. IL1RL1 asthma risk variants regulate airway type 2 inflammation. JCI insight 1. 2016.

  88. Vocca L, Di Sano C, Uasuf CG, Sala A, Riccobono L, Gangemi S, Albano GD, Bonanno A, Gagliardo R, Profita M. IL-33/ST2 axis controls Th2/IL-31 and Th17 immune response in allergic airway diseases. Immunobiology. 2015;220:954–63.

    Article  CAS  PubMed  Google Scholar 

  89. Li Y, Wang W, Lv Z, Li Y, Chen Y, Huang K, Corrigan CJ, Ying S. Elevated expression of IL-33 and TSLP in the airways of human asthmatics in vivo: a potential biomarker of severe refractory disease. J Immunol. 2018;200:2253–62.

    Article  CAS  PubMed  Google Scholar 

  90. Christianson CA, Goplen NP, Zafar I, Irvin C, Good JT Jr, Rollins DR, Gorentla B, Liu W, Gorska MM, Chu H. Persistence of asthma requires multiple feedback circuits involving type 2 innate lymphoid cells and IL-33. J Allergy Clin Immunol. 2015;136:59–68. e14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Gordon ED, Simpson LJ, Rios CL, Ringel L, Lachowicz-Scroggins ME, Peters MC, Wesolowska-Andersen A, Gonzalez JR, MacLeod HJ, Christian LS. (2016) Alternative splicing of interleukin-33 and type 2 inflammation in asthma. Proceedings of the National Academy of Sciences 113: 8765–8770.

  92. Suzukawa M, Koketsu R, Iikura M, Nakae S, Matsumoto K, Nagase H, Saito H, Matsushima K, Ohta K, Yamamoto K. Interleukin-33 enhances adhesion, CD11b expression and survival in human eosinophils. Lab Invest. 2008;88:1245–53.

    Article  CAS  PubMed  Google Scholar 

  93. van de Veen W, Akdis M. The use of biologics for immune modulation in allergic disease. J Clin Investig. 2019;129:1452–62.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Nnane I, Frederick B, Yao Z, Raible D, Shu C, Badorrek P, van den Boer M, Branigan P, Duffy K, Baribaud F. The first-in‐human study of CNTO 7160, an anti‐interleukin‐33 receptor monoclonal antibody, in healthy subjects and patients with asthma or atopic dermatitis. Br J Clin Pharmacol. 2020;86:2507–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Okragly AJ, Corwin KB, Elia M, He D, Schroeder O, Zhang Q, Shiyanova T, Bright S, Dicker SB, Chlewicki L. Generation and characterization of torudokimab (LY3375880): a monoclonal antibody that neutralizes interleukin-33. J Inflamm Res. 2021;14:3823.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Augé J, Vent J, Agache I, Airaksinen L, Campo Mozo P, Chaker A, Cingi C, Durham S, Fokkens W, Gevaert P. EAACI position paper on the standardization of nasal allergen challenges. Allergy. 2018;73:1597–608.

    Article  PubMed  Google Scholar 

  97. Bousquet J, Arnavielhe S, Bedbrook A, Fonseca J, Morais Almeida M, Todo Bom A, Annesi-Maesano I, Caimmi D, Demoly P, Devillier P. The allergic Rhinitis and its impact on Asthma (ARIA) score of allergic rhinitis using mobile technology correlates with quality of life: the MASK study. Allergy. 2018;73:505–10.

    Article  CAS  PubMed  Google Scholar 

  98. Wheatley LM, Togias A. Allergic rhinitis. N Engl J Med. 2015;372:456–63.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Small P, Keith PK, Kim H. Allergic rhinitis. Allergy Asthma Clin Immunol. 2018;14:1–11.

    Article  Google Scholar 

  100. Sakashita M, Yoshimoto T, Hirota T, Harada M, Okubo K, Osawa Y, Fujieda S, Nakamura Y, Yasuda K, Nakanishi K. Association of serum interleukin-33 level and the interleukin‐33 genetic variant with Japanese cedar pollinosis. Clin Experimental Allergy. 2008;38:1875–81.

    Article  CAS  Google Scholar 

  101. Mjösberg JM, Trifari S, Crellin NK, Peters CP, Van Drunen CM, Piet B, Fokkens WJ, Cupedo T, Spits H. Human IL-25-and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol. 2011;12:1055–62.

    Article  PubMed  Google Scholar 

  102. Akasaki S, Matsushita K, Kato Y, Fukuoka A, Iwasaki N, Nakahira M, Fujieda S, Yasuda K, Yoshimoto T. Murine allergic rhinitis and nasal T h2 activation are mediated via TSLP-and IL-33-signaling pathways. Int Immunol. 2016;28:65–76.

    Article  CAS  PubMed  Google Scholar 

  103. Haenuki Y, Matsushita K, Futatsugi-Yumikura S, Ishii KJ, Kawagoe T, Imoto Y, Fujieda S, Yasuda M, Hisa Y, Akira S. A critical role of IL-33 in experimental allergic rhinitis. J Allergy Clin Immunol. 2012;130:184–94. e111.

    Article  CAS  PubMed  Google Scholar 

  104. Kamekura R, Kojima T, Takano K, Go M, Sawada N, Himi T. The role of IL-33 and its receptor ST 2 in human nasal epithelium with allergic rhinitis. Clin Experimental Allergy. 2012;42:218–28.

    Article  CAS  Google Scholar 

  105. Rondon C, Campo P, Eguiluz-Gracia I, Plaza C, Bogas G, Galindo P, Mayorga C, Torres M. Local allergic rhinitis is an independent rhinitis phenotype: the results of a 10‐year follow‐up study. Allergy. 2018;73:470–8.

    Article  CAS  PubMed  Google Scholar 

  106. Shaw JL, Fakhri S, Citardi MJ, Porter PC, Corry DB, Kheradmand F, Liu Y-J, Luong A. IL-33–responsive innate lymphoid cells are an important source of IL-13 in chronic rhinosinusitis with nasal polyps. Am J Respir Crit Care Med. 2013;188:432–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Lam EP, Kariyawasam HH, Rana BM, Durham SR, McKenzie AN, Powell N, Orban N, Lennartz-Walker M, Hopkins C, Ying S. IL-25/IL-33–responsive TH2 cells characterize nasal polyps with a default TH17 signature in nasal mucosa. J Allergy Clin Immunol. 2016;137:1514–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Du Y, Luo Y, Yang C, Liu J, Wan J, Wang K. Discussion IL-33 and its receptor ST2 associated with the pathogenesis of allergic rhinitis. Lin Chuang Er Bi Yan Hou Tou Jing Wai Ke Za Zhi = Journal of Clinical Otorhinolaryngology. Head Neck Surg. 2015;29:811–4.

    Google Scholar 

  109. Kim Y, Yang T, Park CS, Ahn SH, Son B, Kim J, Lim D, Jang T. Anti-IL‐33 antibody has a therapeutic effect in a murine model of allergic rhinitis. Allergy. 2012;67:183–90.

    Article  CAS  PubMed  Google Scholar 

  110. Kitano M, Fukuoka S, Adachi N, Hisamitsu T, Sunagawa M. (2022) Shoseiryuto Ameliorated TDI-Induced Allergic Rhinitis by Suppressing IL-33 Release from Nasal Epithelial Cells. Pharmaceutics 14: 2083.

  111. Renz H, Allen KJ, Sicherer SH, Sampson HA, Lack G, Beyer K, Oettgen HC. Food allergy. Nat Reviews Disease Primers. 2018;4:1–20.

    Google Scholar 

  112. Burks AW, Tang M, Sicherer S, Muraro A, Eigenmann PA, Ebisawa M, Fiocchi A, Chiang W, Beyer K, Wood R. ICON: food allergy. J Allergy Clin Immunol. 2012;129:906–20.

    Article  PubMed  Google Scholar 

  113. Gupta RS, Warren CM, Smith BM, Jiang J, Blumenstock JA, Davis MM, Schleimer RP, Nadeau KC. Prevalence and severity of food allergies among US adults. JAMA Netw open. 2019;2:e185630–185630.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Turnbull J, Adams H, Gorard DA. The diagnosis and management of food allergy and food intolerances. Aliment Pharmacol Ther. 2015;41:3–25.

    Article  CAS  PubMed  Google Scholar 

  115. Żukiewicz-Sobczak WA, Wróblewska P, Adamczuk P, Kopczyński P. Causes, symptoms and prevention of food allergy. Adv Dermatology Allergology/Postępy Dermatologii i Alergologii. 2013;30:113–6.

    Article  Google Scholar 

  116. Castillo-Courtade L, Han S, Lee S, Mian F, Buck R, Forsythe P. Attenuation of food allergy symptoms following treatment with human milk oligosaccharides in a mouse model. Allergy. 2015;70:1091–102.

    Article  CAS  PubMed  Google Scholar 

  117. Liu J-Q, Hu T-Y, Diao K-Y, Yu D, Song Y-N, Mo L-H, Yang G, Liu Z-Q, Liu Z-G, Yang P-C. Cold stress promotes IL-33 expression in intestinal epithelial cells to facilitate food allergy development. Cytokine. 2020;136:155295.

    Article  CAS  PubMed  Google Scholar 

  118. Muto T, Fukuoka A, Kabashima K, Ziegler SF, Nakanishi K, Matsushita K, Yoshimoto T. The role of basophils and proallergic cytokines, TSLP and IL-33, in cutaneously sensitized food allergy. Int Immunol. 2014;26:539–49.

    Article  CAS  PubMed  Google Scholar 

  119. Travers J, Rochman M, Miracle CE, Cohen JP, Rothenberg ME. Linking impaired skin barrier function to esophageal allergic inflammation via IL-33. J Allergy Clin Immunol. 2016;138:1381–3.

    Article  PubMed  PubMed Central  Google Scholar 

  120. 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.

    Article  CAS  PubMed  Google Scholar 

  121. Rivas MN, Burton OT, Oettgen HC, Chatila T. IL-4 production by group 2 innate lymphoid cells promotes food allergy by blocking regulatory T-cell function. J Allergy Clin Immunol. 2016;138:801–11. e809.

    Article  PubMed Central  Google Scholar 

  122. Khodoun MV, Tomar S, Tocker JE, Wang YH, Finkelman FD. Prevention of food allergy development and suppression of established food allergy by neutralization of thymic stromal lymphopoietin, IL-25, and IL-33. J Allergy Clin Immunol. 2018;141:171–9. e171.

    Article  CAS  PubMed  Google Scholar 

  123. Chinthrajah S, Cao S, Liu C, Lyu S-C, 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 4. 2019.

  124. Torres T, Ferreira EO, Gonçalo M, Mendes-Bastos P, Selores M, Filipe P. Update on atopic dermatitis. Acta Med Port. 2019;32:606–13.

    Article  CAS  PubMed  Google Scholar 

  125. Bylund S, von Kobyletzki LB, Svalstedt M, Svensson Å. Prevalence and incidence of atopic dermatitis: a systematic review. Acta dermato-venereologica. 2020;100:320–9.

    Article  Google Scholar 

  126. Kowalska-Olędzka E, Czarnecka M, Baran A. Epidemiology of atopic dermatitis in Europe. J drug Assess. 2019;8:126–8.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Fuxench ZCC, Block JK, Boguniewicz M, Boyle J, Fonacier L, Gelfand JM, Grayson MH, Margolis DJ, Mitchell L, Silverberg JI. Atopic dermatitis in America study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. J Invest Dermatology. 2019;139:583–90.

    Article  Google Scholar 

  128. Brandt EB, Sivaprasad U. (2011) Th2 cytokines and atopic dermatitis. J Clin & cellular immunology 2 (3)

  129. Frazier W, Bhardwaj N. Atopic dermatitis: diagnosis and treatment. Am Family Phys. 2020;101:590–8.

    Google Scholar 

  130. Seltmann J, Werfel T, Wittmann M. Evidence for a regulatory loop between IFN-γ and IL‐33 in skin inflammation. Exp Dermatol. 2013;22:102–7.

    Article  CAS  PubMed  Google Scholar 

  131. Cevikbas F, Steinhoff M. IL-33: a novel danger signal system in atopic dermatitis. J Invest Dermatology. 2012;132:1326–9.

    Article  CAS  Google Scholar 

  132. Imai Y. Interleukin-33 in atopic dermatitis. J Dermatol Sci. 2019;96:2–7.

    Article  CAS  PubMed  Google Scholar 

  133. Kostner L, Anzengruber F, Guillod C, Recher M, Schmid-Grendelmeier P, Navarini AA. Allergic contact dermatitis. Immunol Allergy Clin. 2017;37:141–52.

    Google Scholar 

  134. Meephansan J, Tsuda H, Komine M, Tominaga S-i, Ohtsuki M. Regulation of IL-33 expression by IFN-γ and tumor necrosis factor-α in normal human epidermal keratinocytes. J Invest Dermatology. 2012;132:2593–600.

    Article  CAS  Google Scholar 

  135. Shimizu M, Matsuda A, Yanagisawa K, Hirota T, Akahoshi M, Inomata N, Ebe K, Tanaka K, Sugiura H, Nakashima K. Functional SNPs in the distal promoter of the ST2 gene are associated with atopic dermatitis. Hum Mol Genet. 2005;14:2919–27.

    Article  CAS  PubMed  Google Scholar 

  136. Peng G, Mu Z, Cui L, Liu P, Wang Y, Wu W, Han X. Anti-IL-33 antibody has a therapeutic effect in an atopic dermatitis murine model induced by 2, 4-dinitrochlorobenzene. Inflammation. 2018;41:154–63.

    Article  CAS  PubMed  Google Scholar 

  137. Bielory B, Bielory L. Atopic dermatitis and keratoconjunctivitis. Immunol Allergy Clin. 2010;30:323–36.

    Google Scholar 

  138. Matsuda A, Okayama Y, Terai N, Yokoi N, Ebihara N, Tanioka H, Kawasaki S, Inatomi T, Katoh N, Ueda E. The role of interleukin-33 in chronic allergic conjunctivitis. Investig Ophthalmol Vis Sci. 2009;50:4646–52.

    Article  Google Scholar 

  139. Matsuba-Kitamura S, Yoshimoto T, Yasuda K, Futatsugi-Yumikura S, Taki Y, Muto T, Ikeda T, Mimura O, Nakanishi K. Contribution of IL-33 to induction and augmentation of experimental allergic conjunctivitis. Int Immunol. 2010;22:479–89.

    Article  CAS  PubMed  Google Scholar 

  140. Asada Y, Nakae S, Ishida W, Hori K, Sugita J, Sudo K, Fukuda K, Fukushima A, Suto H, Murakami A. Roles of epithelial cell–derived type 2–Initiating cytokines in experimental allergic conjunctivitis. Investig Ophthalmol Vis Sci. 2015;56:5194–202.

    Article  CAS  Google Scholar 

  141. Bracken SJ, Abraham S, MacLeod AS. Autoimmune theories of chronic spontaneous urticaria. Front Immunol. 2019;10:627.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Maurer M, Khan DA, Komi DEA, Kaplan AP. Biologics for the use in chronic spontaneous urticaria: when and which. J Allergy Clin Immunology: Pract. 2021;9:1067–78.

    CAS  Google Scholar 

  143. Kaplan A, Lebwohl M, Giménez-Arnau AM, Hide M, Armstrong AW, Maurer M. Chronic spontaneous urticaria: focus on pathophysiology to unlock treatment advances. Allergy. 2023;78:389–401.

    Article  CAS  PubMed  Google Scholar 

  144. Chaowattanapanit S, Choonhakarn C, Salao K, Winaikosol K, Julanon N, Wongjirattikarn R, Foocharoen C, Sompornrattanaphan M. Increased serum IL-31 levels in chronic spontaneous urticaria and psoriasis with pruritic symptoms. Heliyon 6. 2020.

  145. Kay A, Clark P, Maurer M, Ying S. Elevations in T-helper‐2‐initiating cytokines (interleukin‐33, interleukin‐25 and thymic stromal lymphopoietin) in lesional skin from chronic spontaneous (‘idiopathic’) urticaria. Br J Dermatol. 2015;172:1294–302.

    Article  CAS  PubMed  Google Scholar 

  146. Trier AM, Ver Heul A, Fredman A, Le V, Wang Z, Auyeung K, Meixiong J, Lovato P, Holtzman MJ, Wang F. IL-33 potentiates histaminergic itch. Journal of Allergy and Clinical Immunology; 2023.

  147. Pushparaj PN, Tay HK, H’ng SC, Pitman N, Xu D, McKenzie A, Liew FY, Melendez AJ. (2009) The cytokine interleukin-33 mediates anaphylactic shock. Proceedings of the National Academy of Sciences 106: 9773–9778.

  148. West PW, Bahri R, Garcia-Rodriguez KM, Sweetland G, Wileman G, Shah R, Montero A, Rapley L, Bulfone-Paus S. Interleukin-33 amplifies human mast cell activities induced by complement anaphylatoxins. Front Immunol. 2021;11:615236.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Komai-Koma M, Brombacher F, Pushparaj P, Arendse B, McSharry C, Alexander J, Chaudhuri R, Thomson N, McKenzie A, McInnes I. Interleukin‐33 amplifies I g E synthesis and triggers mast cell degranulation via interleukin‐4 in naïve mice. Allergy. 2012;67:1118–26.

    Article  CAS  PubMed  Google Scholar 

  150. Antunes MM, Coelho BSL, Vichi TM, Dos Santos EA, Gondim FKB, Diniz AB, Aguilar EC, Cara DC, Porto LCJ, de Castro IC. Oral supplementation with capsaicin reduces oxidative stress and IL-33 on a food allergy murine model. World Allergy Organ J. 2019;12:100045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Kim E, Kim J, Samivel R, Bae JS, Chung YJ, Chung PS, Lee S, Mo JH. Intralymphatic treatment of flagellin–ovalbumin mixture reduced allergic inflammation in murine model of allergic rhinitis. Allergy. 2016;71:629–39.

    Article  CAS  PubMed  Google Scholar 

  152. Li J, Zhang L, Chen X, Chen D, Hua X, Bian F, Deng R, Lu F, Li Z, Pflugfelder SC. Pollen/TLR4 innate immunity signaling initiates IL-33/ST2/Th2 pathways in allergic inflammation. Sci Rep. 2016;6:36150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Zhiguang X, Wei C, Steven R, Wei D, Wei Z, Rong M, Zhanguo L, Lianfeng Z. Over-expression of IL-33 leads to spontaneous pulmonary inflammation in mIL-33 transgenic mice. Immunol Lett. 2010;131:159–65.

    Article  PubMed  Google Scholar 

  154. Liu X, Li M, Wu Y, Zhou Y, Zeng L, Huang T. Anti-IL-33 antibody treatment inhibits airway inflammation in a murine model of allergic asthma. Biochem Biophys Res Commun. 2009;386:181–5.

    Article  CAS  PubMed  Google Scholar 

  155. Kearley J, Buckland KF, Mathie SA, Lloyd CM. Resolution of allergic inflammation and airway hyperreactivity is dependent upon disruption of the T1/ST2–IL-33 pathway. Am J Respir Crit Care Med. 2009;179:772–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Oshikawa K, Yanagisawa K, Tominaga S, Sugiyama Y. Expression and function of the ST2 gene in a murine model of allergic airway inflammation. Clin Experimental Allergy. 2002;32:1520–6.

    Article  CAS  Google Scholar 

  157. Allinne J, Scott G, Birchard D, Agrawal A, Ben L-H, Lim WK, Kim JH, Kamat V, Murphy A, Sleeman M. IL-33 blockade impacts mediators of persistence and exacerbation of chronic airway inflammationEur respiratory soc. 2018.

  158. Taniguchi K, Yamamoto S, Hitomi E, Inada Y, Suyama Y, Sugioka T, Hamasaki Y. Interleukin 33 is induced by tumor necrosis factor alpha and interferon gamma in keratinocytes and contributes to allergic contact dermatitis. J Investig Allergol Clin Immunol. 2013;23:428–34.

    CAS  PubMed  Google Scholar 

  159. Salamon P, Shefler I, Moshkovits I, Munitz A, Horwitz Klotzman D, Mekori Y, Hershko A. IL-33 and IgE stimulate mast cell production of IL‐2 and regulatory T cell expansion in allergic dermatitis. Clin Experimental Allergy. 2017;47:1409–16.

    Article  CAS  Google Scholar 

  160. Hayakawa H, Hayakawa M, Kume A, Tominaga S-i. Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation. J Biol Chem. 2007;282:26369–80.

    Article  CAS  PubMed  Google Scholar 

  161. Anderson E, Kobayashi T, Iijima K, Bartemes K, Chen CC, Kita H. IL-33 mediates reactive eosinophilopoiesis in response to airborne allergen exposure. Allergy. 2016;71:977–88.

    Article  CAS  PubMed  Google Scholar 

  162. Cherry WB, Yoon J, Bartemes KR, Iijima K, Kita H. A novel IL-1 family cytokine, IL-33, potently activates human eosinophils. J Allergy Clin Immunol. 2008;121:1484–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Stolarski B, Kurowska-Stolarska M, Kewin P, Xu D, Liew FY. IL-33 exacerbates eosinophil-mediated airway inflammation. J Immunol. 2010;185:3472–80.

    Article  CAS  PubMed  Google Scholar 

  164. Ali M, Zhang G, Thomas W, McLean C, Bizzintino J, Laing I, Martin A, Goldblatt J, Le Souëf P, Hayden C. Investigations into the role of ST2 in acute asthma in children. Tissue Antigens. 2009;73:206–12.

    Article  CAS  PubMed  Google Scholar 

  165. Gasiuniene E, Janulaityte I, Zemeckiene Z, Barkauskiene D, Sitkauskiene B. Elevated levels of interleukin-33 are associated with allergic and eosinophilic asthma. Scand J Immunol. 2019;89:e12724.

    Article  PubMed  Google Scholar 

  166. Schröder PC, Casaca VI, Illi S, Schieck M, Michel S, Böck A, Roduit C, Frei R, Lluis A, Genuneit J. IL-33 polymorphisms are associated with increased risk of hay fever and reduced regulatory T cells in a birth cohort. Pediatr Allergy Immunol. 2016;27:687–95.

    Article  PubMed  Google Scholar 

  167. Queiroz GA, Costa RdS, Alcantara-Neves N, Nunes de Oliveira Costa G, Barreto ML, Carneiro VL, Figueiredo C. IL33 and IL1RL1 variants are associated with asthma and atopy in a Brazilian population. Int J Immunogenet. 2017;44:51–61.

    Article  CAS  PubMed  Google Scholar 

  168. Riikonen R, Teräsjärvi J, Lauhkonen E, Nuolivirta K, He Q, Korppi M. Interleukin 1 receptor-like 1 rs13408661/13431828 polymorphism is associated with persistent post‐bronchiolitis asthma at school age. Acta Paediatr. 2022;111:628–35.

    Article  CAS  PubMed  Google Scholar 

  169. Nakanishi W, Yamaguchi S, Matsuda A, Suzukawa M, Shibui A, Nambu A, Kondo K, Suto H, Saito H, Matsumoto K. IL-33, but not IL-25, is crucial for the development of house dust mite antigen-induced allergic rhinitis. PLoS ONE. 2013;8:e78099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Londei M, Kenney B, Los G, Marino MH. A phase 1 study of ANB020, an anti-IL-33 monoclonal antibody in healthy volunteers. J Allergy Clin Immunol. 2017;139:AB73.

    Article  Google Scholar 

  171. Asaka D, Yoshikawa M, Nakayama T, Yoshimura T, Moriyama H, Otori N. Elevated levels of interleukin-33 in the nasal secretions of patients with allergic rhinitis. Int Arch Allergy Immunol. 2012;158:47–50.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors extend their appreciation to the Deanship of Postgraduate Studies and Scientific Research at Majmaah University for funding this research work through the project number (R- 2025-1628). Dr. Fuzail Ahmad would like to thank Almareefa University.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

Mohammad Chand Jamali, Azfar Jamal and Mohammad Azhar Kamal: Methodology, Investigation, Writing–original draft. Waleed Al Abdulmonem, Bashar Abdullah Saeed, Nasrin Mansuri, and Fuzail Ahmad: Visualization, Writing– review & editing. Ayyub Ali Patel and Alaa Shafie: Writing - original draft. Asma’a H. Mohamed: Project administration, Supervision, Writing–original draft, and Writing - review & editing. Haroonrashid M. Hattiwale: Project administration, Supervision, and Writing - review & editing.

Corresponding authors

Correspondence to Asma’a H. Mohamed or Haroonrashid M. Hattiwale.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jamali, M.C., Mohamed, A.H., Jamal, A. et al. Biological mechanisms and therapeutic prospects of interleukin-33 in pathogenesis and treatment of allergic disease. J Inflamm 22, 17 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12950-025-00438-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12950-025-00438-w

Keywords