Skip to main content

Decoding innate lymphoid cells and innate-like lymphocytes in asthma: pathways to mechanisms and therapies

Abstract

Asthma is a chronic inflammatory lung disease driven by a complex interplay between innate and adaptive immune components. Among these, innate lymphoid cells (ILCs) and innate-like lymphocytes have emerged as crucial players in shaping the disease phenotype. Within the ILC family, group 2 ILCs (ILC2s), in particular, contribute significantly to type 2 inflammation through their rapid production of cytokines such as IL-5 and IL-13, promoting airway eosinophilia and airway hyperreactivity. On the other hand, innate-like lymphocytes such as invariant natural killer T (iNKT) cells can play either pathogenic or protective roles in asthma, depending on the stimuli and lung microenvironment. Regulatory mechanisms, including cytokine signaling, metabolic and dietary cues, and interactions with other immune cells, play critical roles in modulating their functions. In this review, we highlight current findings on the role of ILCs and innate-like lymphocytes in asthma development and pathogenesis. We also examine the underlying mechanisms regulating their function and their interplay with other immune cells. Finally, we explore current therapies targeting these cells and their effector cytokines for asthma management.

Introduction

Asthma is a chronic respiratory disease affecting more than 300 million people of all ages worldwide [1]. The hallmark features of asthma include inflammation and airway constriction, causing symptoms like coughing, wheezing, chest tightness, and dyspnea. Inhaled corticosteroids (ICS) remain the mainstay therapy for asthma management, and can be administered individually or in combination with a controller medication such as long-acting beta agonists (LABA). Nevertheless, approximately 5–10% of patients are refractory to this conventional treatment, experiencing poor symptom control and frequent exacerbations [2]. These patients are categorized as having severe asthma and they account for a significant portion of asthma-related morbidity and healthcare expenditures [3].

In general, asthma can be classified into two endotypes: type 2 (T2)-high and T2-low/non-T2. T2-high asthma is defined by eosinophilic inflammation and elevated levels of T helper 2 (Th2) cytokines, mainly IL-4, IL-5, and IL-13 [4]. It can present as either an allergic or non-allergic phenotype and accounts for 50% of mild-to-moderate asthma, as well as a significant proportion of patients with severe asthma [5, 6]. T2-low asthma, on the other hand, is defined by the absence of T2-high features and can be further classified as either type 17 (T17)-high asthma, marked by neutrophilic inflammation and high circulating levels of IL-17 and IL-22, or T2-low and T17-low asthma [7]. This endotype is often severe and patients tend to be refractory to high doses of ICS [8].

Although initially thought to be mediated by the adaptive immune responses of B and T effector cells, recent studies have underscored the pivotal roles of innate lymphoid cells (ILCs) and innate-like lymphocytes in shaping the adaptive immunity and influencing disease outcomes in asthma. ILCs are non-B, non-T cells that lack antigen-specific receptors and exhibit phenotypes and functions similar to those of T cells. They are divided into five distinct subsets: natural killer (NK) cells, ILC1, ILC2, ILC3, and lymphoid tissue inducer cells (LTi) [9]. Among the ILCs, ILC2s are the most extensively studied and are particularly notable for their role in the pathogenesis of asthma. Innate-like lymphocytes include γδ T cells, invariant natural killer T (iNKT) cells, and mucosal-associated invariant T (MAIT) cells. These unconventional T cells are enriched in mucosal tissues such as the lungs and liver and respond rapidly to stress by secreting copious amounts of cytokines in a T cell antigen receptor (TCR)-dependent or independent manner [10]. In asthma, the role of innate-like lymphocytes remains controversial, with some studies reporting a protective function while others demonstrate a pathogenic effect.

In this review, we detail key characteristics of ILCs and innate-like lymphocytes, focusing primarily on ILC2s and iNKT cells, and examine their roles in the development of, or protection against, asthma in both human and mouse models. We explore the cellular and environmental triggers that influence the functions of these cells and highlight current existing therapies that could potentially be employed to target them.

Characteristics of ILCs and innate-like lymphocytes

ILCs

NK cells and ILC1s

NK cells and ILC1s are categorized as group 1 ILCs due to their shared ability to produce type 1 cytokines like interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), as well as their dependence on the T-box transcription factor (T-bet) for lineage specification [11]. NK cells, identified as CD3 CD56+ cells in humans and CD3 NK1.1+ CD49b+ Nkp46+ cells in mice [12], make up 10–15% of lung lymphocytes [13]. Unlike ILC1s, the development of NK cells requires the transcription factor EOMES [14]. NK cells are highly cytotoxic and play a key role in eliminating cancer and virus-infected cells through the secretion of perforins and granzymes [15]. They recognize altered cell surface proteins via activating receptors like NKG2D and natural cytotoxicity receptors (NCRs), while inhibitory receptors such as NKG2A and killer immunoglobulin-like receptors (KIRs) modulate their activity [12, 16].

ILC1s, in contrast, are tissue-resident and respond to cytokines like IL-15, IL-12, and IL-18, particularly in response to intracellular pathogens [17]. Predominantly found in the intestinal intraepithelial compartment, ILC1s account for 25–50% of the total ILCs [18]. In humans, intraepithelial ILC1s have been described in gut and tonsil [19]. ILC1s express surface molecules that facilitate tissue retention, such as CD103, CD49a, and CD69 [20], and are crucial for defense against intracellular pathogens such as the parasite Toxoplasma gondii. They provide innate protection by producing IFN-γ to control the parasite burden [21]. ILC1s are also implicated in both cancer progression and protective antitumor responses, depending on the tumor type [22].

ILC2s

Identified in 2010 in the adipose tissue and small intestine of mice [23, 24], ILC2s are regarded as the innate counterparts of Th2 cells, requiring GATA3 for differentiation, maintenance, and function [25], and produce Th2-associated cytokines IL-13 and IL-5 [23]. In mice, ILC2s are present in the lungs, small intestine, adipose tissue, and skin, each exhibiting tissue-specific properties [26]. Lung ILC2s, or natural ILC2s, respond to IL-33 and express the IL-33 receptor ST2 [27]. In contrast, gut ILC2s become pro-inflammatory upon activation by IL-25 and are characterized by the expression of IL-25R and KLRG1 [28]. These gut ILC2s can migrate to the lungs during Nippostrongylus brasiliensis infection through sphingosine 1-phosphate (S1P) but will eventually revert to the natural ILC2 phenotype [28, 29]. While adipose tissue ILC2s are phenotypically more similar to lung ILC2s, dermal ILC2s express thymic stromal lymphopoietin (TSLP) receptors and are activated by IL-18 [26]. In humans, ILC2s are the predominant ILC population in the skin and can also be found in the blood, lungs, and tonsils, though they are rare in the small intestine [30, 31]. ILC2s have been shown to exhibit a memory phenotype upon exposure to allergens [32]. They also exhibit high functional plasticity, and can polarize to either ILC1- or ILC3-like cells upon stimulation with the respective activating cytokines [33,34,35,36]. ILC2s are associated with the development of many allergic diseases including atopic dermatitis, allergic rhinitis, and asthma [37, 38]. Nevertheless, they also have beneficial roles, such as promoting thermogenesis [39], aiding tissue repair through the production of amphiregulin [27], facilitating the expulsion of helminths [40], and enhancing antitumor immunity [41, 42].

ILC3s

ILC3s are the predominant ILC subset in the human lung and are also found in the intestine in both mice and humans [43,44,45]. They are a diverse population with at least four identified subsets: IL-17-producing Nkp46 RORγt+ ILC3s [46], IL-22-producing Nkp46+ RORγt+ ILC3s [44], Nkp46+ RORγt ILC3s (‘ex-RORγt+’ ILC3s) [21], and LTi-like ILC3s [47]. The surface markers of ILC3s are highly heterogeneous, varying according to the specific subset and the tissue in which they reside [9]. In the lung, Cd36 has been identified as a lung-specific marker for ILC3 [48]. ILC3s require the transcription factor retinoic acid receptor-related orphan receptor γt (RORγt) for development and produce IL-17A, IL-22, and granulocyte–macrophage colony-stimulating factor (GM-CSF) in response to IL-1β and IL-23 [49]. Notably, ILC3s exhibit phenotypic plasticity and can acquire an ILC1-like phenotype, a process regulated by the transcription factor c-Maf [50, 51]. Similar to ILC2s, lung ILC3s can acquire a memory-like phenotype following exposure to cigarette smoke and contribute to asthma exacerbation [52].

Innate-like lymphocytes

iNKT cells

NKT cells are divided into type I and II based on differences in TCR usage, with iNKT cells belonging to the type I subset [53]. iNKT cells have a restricted TCRα chain (Vα14-Jα18 in mice and Vα24-Jα18 in humans), which mainly recognizes self-antigens and foreign lipid antigens presented by CD1d on antigen-presenting cells [54, 55]. They share common phenotypic and functional features with the conventional T cells and NK cells [56], and can express either CD4 or exhibit a double-negative (CD4 CD8) phenotype in both humans and mice [56]. There are three main iNKT cell subtypes: NKT1, NKT2, and NKT17, which primarily produce IFN-γ, IL-4, and IL-17A, respectively [57]. This cytokine profile enables iNKT cells to either suppress or amplify allergic responses. For example, iNKT cells activated by α-GalCer prior to ovalbumin (OVA) challenge have been shown to exert a suppressive effect, whereas iNKT cells activated by house dust mite (HDM) extracts during the OVA sensitization period enhance the adjuvant effects of HDM extracts, thereby amplifying inflammation [58, 59]. Their cytokine profile, along with their cognate interactions, orchestrates the functions of various immune cells, making them attractive targets for therapeutic intervention.

γδ T cells

γδ T cells are a specialized subset of T cells that express a γδ TCR instead of the conventional αβ TCR. They are classified by the TCR V gene usage, with various γ (γ2–5, γ8, γ9, and γ11) and δ (δ1–3) genes used for classification in humans, while distinct γ chains define subsets in mice [60, 61]. γδ T cells make up approximately 10% of total T cells in human peripheral blood mononuclear cells (PBMCs) but can be expanded at mucosal barriers and in peripheral non-lymphoid organs [62]. These cells recognize endogenous soluble and non-protein antigens, such as stress-induced molecules (MICA/B) and phospho-antigens, without requiring antigen processing and major histocompatibility complex (MHC) presentation [63]. Two major subsets have been defined in mice based on cytokine production: IL-17- and IFN-γ-producing subsets [64]. Their role in asthma is complex, as studies suggest they can have both protective and pathogenic effects in allergen-induced asthma [65, 66]. Specifically, γδ T cells promote allergic airway inflammation during a short-term challenge (4-day) with cockroach extract [66], but reverse airway hyperreactivity (AHR) after a long-term challenge (52-day) with OVA [65].

MAIT cells

MAIT cells express the MHC class I-related molecule MR1 and detect microbial metabolites derived from vitamin B9 or B2 (riboflavin) [67]. These metabolites, produced by bacteria and fungi capable of synthesizing riboflavin, enable MAIT cells to detect microbial infections [68]. MAIT cells feature a semi-invariant TCR, comprising a conserved Vα chain (Vα7.2 in humans and Vα19 in mice) paired with a limited range of Vβ chains (Vβ2/Vβ13 in humans and Vβ6/Vβ8 in mice) [69]. MAIT cells can respond to a variety of inflammatory stimuli, including IL-7, IL-12, IL-15, IL-18, and IL-23, through the expression of their corresponding cytokine receptors [62]. Upon activation, they rapidly produce cytokines such as IFN-γ, TNF-α, and IL-17 and cytotoxic substances such as perforin and granzyme, contributing to antimicrobial defense and inflammation [69]. MAIT cells have been implicated in several lung diseases, including asthma, chronic obstructive pulmonary disease (COPD), pneumonia, tuberculosis, and lung cancer [70, 71].

ILCs and innate-like lymphocytes in asthma

Extensive evidence highlights the diverse roles of ILCs and innate-like lymphocytes in the pathogenesis of asthma across various inflammatory phenotypes [72, 73]. The importance of ILCs and innate-like lymphocytes in asthma development has been well-established in both asthmatic murine models [74, 75] and patients with asthma [72, 76].

ILCs

Group 1 ILCs, including ILC1s and NK cells, exhibit complex roles in asthma. ILC1s are reduced in the blood of patients with allergic and non-allergic eosinophilic asthma [77], whereas their levels are elevated in the sputum of asthma patients with neutrophilic inflammation, where they are linked to inflammasome activation, IFN-γ, and TNF signaling [78]. ILC1s, together with ILC3s, induce M1 macrophage polarization through IFN-γ and IL-17A, and contribute to asthma pathogenesis [79]. NK cells, on the other hand, exhibit elevated frequencies and cytotoxic activity in both asthmatic adults and children, with distinct cytokine profiles observed, including increased IL-4 and reduced IFN-γ production [80,81,82,83]. In mice, NK cells exhibit dual roles: they promote eosinophilic inflammation and enhance Th2 response in allergic asthma models such as OVA and HDM [84, 85], but also provide protection against respiratory syncytial virus (RSV)-induced allergic airway inflammation through IFN-γ production [86]. In humans, NK cells are highly activated in severe asthma and help down-modulate the inflammatory response by inducing eosinophil apoptosis [87]. Collectively, these findings highlight the ambiguous roles of NK cells in asthma, highlighting the need for further investigation.

ILC2s are increased in the peripheral blood of asthma patients compared to healthy individuals or those with allergic rhinitis [88, 89]. Sputum analysis also revealed that allergen exposure increases ILC2 numbers in asthma patients [90]. Clinical studies highlighted a correlation between the alarmin cytokine IL-33 and ILC2s in peripheral blood [91] and bronchoalveolar lavage fluid (BALF) [92], influencing allergic asthma symptoms. Animal studies using asthma models (RSV, OVA, HDM, or fungi) demonstrated that epithelial alarmin-ILC2 interactions drive eosinophil infiltration, AHR, and mucus production [93,94,95,96,97,98]. Recent studies have revealed the diversity of human circulating ILC2s. Initially, human ILC2s were identified by their expression of CD127 (IL-7 receptor α-subunit), CD161, and CRTH2 (prostaglandin D2 receptor) [31]. However, subsequent research has identified some ILC2-like cells in human peripheral blood that lack CRTH2 expression but instead express KLRG1 [99,100,101]. Maho et al. further demonstrated that KLRG1+ ILCs can differentiate into other ILC subsets [100]. CRTH2 ILC2s are enriched in the blood and BALF of patients with asthma [101], and another study discovered that the dynamic regulation of CRTH2 expression might be linked to the migration of human ILC2s into lung tissues. In mice, the accumulation of ILC2s and development of type 2 inflammation in the lung depend on CRTH2 [99]. Other ILC2 subsets, such as CCR10+ ILC2s, have been found to be enriched in the blood of both allergic and non-allergic severe asthmatic patients. CCR10+ ILC2s secrete minimal Th2 cytokines but possess ILC1-like features, including IFN-γ production [102]. In an allergen-induced asthma model, depletion of CCR10+ ILC2s resulted in the exacerbation of AHR [102]. These findings indicate the protective role of CCR10+ ILC2s in allergic inflammation.

Studies on the role of ILC3s in asthma remain limited. Notably, IL-17 has been reported to play a pertinent role in the pathogenesis of severe asthma and is associated with the recruitment and activation of neutrophils in the airways [103]. Human studies have also shown that patients with severe asthma exhibit higher numbers of IL-17-producing ILC3s in BALF compared to individuals with mild asthma or those without asthma [104]. Based on these observations, Kim et al. further demonstrated that high fat diet (HFD) up-regulated IL-1β expression, which potently stimulates and increases ILC3s in obese lungs. Furthermore, IL-17-producing ILC3s were found to be essential for the development of obesity-related AHR [104]. Collectively, these studies highlight the potential role of the ILC3-IL-17 axis in the pathogenesis of asthma.

Innate-like lymphocytes

The role of iNKT cells in asthma has been extensively studied in various murine models, including those induced by allergens [105, 106], ozone [107, 108], and viruses [109]. Clinically, iNKT cells have been found in lung [110, 111], sputum [112], BALF [113], and sinus mucosa [114] samples from patients with asthma. However, significant variability exists in the reported numbers of iNKT cells in asthmatic patients. While some studies have observed an increase in iNKT cells, others have reported a decrease compared to healthy individuals. This variability likely reflects differences in patient populations, tissues analyzed, and detection methods. Several novel cytokines have been found to modulate iNKT cells, thereby contributing to the development of asthma. Notably, blood iNKT cells in asthmatic patients exhibit a Th2-like phenotype, with IL-4-producing iNKT cells potentially playing a critical role in modulating lung function [115]. Cytokines such as IL-25, IL-33 and TSLP activate iNKT cells, contributing to the development of AHR [116,117,118]. While murine models highlight the role of iNKT subsets in asthma, their relevance to human disease warrants further study.

Reports on the frequency of γδ T cells in asthma patients show varying results. Early studies demonstrated a decreased percentage of blood γδ T cells in asthmatic patients [119, 120]. In contrast, another study found no significant difference in γδ T cell levels between asthmatic patients and healthy controls across peripheral blood, induced sputum, or BALF [121]. γδ T cells are known to produce cytokines that contribute to the pathogenesis of neutrophilic asthma, such as IFN-γ and IL-17 [122, 123]. In allergic asthma patients, an increased frequency of IL-4-producing γδ T cells and a reduced frequency of IFN-γ-producing γδ T cells were observed compared to healthy controls [124]. Moreover, Krug et al. found an increased frequency of IL-5+ γδ T cells and IL-13+ γδ T cells in the BALF of asthmatic patients after segmental allergen challenge [125], suggesting a possible skew towards a Th2-type response and a potential involvement in allergic asthma. Overall, these studies underscore the complexity of the γδ T cell compartment in asthma and its potential significance in asthma pathogenesis.

Several studies have reported an association between MAIT cells and asthma. The frequency of MAIT cells is reduced in the peripheral blood, sputum, BALF, and lungs of asthmatic patients compared to healthy individuals [126,127,128,129]. In children with asthma, IL-17-producing MAIT cells have been observed to correlate with asthma severity [129]. Moreover, in severe asthma, the frequencies of MAIT cells, NK cells, ILC1s, ILC2s, and ILC3s show a positive correlation, indicating potential interactions between MAIT cells and these innate lymphoid cells [127]. In HDM and Alternaria alternata-induced mouse models, MAIT cells have been shown to regulate airway inflammation by suppressing ILC2 proliferation and activity [71]. Moreover, MAIT cells have also been found to protect against asthma by regulating the mucosal microbiota [130]. Collectively, the studies in both humans and mice reveal the involvement of MAIT cells in asthma pathogenesis.

ILC and innate-like lymphocyte regulation and immune crosstalk in asthma

Understanding the regulation of ILCs and innate-like lymphocytes is key to uncovering their role in driving asthma. Targeting these regulatory mechanisms offers potential for precise therapies to modulate inflammation and restore immune balance. This section examines how cytokines, non-cytokine mediators, and crosstalk with other effector cells influence the function of ILCs (Fig. 1) and innate-like lymphocytes (Fig. 2).

Fig. 1
figure 1

Modulation of ILC function in asthma. A ILC2 activation is influenced by cytokines and other mediators. IL-33, TSLP, and IL-25 are primary activators, while IL-2, IL-7, IL-9, IL-4, and TNF superfamily members act as co-factors. Meanwhile, type I/II IFNs, IL-27, and IL-10 inhibit ILC2s. Activation leads to GATA3 and NF-κB phosphorylation, driving IL-13 and IL-5 production, proliferation, and survival. IL-1β, IL-12, and IL-18 promote ILC1-like differentiation, marked by IFN-γ production and T-bet expression, while IL-1β, IL-23, and TGF-β drive polarization into IL-17A-producting ILC3-like cells. Notably, IL-4 counteracts these effects. Non-cytokine regulators, including neuropeptides (NMU, VIP, CGRP), prostaglandins (PGI2, PGE2, PGD2), and leukotrienes, can either inhibit or enhance ILC2 activation. Butyrate inhibits ILC2s via HDAC suppression, while BHB indirectly suppresses ILC2s by limiting IL-2 from mast cells. Tregs (ICOS-ICOSL) and SLAMF receptors inhibit ILC2s, while Pla2g5+ macrophages (FFAs, IL-33) activate ILC2s. ILC2s also influence CD4+ T cells via BTN2A2, PD-L1, and OX40L, promoting Th2 differentiation. Notably, GATA3-expressing Tregs inhibit OX40L expression on ILC2s. B Resolvin-E1 promotes NK cell migration and enhances cytotoxic abilities, while lipoxin A4 promotes eosinophil apoptosis and clearance, together boosting pro-resolving functions of NK cells. In contrast, cannabinoids and PGI2 inhibit IFN-γ production by NK cells, subsequently attenuating ILC2 function. Type I IFNs from pDCs drive NK cell IFN-γ production. NK cell pathogenicity involves TLR3-induced IL-17A production, worsening asthma. NKG2D+ NK cells, via MULT-1, elevate IgE and eosinophil levels, promoting allergic airway inflammation. C ILC3s drive neutrophilic inflammation via IL-17. Cigarette smoke induces IL-1β from airway epithelium, generating memory-like ILC3s that worsen neutrophilic asthma. Conversely, MHC-II engagement on ILC3s inhibits microbe-specific Th17 and allergen-specific Th2 cells, reducing neutrophilic and eosinophilic asthma, respectively. NCR+ ILC3s interact with the microbiome to enhance protective functions via an unknown mechanism

Fig. 2
figure 2

Regulation of the pathogenic or protective functions of innate-like T cells in asthma. A IL-4 secretion by iNKT cells promotes allergic inflammation, with progranulin enhancing IL-4 production by downregulating EZH2 expression, thereby facilitating PLZF translocation and subsequent IL-4 expression. CD40-CD40L interaction between iNKT cells and DCs promotes Th2 differentiation, contributing to allergic inflammation. ACC1 mediates de novo fatty acid synthesis by upregulating PPARγ and FABPs, which promotes iNKT cell survival and enhances their deleterious functions. The protective functions of iNKT cells have been demonstrated in a PM2.5 model, where phosphatidylserine-expressing apoptotic epithelial cells activate suppressive CD38+ CD4 iNKT cells, leading to the upregulation of PD-L1 expression through interaction with Tim-1. PD-L1/PD-1 interaction between CD4 iNKT cells and γδ T cells inhibits IL-17A production by γδ T cells, thereby reducing neutrophilic inflammation. CCL21 can recruit CCR7+ PLZF+ iNKT cells into inflamed lungs to restrain Th2 response and mitigate eosinophilic inflammation. B CXCR5+ γδ T cells in allergic inflammation exhibit a Th2 phenotype (producing IL-4 and IL-10), promoting antibody production. Notably, Itk suppresses their development. IL-17A-producing γδ T cells contribute to neutrophilic inflammation via IL-6 trans-signaling, where IL-6 forms a complex with soluble IL-6R (sIL-6R) and binds to the GP130 receptor, amplifying inflammation. C MAIT cells produce IFN-γ which suppresses ILC2 cytokine production, attenuating airway inflammation

ILCs

ILC2 proliferation and effector function can be regulated by various factors including cytokines, lipid mediators, neurotransmitters, dietary metabolites, and cellular crosstalk (Fig. 1A). Beyond the well-established role of epithelial-derived cytokines in activating ILC2s, additional co-stimulatory cytokines have been identified as key regulators of ILC2 activity. Both IL-7 and IL-2 work synergistically with IL-33 to increase ILC2 numbers and induce corticosteroid resistance in vitro [131], while IL-9 supports ILC2 proliferation and type 2 cytokine production [132]. Basophil-derived IL-4 promotes ILC2 effector function in papain-induced airway inflammation [133]. Furthermore, members of the TNF superfamily, TL1A and TNF-α, amplify ILC2-mediated airway inflammation by enhancing ILC2 survival, expansion, and cytokine production [134, 135]. Conversely, ILC2 activation and effector functions are directly counteracted by inhibitory cytokines such as type I and type II interferons. Our group has identified an indirect mechanism through which type I IFN suppresses lung ILC2 function by stimulating IFN-γ production by NK cells. Both type I and II IFNs inhibit ILC2 proliferation and type 2 cytokine production via the signal transducer and activator of transcription 1 (STAT1) signaling pathway [136]. Additionally, IL-27 directly inhibits ILC2 function through STAT1 [137], while IL-10, another anti-inflammatory cytokine, attenuates ILC2 activation and function in humans and mice [96, 138]. Interestingly, IL-10 specifically inhibits IL-33-mediated ILC2 activation but does not suppress ILC2 responses to IL-25 [96]. ILC2s also exhibit a high degree of plasticity, predominantly demonstrated in human systems, with cytokines driving their differentiation into ILC1- and ILC3-like cells, based on transcription factor expression and cytokine profiles [33, 139, 140]. Notably, this plasticity is inhibited by IL-4 [36]. Through this plasticity, ILC2s contribute to both allergic and non-allergic forms of asthma.

Non-cytokine factors also play a significant role in regulating ILC2 function. Earlier studies have highlighted the roles of leukotrienes, prostaglandins, and neuropeptides in modulating lung ILC2 activity. Elevated levels of leukotrienes have been detected in the sputum and plasma of individuals with asthma [141, 142], and preclinical studies have shown that leukotrienes synergize with IL-33 via the nuclear factor of activated T cells (NFAT) pathway to activate ILC2s during type 2 inflammation [143]. Conversely, prostaglandin I2 (PGI2) and prostaglandin E2 (PGE2) inhibit ILC2 function through the cyclic AMP (cAMP) pathway [144, 145], while prostaglandin D2 (PGD2) promotes ILC2 activity [146]. Neuroimmune regulation is equally crucial in modulating lung ILC2 function. Neuropeptides and neurotransmitters, such as vasoactive intestinal peptide (VIP) and neuromedin U, have been shown to activate ILC2s in mouse models of asthma [147, 148]. In contrast, calcitonin gene-related peptide (CGRP) and dopamine attenuate ILC2 effector functions [149, 150]. More recent studies have demonstrated the importance of calcium ion channels (Orai1) and iron in promoting ILC2 function by modulating ILC2 metabolism, leading to AHR and type 2 inflammation [151, 152]. Our group has particularly focused on metabolic factors, such as the short-chain fatty acid (SCFA) butyrate and the ketone body β-hydroxybutyrate (BHB). Butyrate, predominantly produced by the gut microbiome, directly inhibits ILC2 proliferation and type 2 cytokine production through inhibition of histone deacetylase (HDAC) signaling [153]. Although we did not identify the specific HDAC involved in this study, our recent findings in dermal ILC2s suggest that butyrate acts via HDAC3 [154]. A subsequent study further corroborated our findings, showing that butyrate inhibits ILC2-driven allergic airway inflammation by reducing IL-13 and IL-5 production while promoting IL-17A production by ILC2s [155]. Additionally, Karagiannis et al. reported that the ketogenic diet attenuates lung ILC2 function by impairing fatty acid metabolism [156]. BHB is one of the major ketone bodies produced during ketogenic diet feeding [157]. Building on this research, we demonstrated that BHB suppresses ILC2 function and mitigates allergic airway inflammation in the Alternaria alternata model [158]. Unlike butyrate, BHB does not act directly on ILC2 but instead targets mast cells, inhibiting IL-2 production via G protein-coupled receptor 109 A (GPR109A). Collectively, these studies underscore the potential impact of diet on asthma management.

ILC2s coordinate with various immune cells in a complex network of cellular crosstalk, influencing immune responses and contributing to asthma development. ILC2-derived IL-13 acts on dendritic cells (DCs) to potentiate Th2 effector cell generation [159]. ILC2s also regulate Th2 cell differentiation by expressing co-stimulatory molecules, such as OX40L and butyrophilin 2a2 (BTN2A2), as well as PD-L1, in response to epithelial-derived cytokines. BTN2A2 interaction with its receptor on T cells inhibits T cell effector function [160], while OX40L engagement with its receptor promotes Th2 cell expansion [161]. On the other hand, engagement of PD-L1 with PD-1 on T cells promotes GATA3 expression and IL-13 production by Th2 cells [162]. A recent study has shown that GATA3hi regulatory T cells (Tregs) negatively regulate the availability of OX40L on ILC2s, thereby limiting the expansion of effector memory Th2 cells and attenuating airway inflammation [163]. In addition, interactions between ILC2s and bystander T cells in the mediastinal lymph node through signaling lymphocyte activation molecule 3 (SLAMF3) and SLAMF5 suppress ILC2-derived IL-13 production, which in turn inhibits DC activation and Th2 differentiation in the papain model of allergic asthma [164]. ILC2 function is further modulated through interactions with other lung immune cells. Group V phospholipase A2 (Pla2g5) regulates IL-33 and free fatty acid (FFA) production by lung macrophages, which, in turn, stimulates ILC2 activation and expansion upon exposure to Alternaria alternata [165]. Eosinophils play a reciprocal role in promoting ILC2 accumulation in the lungs through IL-4 and IL-13 [166]. Additionally, Tregs can inhibit ILC2 activity through inducible T cell co-stimulator (ICOS):ICOSL interaction [167].

Although the involvement of ILC1s in asthma has not been extensively studied, several reports have highlighted a dual role for NK cells in allergic asthma. NK cell protective function is differentially regulated by various eicosanoids, including resolvin-E1, prostaglandins, lipoxin-A4, and cannabinoid receptor 2 (CB2). Both resolvin-E1 and lipoxin-A4 enhance NK cell protective functions. Resolvin-E1 promotes NK cell migration and cytotoxicity, thereby attenuating allergic inflammation [168]. Similarly, lipoxin-A4 activates NK cells by interacting with pro-resolving ALX/FPR2 receptors, enhancing their ability to clear airway eosinophils via apoptosis [87]. In contrast, PGI2 and CB2 suppress NK cell activity. In PGI-deficient mice, an increase in IFN-γ-producing NK cells inhibits ILC2 function, reducing inflammation and highlighting an anti-inflammatory role for NK cells [169]. Likewise, the absence of the CB2 receptor increases IFN- γ-producing pulmonary NK cells, leading to reduced ILC2 numbers, airway eosinophilia, and lower type 2 cytokine production in response to HDM exposure [170]. Beyond eicosanoids, NK cell function can also be regulated by other factors, such as Toll-like receptors (TLRs). Notably, poly(I:C)-mediated TLR3 activation of NK cells stimulates IL-17A production, leading to asthma exacerbation [171]. Additionally, TLR9 can indirectly stimulate NK cell production of IFN-γ via plasmacytoid dendritic cell (pDC)-derived type I IFNs, which suppress ILC2s and alleviate airway inflammation [136]. Finally, NK cell activation through the activating receptor NKG2D, triggered by its ligand murine UL16 binding protein-like transcript (MULT-1) and granzyme B production, further promotes HDM-mediated allergic inflammatory responses [85] (Fig. 1B).

Research into the mechanisms regulating ILC3 function and their role in orchestrating immune responses in asthma remains limited. However, several recent studies have begun to address this gap. IL-1β is recognized as a key cytokine driving ILC3 activation and the subsequent production of IL-17A, which contributes to neutrophilic airway inflammation [172]. A recent study further reported that IL-1β production by airway epithelial cells is elevated in smokers and is associated with the development of memory-like ILC3s, which exacerbate asthma symptoms [52]. The positive association between NCR+ ILC3s and the gut microbiome has also been implicated, with a potential role for NCR+ ILC3s in improving lung function in severe asthmatics [173]. Additionally, ILC3s, through MHC-II interactions with allergen-specific Th2 cells and microbe-specific Th17 cells, limit eosinophilic and neutrophilic inflammation, respectively [174] (Fig. 1C).

Innate-like lymphocytes

iNKT cells can play both protective and pathogenic roles in asthma (Fig. 2A). From a pathogenic perspective, iNKT cells are linked to the development of allergic asthma. A recent study reported that IL-4 production by lung iNKT cells is modulated by progranulin through the regulation of enhancer of zeste homolog 2 (EZH2) expression [175], a transcription activator previously shown to restrain pathogenic iNKT cell development [176]. In another study, de novo-fatty acid synthesis, mediated by acetyl-coA-carboxylase 1 (ACC1), promotes iNKT survival and exacerbates their deleterious effects in OVA and HDM models of allergic asthma [177]. iNKT cells can further enhance the immune response by upregulating the co-stimulatory molecule CD40L, which promotes DC maturation and subsequent CD4+ T cell differentiation [178]. Similarly, Deng et al. highlighted the potential importance of CD40-CD40L interactions between iNKT cells and DCs in promoting an asthma-like phenotype in the OVA model, using CD1d-deficient mice [179]. This highlights the critical role of iNKT cells in bridging innate and adaptive immunity in asthma pathogenesis.

While the aforementioned studies highlight the deleterious effects of iNKT cells, we and others have characterized a suppressive subset of iNKT cells and elucidated the regulatory pathways underlying their immunosuppressive functions. In an earlier study, Chuang et al. reported that influenza infection in neonatal mice preferentially expanded a suppressive CD4 CD8 iNKT population that highly expresses CD38 and can mitigate CD4+ T cell proliferation and the subsequent development of asthma [180]. Our study also identified a similar population that was induced by PM2.5 exposure in the absence of any glycolipid stimulation. Notably, we found that this subset is activated upon recognition of apoptotic cells through T-cell immunoglobulin and mucin domain 1 (TIM-1). This activation led to upregulation of PD-L1, which, through PD-1 signaling, suppressed the effector function of γδ T cells, resulting in reduced neutrophilic inflammation and attenuated AHR [181]. In a separate study, the chemokine CCL21 was shown to facilitate the migration of promyelocytic leukemia zinc finger protein (PLZF)+ iNKT cells to the lung, contributing to decreased airway resistance and increased asthma tolerance [182]. Nevertheless, further studies are required to delineate the exact mechanism by which these cells promote asthma tolerance.

In contrast to iNKT cells, the specific co-stimulatory mediators regulating γδ T cell and MAIT cell effector functions in the context of asthma remain poorly characterized. Nonetheless, studies have linked γδ T cells to the regulation of IgE production and neutrophilic asthma (Fig. 2B). Specifically, allergen-induced IL-6 trans-signaling promotes IL-17A production by γδ T cells, driving neutrophilic recruitment into the airways [66]. Consistently, asthmatic patients exhibiting elevated trans-signaling also present with increased neutrophilic and mixed granulocytic subtypes [66]. In non-allergic settings, PM2.5-driven neutrophilic inflammation is also mediated by γδ T cells in an IL-17A-dependent manner [181]. Crosstalk between γδ T cells and B cells has also been reported, where IL-4 and IL-10-producing CXCR5+ γδ T cells facilitate B cell antibody production [183]. Development of these Th2 cytokine-producing γδ T cells is negatively regulated by Itk, as Itk-deficient γδ T cells produce more Th2 cytokines, accompanied by increased IgE levels [184]. This effect is subset-specific: Vγ1 γδ T cells promote IgE production, whereas Vγ4 γδ T cells have an opposing function [185]. MAIT cells, on the other hand, can produce IFN-γ, which has been shown to suppress ILC2 function in Alternaria alternata-induced allergic airway inflammation [186] (Fig. 2C).

Potential therapeutic approaches targeting ILCs and innate-like lymphocytes

With growing research elucidating the roles of ILCs and innate-like lymphocytes in asthma, these immune cell populations have emerged as promising therapeutic targets. Here, we will discuss recent findings in the therapeutic manipulation of ILCs and innate-like lymphocytes in both animal models and human studies (Summarized in Table 1).

Table 1 Potential therapies for asthma and their mechanisms of action

Corticosteroids

Glucocorticoids, widely used anti-inflammatory agents in asthma, have been shown to reduce ILC2 levels in asthmatic patients with allergic rhinitis. Additionally, glucocorticoids inhibit ILC2 function by suppressing IL-5, IL-13, and IL-9 production via MEK/JAK-STAT signaling pathways, as demonstrated in studies using ILC2s sorted from asthma patients’ PBMCs [187].

Alarmins

ILC2 activity, survival, and cytokine production are regulated by alarmins such as IL-33, TSLP, and IL-25. There are several drugs that target these ILC2 activators. Itepekimab, a monoclonal antibody targeting IL-33, demonstrated improved lung function in patients with COPD during a phase 3 trial [188]. Additionally, a phase 2 trial revealed its efficacy in improving lung function in individuals with moderate-to-severe asthma [189]. Astegolimab is an anti-ST2, the IL-33 receptor, monoclonal antibody. A phase 2 trial for astegolimab reported a reduced asthma exacerbation rate (AER) in asthmatic patients, and is currently undergoing phase 3 trials for COPD [190]. However, in a phase 2 trial, astegolimab did not show significant improvement in disease outcomes for patients with atopic dermatitis [191]. Tozorakimab is an anti-IL-33 monoclonal antibody currently undergoing a phase 3 clinical trial to assess its efficacy in COPD and in patients hospitalized with viral lower respiratory tract infections [192, 193]. Tezepelumab is a monoclonal antibody that blocks TSLP. A phase 3 trial showed that patients with severe asthma experienced fewer exacerbations and improved lung function. Tezepelumab has already been approved for the treatment of asthma [194, 195]. On the other hand, drugs targeting IL-25 have not yet progressed to clinical trials. Although risankizumab, a monoclonal antibody targeting the ILC3 activator IL-23, was approved for the treatment in psoriasis, it failed to show significant improvement in severe asthma in a phase 2 trial [196].

Effector cytokines

Dupilumab, a monoclonal antibody that targets the IL-4 receptor, has been approved for the treatment of eosinophilic esophagitis. A phase 3 trial demonstrated significant improvements in lung function and asthma control in patients with severe asthma, leading to FDA approval for its use in moderate and severe asthma patients [197]. The drug is also undergoing clinical trials for COPD [198, 199]. Mepolizumab and reslizumab, monoclonal antibodies targeting IL-5, are approved for asthma treatment, with their use in COPD currently in a phase 3 trial [200,201,202]. Benralizumab, a monoclonal antibody against the IL-5 receptor, showed improvements in clinical outcomes for asthma and COPD patients in a phase 2 trial [203]. Although drugs targeting IL-13 are under development, results have been disappointing so far [204]. Ixekizumab and secukinumab, which target the ILC3-related cytokine IL-17, are approved for psoriasis treatment. However, secukinumab failed to improve asthma control, as indicated by the Asthma Control Questionnaire (ACQ) scores in patients with uncontrolled asthma, leading to its termination (ClinicalTrials.gov Identifier: NCT01478360) [205, 206]. Brodalumab, a monoclonal antibody targeting the interleukin-17 receptor, has also been approved for psoriasis but did not improve asthma outcomes in a phase 2 trial [207].

Bronchodilators

In addition to their established role as bronchodilators in asthma treatment, β2-agonists, muscarinic antagonists, and leukotriene receptor antagonists have been shown to regulate ILC2 function. β2-agonists, commonly used to relax airway smooth muscles, have been found to negatively regulate ILC2 proliferation and effector function [208]. Muscarinic antagonists, traditionally used to alleviate bronchoconstriction, also demonstrate inhibitory effects on ILC2 activity [209, 210]. Montelukast and zafirlukast are both cysteinyl leukotriene 1 (CysLT1) receptor antagonists approved for asthma management. Notably, montelukast has been shown to suppress ILC2 activity [211,212,213].

Signaling pathways

JAK3 inhibitors have been shown to suppress ILC2 survival, proliferation, and cytokine production, thereby alleviating ILC2-driven asthma in murine models [214]. Moreover, ILC2 activation and function are highly dependent on cellular metabolic processes. Inhibiting hypoxia-inducible factor 1-alpha (HIF-1α) or disrupting glycolytic metabolism has been demonstrated to suppress ILC2 activity and mitigate airway inflammation in mouse models of asthma [215].

Glycolipids

iNKT cells play a complex role in asthma, with certain subsets contributing to AHR and allergic inflammation, while others provide a protective effect. Activation of pulmonary iNKT cells by glycolipid antigens, such as α-GalCer and asperamide B, has been shown to induce AHR and eosinophilic inflammation in mice [216, 217]. However, other studies have shown that α-GalCer activation of iNKT cells can suppress AHR and allergic inflammation in an OVA-induced mouse model of asthma [218]. CD38hi iNKT cells, a specific subset of iNKT cells, have been shown to ameliorate the development of AHR in mice. Moreover, treatment with NU–α-GalCer can increase the number of CD38hi iNKT cells in the lungs and protect against allergen-induced AHR [180]. Additionally, α-LacCer, a weaker activator of iNKT cells compared to α-GalCer, can reduce AHR induced by both α-GalCer and glycosphingolipid (GSL)-1 [219]. Efforts to enhance therapeutic outcomes by developing alternative glycolipids have also been made, including the recent development of sp2-iminoglycolipids with enhanced biostability [220, 221]. These findings suggest that controlled activation of iNKT cells, potentially by using different glycolipid antigens and promoting regulatory subsets, may help reduce allergic airway inflammation.

Other potential approaches

Vitamin D3 supplementation has been associated with less severe asthma in both humans and murine models, with its effects attributed to the modulation of ILC2 function [222]. Piezo1, a calcium ion channel, plays a crucial role in regulating ILC2 activity and AHR in asthma. In murine models, activation of Piezo1 using Yoda1 treatment significantly reduced ILC2 cytokine secretion, proliferation, and survival, leading to diminished AHR and lung inflammation [223]. Additionally, targeting CD226 has also shown potential in asthma therapy. In a mouse model, administration of an anti-CD226 antibody effectively reduced cytokine production by ILC2s and alleviated AHR triggered by IL-33 or Alternaria alternata [224]. Signal regulatory protein alpha (SIRPα), an inhibitory receptor that regulates ILC2 effector functions and AHR, has been shown to suppress ILC2 proliferation and cytokine secretion when engaged with CD47Fc, thereby mitigating AHR and lung inflammation in murine models [225].

Conclusion

In conclusion, ILCs and innate-like lymphocytes are key regulators of immune responses in asthma, responding rapidly to environmental cues and allergen exposure. Their ability to amplify or dampen inflammation, depending on the microenvironment, highlights their functional plasticity and potential as therapeutic targets. However, the dynamics of their frequency, cytokine milieu, and receptor expression in the asthmatic lung remain unclear, including whether they shift from protective to pathogenic roles during disease progression. Future studies should focus on characterizing ILC subsets, their interplay with other immune cells, and developing tools to manipulate these cells in asthma models. Advancing our understanding of their biology could pave the way for novel immunotherapies to better manage or prevent asthma.

Availability of data and materials

Not applicable.

Abbreviations

ACC1:

Acetyl-coA-carboxylase 1

ACQ:

Asthma Control Questionnaire

AHR:

Airway hyperreactivity

BALF:

Bronchoalveolar lavage fluid

BCL-3:

B-cell lymphoma 3

BHB:

β-Hydroxybutyrate

BTN2A2:

Butyrophilin 2a2

cAMP:

Cyclic AMP

CB2:

Cannabinoid receptor 2

CGRP:

Calcitonin gene-related peptide

CMKLR1:

Chemerin chemokine-like receptor 1

COPD:

Chronic obstructive pulmonary disease

CysLT1:

Cysteinyl leukotriene 1

DCs:

Dendritic cells

EZH2:

Enhancer of zeste homolog 2

FABPs:

Fatty acid binding proteins

FAO:

Fatty acid oxidation

FFA:

Free fatty acid

FPR2:

Formyl peptide receptor 2

GATA3:

GATA binding protein 3

GM-CSF:

Granulocyte–macrophage colony-stimulating factor

GPR109A:

G protein-coupled receptor 109 A

GSL-1:

Glycosphingolipid

HDAC:

Histone deacetylase

HDM:

House dust mite

HFD:

High fat diet

HIF-1α:

Hypoxia-inducible factor 1-alpha

ICOS:

Inducible T cell co-stimulator

ICS:

Inhaled corticosteroid

IFN:

Interferon

IFNAR:

Interferon-α/β receptor

IFN-g:

Interferon-gamma

ILCs:

Innate lymphoid cells

ILC1:

Group 1 innate lymphoid cells

ILC2:

Group 2 innate lymphoid cells

ILC3:

Group 3 innate lymphoid cells

iNKT:

Invariant natural killer T cells

KIRs:

Killer immunoglobulin-like receptors

LABA:

Long-acting beta agonists

LTi:

Lymphoid tissue inducer cells

MAIT:

Mucosal-associated invariant T cells

MHC-II:

Major histocompatibility complex class II

MULT-1:

Murine UL16 binding protein-like transcript

NCRs:

Natural cytotoxicity receptors

NFAT:

Nuclear factor of activated T cells

NF-κB:

Nuclear factor kappa B

NK cells:

Natural killer cells

NKG2D:

Natural killer group 2, member D

NMU:

Neuromedin U

OVA:

Ovalbumin

PBMC:

Peripheral blood mononuclear cells

PD-1:

Programmed cell death protein 1

pDC:

Plasmacytoid DC

PD-L1:

Programmed death-ligand 1

PGD2:

Prostaglandin D2

PGE2:

Prostaglandin E2

PGI2:

Prostaglandin I2

Pla2g5:

Group V phospholipase A2

PLZF:

Promyelocytic leukemia zinc finger protein

PPAR-γ:

Peroxisome proliferator-activated receptor gamma

RORgt:

Retinoic acid receptor-related orphan receptor γt

RSV:

Respiratory syncytial virus

SCFA:

Short-chain fatty acid

SIRPα:

Signal regulatory protein alpha

SLAMF:

Signaling lymphocyte activation molecule family

STAT1:

Signal transducer and activator of transcription 1

T2:

Type 2

T17:

Type 17

T-bet:

T-box transcription factor

TCR:

T cell antigen receptor

Th2:

T helper 2

TIM1:

T-cell immunoglobulin and mucin domain 1

TLRs:

Toll-like receptors

TNF-a:

Tumor necrosis factor-alpha

Treg:

Regulatory T cells

TSLP:

Thymic stromal lymphopoietin

VIP:

Vasoactive intestinal peptide

References

  1. Yang CH, Lv JJ, Li XY, Yang XT, Yin MY. Global burden of asthma in young adults in 204 countries and territories, 1990–2019: systematic analysis of the Global burden of disease study 2019. Prev Med Rep. 2024;37: 102531.

    Article  PubMed  Google Scholar 

  2. Davison J, Doe S. A patient case demonstrating the efficacy of benralizumab in uncontrolled severe eosinophilic asthma refractory to omalizumab and mepolizumab treatment. Respir Med Case Rep. 2021;34: 101557.

    PubMed  PubMed Central  Google Scholar 

  3. Lang DM. Severe asthma: epidemiology, burden of illness, and heterogeneity. Allergy Asthma Proc. 2015;36(6):418–24.

    Article  PubMed  Google Scholar 

  4. Kuruvilla ME, Lee FE, Lee GB. Understanding asthma phenotypes, endotypes, and mechanisms of disease. Clin Rev Allergy Immunol. 2019;56(2):219–33.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Ricciardolo FLM, Sprio AE, Baroso A, Gallo F, Riccardi E, Bertolini F, et al. Characterization of T2-low and T2-high asthma phenotypes in real-life. Biomedicines. 2021;9(11):1684.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fahy JV. Type 2 inflammation in asthma–present in most, absent in many. Nat Rev Immunol. 2015;15(1):57–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mishra PE, Melen E, Koppelman GH, Celedon JC. T2-low asthma in school-aged children: unacknowledged and understudied. Lancet Respir Med. 2023;11(12):1044–5.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Kyriakopoulos C, Gogali A, Bartziokas K, Kostikas K. Identification and treatment of T2-low asthma in the era of biologics. ERJ Open Res. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/23120541.00309-2020.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Meininger I, Carrasco A, Rao A, Soini T, Kokkinou E, Mjosberg J. Tissue-specific features of innate lymphoid cells. Trends Immunol. 2020;41(10):902–17.

    Article  CAS  PubMed  Google Scholar 

  10. Victor JR, Lezmi G, Leite-de-Moraes M. New insights into asthma inflammation: focus on iNKT, MAIT, and gammadeltaT cells. Clin Rev Allergy Immunol. 2020;59(3):371–81.

    Article  CAS  PubMed  Google Scholar 

  11. Sudan R, Gilfillan S, Colonna M. Group 1 ILCs: heterogeneity, plasticity, and transcriptional regulation. Immunol Rev. 2024;323(1):107–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Spits H, Bernink JH, Lanier L. NK cells and type 1 innate lymphoid cells: partners in host defense. Nat Immunol. 2016;17(7):758–64.

    Article  CAS  PubMed  Google Scholar 

  13. Haspeslagh E, van Helden MJ, Deswarte K, De Prijck S, van Moorleghem J, Boon L, et al. Role of NKp46(+) natural killer cells in house dust mite-driven asthma. EMBO Mol Med. 2018;10(4): e8657.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zhang J, Le Gras S, Pouxvielh K, Faure F, Fallone L, Kern N, et al. Sequential actions of EOMES and T-BET promote stepwise maturation of natural killer cells. Nat Commun. 2021;12(1):5446.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Yoon SR, Kim TD, Choi I. Understanding of molecular mechanisms in natural killer cell therapy. Exp Mol Med. 2015;47(2): e141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Paul S, Lal G. The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front Immunol. 2017;8:1124.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Elemam NM, Ramakrishnan RK, Hundt JE, Halwani R, Maghazachi AA, Hamid Q. Innate lymphoid cells and natural killer cells in bacterial infections: function, dysregulation, and therapeutic targets. Front Cell Infect Microbiol. 2021;11: 733564.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kim CH, Hashimoto-Hill S, Kim M. Migration and tissue tropism of innate lymphoid cells. Trends Immunol. 2016;37(1):68–79.

    Article  CAS  PubMed  Google Scholar 

  19. Fuchs A, Vermi W, Lee JS, Lonardi S, Gilfillan S, Newberry RD, et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-gamma-producing cells. Immunity. 2013;38(4):769–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Weizman OE, Adams NM, Schuster IS, Krishna C, Pritykin Y, Lau C, et al. ILC1 confer early host protection at initial sites of viral infection. Cell. 2017;171(4):795-808 e12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Klose CSN, Flach M, Mohle L, Rogell L, Hoyler T, Ebert K, et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell. 2014;157(2):340–56.

    Article  CAS  PubMed  Google Scholar 

  22. Curio S, Belz GT. The unique role of innate lymphoid cells in cancer and the hepatic microenvironment. Cell Mol Immunol. 2022;19(9):1012–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature. 2010;463(7280):540–4.

    Article  CAS  PubMed  Google Scholar 

  24. Neill DR, Wong SH, Bellosi A, Flynn RJ, Daly M, Langford TK, et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature. 2010;464(7293):1367–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhou L. Striking similarity: GATA-3 regulates ILC2 and Th2 cells. Immunity. 2012;37(4):589–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ricardo-Gonzalez RR, Van Dyken SJ, Schneider C, Lee J, Nussbaum JC, Liang HE, et al. Tissue signals imprint ILC2 identity with anticipatory function. Nat Immunol. 2018;19(10):1093–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Monticelli LA, Sonnenberg GF, Abt MC, Alenghat T, Ziegler CG, Doering TA, et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat Immunol. 2011;12(11):1045–54.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Huang Y, Guo L, Qiu J, Chen X, Hu-Li J, Siebenlist U, et al. IL-25-responsive, lineage-negative KLRG1(hi) cells are multipotential “inflammatory” type 2 innate lymphoid cells. Nat Immunol. 2015;16(2):161–9.

    Article  CAS  PubMed  Google Scholar 

  29. Huang Y, Mao K, Chen X, Sun MA, Kawabe T, Li W, et al. S1P-dependent interorgan trafficking of group 2 innate lymphoid cells supports host defense. Science. 2018;359(6371):114–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Simoni Y, Newell EW. Dissecting human ILC heterogeneity: more than just three subsets. Immunology. 2018;153(3):297–303.

    Article  CAS  PubMed  Google Scholar 

  31. Mjosberg JM, Trifari S, Crellin NK, Peters CP, van Drunen CM, Piet B, et al. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol. 2011;12(11):1055–62.

    Article  PubMed  Google Scholar 

  32. Martinez-Gonzalez I, Matha L, Steer CA, Ghaedi M, Poon GF, Takei F. Allergen-experienced group 2 innate lymphoid cells acquire memory-like properties and enhance allergic lung inflammation. Immunity. 2016;45(1):198–208.

    Article  CAS  PubMed  Google Scholar 

  33. Ohne Y, Silver JS, Thompson-Snipes L, Collet MA, Blanck JP, Cantarel BL, et al. IL-1 is a critical regulator of group 2 innate lymphoid cell function and plasticity. Nat Immunol. 2016;17(6):646–55.

    Article  CAS  PubMed  Google Scholar 

  34. Bielecki P, Riesenfeld SJ, Hutter JC, Torlai Triglia E, Kowalczyk MS, Ricardo-Gonzalez RR, et al. Skin-resident innate lymphoid cells converge on a pathogenic effector state. Nature. 2021;592(7852):128–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lim AI, Menegatti S, Bustamante J, Le Bourhis L, Allez M, Rogge L, et al. IL-12 drives functional plasticity of human group 2 innate lymphoid cells. J Exp Med. 2016;213(4):569–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bal SM, Bernink JH, Nagasawa M, Groot J, Shikhagaie MM, Golebski K, et al. IL-1beta, IL-4 and IL-12 control the fate of group 2 innate lymphoid cells in human airway inflammation in the lungs. Nat Immunol. 2016;17(6):636–45.

    Article  CAS  PubMed  Google Scholar 

  37. Bartemes KR, Kita H. Roles of innate lymphoid cells (ILCs) in allergic diseases: The 10-year anniversary for ILC2s. J Allergy Clin Immunol. 2021;147(5):1531–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Luo CH, Lai AC, Tsai CC, Chen WY, Chang YS, Chung EJ, et al. Staphylococcus aureus exacerbates dermal IL-33/ILC2 axis activation through evoking RIPK3/MLKL-mediated necroptosis of dry skin. JCI Insight. 2024;9(6): e166821.

    PubMed  PubMed Central  Google Scholar 

  39. Lee MW, Odegaard JI, Mukundan L, Qiu Y, Molofsky AB, Nussbaum JC, et al. Activated type 2 innate lymphoid cells regulate beige fat biogenesis. Cell. 2015;160(1–2):74–87.

    Article  CAS  PubMed  Google Scholar 

  40. Oliphant CJ, Hwang YY, Walker JA, Salimi M, Wong SH, Brewer JM, et al. MHCII-mediated dialog between group 2 innate lymphoid cells and CD4(+) T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity. 2014;41(2):283–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Moral JA, Leung J, Rojas LA, Ruan J, Zhao J, Sethna Z, et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature. 2020;579(7797):130–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li Z, Ma R, Tang H, Guo J, Shah Z, Zhang J, et al. Therapeutic application of human type 2 innate lymphoid cells via induction of granzyme B-mediated tumor cell death. Cell. 2024;187(3):624-41 e23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. De Grove KC, Provoost S, Verhamme FM, Bracke KR, Joos GF, Maes T, et al. Characterization and quantification of innate lymphoid cell subsets in human lung. PLoS ONE. 2016;11(1): e0145961.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Satoh-Takayama N, Vosshenrich CA, Lesjean-Pottier S, Sawa S, Lochner M, Rattis F, et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity. 2008;29(6):958–70.

    Article  CAS  PubMed  Google Scholar 

  45. Hepworth MR, Fung TC, Masur SH, Kelsen JR, McConnell FM, Dubrot J, et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4(+) T cells. Science. 2015;348(6238):1031–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Buonocore S, Ahern PP, Uhlig HH, Ivanov II, Littman DR, Maloy KJ, et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature. 2010;464(7293):1371–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. King JI, Melo-Gonzalez F, Malengier-Devlies B, Tacho-Pinot R, Magalhaes MS, Hodge SH, et al. Bcl-2 supports survival and metabolic fitness of quiescent tissue-resident ILC3. Mucosal Immunol. 2023;16(5):658–70.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Zhao M, Shao F, Yu D, Zhang J, Liu Z, Ma J, et al. Maturation and specialization of group 2 innate lymphoid cells through the lung-gut axis. Nat Commun. 2022;13(1):7600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li M, Wang Z, Jiang W, Lu Y, Zhang J. The role of group 3 innate lymphoid cell in intestinal disease. Front Immunol. 2023;14:1171826.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Tizian C, Lahmann A, Holsken O, Cosovanu C, Kofoed-Branzk M, Heinrich F, et al. c-Maf restrains T-bet-driven programming of CCR6-negative group 3 innate lymphoid cells. Elife. 2020;9: e52549.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Parker ME, Barrera A, Wheaton JD, Zuberbuehler MK, Allan DSJ, Carlyle JR, et al. c-Maf regulates the plasticity of group 3 innate lymphoid cells by restraining the type 1 program. J Exp Med. 2020;217(1): e20191030.

    Article  PubMed  Google Scholar 

  52. Ham J, Kim J, Sohn KH, Park IW, Choi BW, Chung DH, et al. Cigarette smoke aggravates asthma by inducing memory-like type 3 innate lymphoid cells. Nat Commun. 2022;13(1):3852.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Bennstein SB. Unraveling natural killer T-cells development. Front Immunol. 2017;8:1950.

    Article  PubMed  Google Scholar 

  54. Brennan PJ, Tatituri RV, Brigl M, Kim EY, Tuli A, Sanderson JP, et al. Invariant natural killer T cells recognize lipid self antigen induced by microbial danger signals. Nat Immunol. 2011;12(12):1202–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Fischer K, Scotet E, Niemeyer M, Koebernick H, Zerrahn J, Maillet S, et al. Mycobacterial phosphatidylinositol mannoside is a natural antigen for CD1d-restricted T cells. Proc Natl Acad Sci U S A. 2004;101(29):10685–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Liao CM, Zimmer MI, Wang CR. The functions of type I and type II natural killer T cells in inflammatory bowel diseases. Inflamm Bowel Dis. 2013;19(6):1330–8.

    Article  PubMed  Google Scholar 

  57. Lee YJ, Holzapfel KL, Zhu J, Jameson SC, Hogquist KA. Steady-state production of IL-4 modulates immunity in mouse strains and is determined by lineage diversity of iNKT cells. Nat Immunol. 2013;14(11):1146–54.

    Article  CAS  PubMed  Google Scholar 

  58. Matsuda H, Suda T, Sato J, Nagata T, Koide Y, Chida K, et al. alpha-Galactosylceramide, a ligand of natural killer T cells, inhibits allergic airway inflammation. Am J Respir Cell Mol Biol. 2005;33(1):22–31.

    Article  CAS  PubMed  Google Scholar 

  59. Wingender G, Rogers P, Batzer G, Lee MS, Bai D, Pei B, et al. Invariant NKT cells are required for airway inflammation induced by environmental antigens. J Exp Med. 2011;208(6):1151–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Silva-Santos B, Mensurado S, Coffelt SB. gammadelta T cells: pleiotropic immune effectors with therapeutic potential in cancer. Nat Rev Cancer. 2019;19(7):392–404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Adams EJ, Gu S, Luoma AM. Human gamma delta T cells: Evolution and ligand recognition. Cell Immunol. 2015;296(1):31–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ruf B, Greten TF, Korangy F. Innate lymphoid cells and innate-like T cells in cancer - at the crossroads of innate and adaptive immunity. Nat Rev Cancer. 2023;23(6):351–71.

    Article  CAS  PubMed  Google Scholar 

  63. Zheng R, Yang Q. The role of the gamma delta T cell in allergic diseases. J Immunol Res. 2014;2014: 963484.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Shibata K, Yamada H, Nakamura M, Hatano S, Katsuragi Y, Kominami R, et al. IFN-gamma-producing and IL-17-producing gammadelta T cells differentiate at distinct developmental stages in murine fetal thymus. J Immunol. 2014;192(5):2210–8.

    Article  CAS  PubMed  Google Scholar 

  65. Cui ZH, Joetham A, Aydintug MK, Hahn YS, Born WK, Gelfand EW. Reversal of allergic airway hyperreactivity after long-term allergen challenge depends on gammadelta T cells. Am J Respir Crit Care Med. 2003;168(11):1324–32.

    Article  PubMed  Google Scholar 

  66. Ullah MA, Revez JA, Loh Z, Simpson J, Zhang V, Bain L, et al. Allergen-induced IL-6 trans-signaling activates gammadelta T cells to promote type 2 and type 17 airway inflammation. J Allergy Clin Immunol. 2015;136(4):1065–73.

    Article  CAS  PubMed  Google Scholar 

  67. Kjer-Nielsen L, Patel O, Corbett AJ, Le Nours J, Meehan B, Liu L, et al. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature. 2012;491(7426):717–23.

    Article  CAS  PubMed  Google Scholar 

  68. Zheng Y, Han F, Ho A, Xue Y, Wu Z, Chen X, et al. Role of MAIT cells in gastrointestinal tract bacterial infections in humans: more than a gut feeling. Mucosal Immunol. 2023;16(5):740–52.

    Article  PubMed  Google Scholar 

  69. Hinks TSC, Zhang XW. MAIT cell activation and functions. Front Immunol. 2020;11:1014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wen X, Zhang X, Nian S, Wei G, Guo X, Yu H, et al. Title of article: Mucosal-associated invariant T cells in lung diseases. Int Immunopharmacol. 2021;94: 107485.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ye L, Pan J, Pasha MA, Shen X, D’Souza SS, Fung ITH, et al. Mucosal-associated invariant T cells restrict allergic airway inflammation. J Allergy Clin Immunol. 2020;145(5):1469-1473 e4.

    Article  CAS  PubMed  Google Scholar 

  72. Jonckheere AC, Bullens DMA, Seys SF. Innate lymphoid cells in asthma: pathophysiological insights from murine models to human asthma phenotypes. Curr Opin Allergy Clin Immunol. 2019;19(1):53–60.

    Article  PubMed  Google Scholar 

  73. Ham J, Lim M, Kim D, Kim HY. Memory-like innate lymphoid cells in the pathogenesis of asthma. Front Immunol. 2022;13:1005517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chang YJ, Kim HY, Albacker LA, Baumgarth N, McKenzie AN, Smith DE, et al. Innate lymphoid cells mediate influenza-induced airway hyper-reactivity independently of adaptive immunity. Nat Immunol. 2011;12(7):631–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kim HY, Umetsu DT, Dekruyff RH. Innate lymphoid cells in asthma: Will they take your breath away? Eur J Immunol. 2016;46(4):795–806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Bartemes KR, Kephart GM, Fox SJ, Kita H. Enhanced innate type 2 immune response in peripheral blood from patients with asthma. J Allergy Clin Immunol. 2014;134(3):671-678 e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kim BK, Lee HS, Lee SY, Park HW. Innate lymphoid cell population distributions and related gene expression characteristics in blood from allergic and nonallergic patients with eosinophilic asthma. Asian Pac J Allergy Immunol. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.12932/AP-140124-1765.

    Article  PubMed  Google Scholar 

  78. Pavlidis S, Guo Y, Sun K, Rossios C, Rowe A, Loza M, et al. Molecular evidence of Group 1 innate lymphoid cell activation in the U-BIOPRED cohort. Eur Respir J. 2017;50(suppl 61):OA278.

    Google Scholar 

  79. Kim J, Chang Y, Bae B, Sohn KH, Cho SH, Chung DH, et al. Innate immune crosstalk in asthmatic airways: Innate lymphoid cells coordinate polarization of lung macrophages. J Allergy Clin Immunol. 2019;143(5):1769–82.

    Article  CAS  PubMed  Google Scholar 

  80. Lin SJ, Chang LY, Yan DC, Huang YJ, Lin TJ, Lin TY. Decreased intercellular adhesion molecule-1 (CD54) and L-selectin (CD62L) expression on peripheral blood natural killer cells in asthmatic children with acute exacerbation. Allergy. 2003;58(1):67–71.

    Article  CAS  PubMed  Google Scholar 

  81. Jira M, Antosova E, Vondra V, Strejcek J, Mazakova H, Prazakova J. Natural killer and interleukin-2 induced cytotoxicity in asthmatics. I. Effect of acute antigen-specific challenge. Allergy. 1988;43(4):294–8.

    Article  CAS  PubMed  Google Scholar 

  82. Timonen T, Stenius-Aarniala B. Natural killer cell activity in asthma. Clin Exp Immunol. 1985;59(1):85–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Wei H, Zhang J, Xiao W, Feng J, Sun R, Tian Z. Involvement of human natural killer cells in asthma pathogenesis: natural killer 2 cells in type 2 cytokine predominance. J Allergy Clin Immunol. 2005;115(4):841–7.

    Article  CAS  PubMed  Google Scholar 

  84. Mathias CB, Guernsey LA, Zammit D, Brammer C, Wu CA, Thrall RS, et al. Pro-inflammatory role of natural killer cells in the development of allergic airway disease. Clin Exp Allergy. 2014;44(4):589–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Farhadi N, Lambert L, Triulzi C, Openshaw PJ, Guerra N, Culley FJ. Natural killer cell NKG2D and granzyme B are critical for allergic pulmonary inflammation. J Allergy Clin Immunol. 2014;133(3):827-835 e3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kaiko GE, Phipps S, Angkasekwinai P, Dong C, Foster PS. NK cell deficiency predisposes to viral-induced Th2-type allergic inflammation via epithelial-derived IL-25. J Immunol. 2010;185(8):4681–90.

    Article  CAS  PubMed  Google Scholar 

  87. Barnig C, Cernadas M, Dutile S, Liu X, Perrella MA, Kazani S, et al. Lipoxin A4 regulates natural killer cell and type 2 innate lymphoid cell activation in asthma. Sci Transl Med. 2013;5(174):174ra26.

    Article  PubMed  Google Scholar 

  88. Hosseini B, Berthon BS, Starkey MR, Collison A, McLoughlin RF, Williams EJ, et al. Children with asthma have impaired innate immunity and increased numbers of type 2 innate lymphoid cells compared with healthy controls. Front Immunol. 2021;12: 664668.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Winkler C, Hochdorfer T, Israelsson E, Hasselberg A, Cavallin A, Thorn K, et al. Activation of group 2 innate lymphoid cells after allergen challenge in asthmatic patients. J Allergy Clin Immunol. 2019;144(1):61-69 e7.

    Article  CAS  PubMed  Google Scholar 

  90. Chen R, Smith SG, Salter B, El-Gammal A, Oliveria JP, Obminski C, et al. Allergen-induced increases in sputum levels of group 2 innate lymphoid cells in subjects with asthma. Am J Respir Crit Care Med. 2017;196(6):700–12.

    Article  CAS  PubMed  Google Scholar 

  91. Liu T, Wu J, Zhao J, Wang J, Zhang Y, Liu L, et al. Type 2 innate lymphoid cells: A novel biomarker of eosinophilic airway inflammation in patients with mild to moderate asthma. Respir Med. 2015;109(11):1391–6.

    Article  PubMed  Google Scholar 

  92. Christianson CA, Goplen NP, Zafar I, Irvin C, Good JT Jr, Rollins DR, et al. Persistence of asthma requires multiple feedback circuits involving type 2 innate lymphoid cells and IL-33. J Allergy Clin Immunol. 2015;136(1):59-68 e14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kim DI, Song MK, Lee K. Comparison of asthma phenotypes in OVA-induced mice challenged via inhaled and intranasal routes. BMC Pulm Med. 2019;19(1):241.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Xi GP, Zhang Q, Yin J. Establishment and characterization of murine models of asthma and subcutaneous immunotherapy for Humulus pollen allergy. Immun Inflamm Dis. 2021;9(2):443–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Woo LN, Guo WY, Wang X, Young A, Salehi S, Hin A, et al. A 4-week model of house dust mite (HDM) induced allergic airways inflammation with airway remodeling. Sci Rep. 2018;8(1):6925.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Morita H, Arae K, Unno H, Miyauchi K, Toyama S, Nambu A, et al. An interleukin-33-mast cell-interleukin-2 axis suppresses papain-induced allergic inflammation by promoting regulatory T cell numbers. Immunity. 2015;43(1):175–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Takai T, Ikeda S. Barrier dysfunction caused by environmental proteases in the pathogenesis of allergic diseases. Allergol Int. 2011;60(1):25–35.

    Article  CAS  PubMed  Google Scholar 

  98. Wu YH, Lai AC, Chi PY, Thio CL, Chen WY, Tsai CH, et al. Pulmonary IL-33 orchestrates innate immune cells to mediate respiratory syncytial virus-evoked airway hyperreactivity and eosinophilia. Allergy. 2020;75(4):818–30.

    Article  CAS  PubMed  Google Scholar 

  99. Wojno ED, Monticelli LA, Tran SV, Alenghat T, Osborne LC, Thome JJ, et al. The prostaglandin D(2) receptor CRTH2 regulates accumulation of group 2 innate lymphoid cells in the inflamed lung. Mucosal Immunol. 2015;8(6):1313–23.

    Article  PubMed  Google Scholar 

  100. Nagasawa M, Heesters BA, Kradolfer CMA, Krabbendam L, Martinez-Gonzalez I, de Bruijn MJW, et al. KLRG1 and NKp46 discriminate subpopulations of human CD117(+)CRTH2(-) ILCs biased toward ILC2 or ILC3. J Exp Med. 2019;216(8):1762–76.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Liu S, Sirohi K, Verma M, McKay J, Michalec L, Sripada A, et al. Optimal identification of human conventional and nonconventional (CRTH2(-)IL7Ralpha(-)) ILC2s using additional surface markers. J Allergy Clin Immunol. 2020;146(2):390–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Beuraud C, Lombardi V, Luce S, Horiot S, Naline E, Neukirch C, et al. CCR10(+) ILC2s with ILC1-like properties exhibit a protective function in severe allergic asthma. Allergy. 2019;74(5):933–43.

    Article  CAS  PubMed  Google Scholar 

  103. Linden A. Role of interleukin-17 and the neutrophil in asthma. Int Arch Allergy Immunol. 2001;126(3):179–84.

    Article  CAS  PubMed  Google Scholar 

  104. Kim HY, Lee HJ, Chang YJ, Pichavant M, Shore SA, Fitzgerald KA, et al. Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity. Nat Med. 2014;20(1):54–61.

    Article  CAS  PubMed  Google Scholar 

  105. Akbari O, Stock P, Meyer E, Kronenberg M, Sidobre S, Nakayama T, et al. Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity. Nat Med. 2003;9(5):582–8.

    Article  CAS  PubMed  Google Scholar 

  106. Lisbonne M, Diem S, de Castro KA, Lefort J, Araujo LM, Hachem P, et al. Cutting edge: invariant V alpha 14 NKT cells are required for allergen-induced airway inflammation and hyperreactivity in an experimental asthma model. J Immunol. 2003;171(4):1637–41.

    Article  CAS  PubMed  Google Scholar 

  107. Lee KA, Kang MH, Lee YS, Kim YJ, Kim DH, Ko HJ, et al. A distinct subset of natural killer T cells produces IL-17, contributing to airway infiltration of neutrophils but not to airway hyperreactivity. Cell Immunol. 2008;251(1):50–5.

    Article  CAS  PubMed  Google Scholar 

  108. Pichavant M, Goya S, Meyer EH, Johnston RA, Kim HY, Matangkasombut P, et al. Ozone exposure in a mouse model induces airway hyperreactivity that requires the presence of natural killer T cells and IL-17. J Exp Med. 2008;205(2):385–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Kim EY, Battaile JT, Patel AC, You Y, Agapov E, Grayson MH, et al. Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease. Nat Med. 2008;14(6):633–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Akbari O, Faul JL, Hoyte EG, Berry GJ, Wahlstrom J, Kronenberg M, et al. CD4+ invariant T-cell-receptor+ natural killer T cells in bronchial asthma. N Engl J Med. 2006;354(11):1117–29.

    Article  CAS  PubMed  Google Scholar 

  111. Matangkasombut P, Marigowda G, Ervine A, Idris L, Pichavant M, Kim HY, et al. Natural killer T cells in the lungs of patients with asthma. J Allergy Clin Immunol. 2009;123(5):1181–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Hamzaoui A, Cheik Rouhou S, Grairi H, Abid H, Ammar J, Chelbi H, et al. NKT cells in the induced sputum of severe asthmatics. Mediators Inflamm. 2006;2006(2):71214.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Mutalithas K, Croudace J, Guillen C, Siddiqui S, Thickett D, Wardlaw A, et al. Bronchoalveolar lavage invariant natural killer T cells are not increased in asthma. J Allergy Clin Immunol. 2007;119(5):1274–6.

    Article  CAS  PubMed  Google Scholar 

  114. Yamamoto H, Okamoto Y, Horiguchi S, Kunii N, Yonekura S, Nakayama T. Detection of natural killer T cells in the sinus mucosa from asthmatics with chronic sinusitis. Allergy. 2007;62(12):1451–5.

    Article  CAS  PubMed  Google Scholar 

  115. Shim JU, Koh YI. Increased Th2-like invariant natural killer T cells in peripheral blood from patients with asthma. Allergy Asthma Immunol Res. 2014;6(5):444–8.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Nagata Y, Kamijuku H, Taniguchi M, Ziegler S, Seino K. Differential role of thymic stromal lymphopoietin in the induction of airway hyperreactivity and Th2 immune response in antigen-induced asthma with respect to natural killer T cell function. Int Arch Allergy Immunol. 2007;144(4):305–14.

    Article  CAS  PubMed  Google Scholar 

  117. Bourgeois E, Van LP, Samson M, Diem S, Barra A, Roga S, et al. The pro-Th2 cytokine IL-33 directly interacts with invariant NKT and NK cells to induce IFN-gamma production. Eur J Immunol. 2009;39(4):1046–55.

    Article  CAS  PubMed  Google Scholar 

  118. Terashima A, Watarai H, Inoue S, Sekine E, Nakagawa R, Hase K, et al. A novel subset of mouse NKT cells bearing the IL-17 receptor B responds to IL-25 and contributes to airway hyperreactivity. J Exp Med. 2008;205(12):2727–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Schauer U, Dippel E, Gieler U, Brauer J, Jung T, Heymanns J, et al. T cell receptor gamma delta bearing cells are decreased in the peripheral blood of patients with atopic diseases. Clin Exp Immunol. 1991;86(3):440–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mota-Pinto A, Todo A, Alves V, Santos A, Santos M. Regulatory T cells in elderly patients with asthma. J Investig Allergol Clin Immunol. 2011;21(3):199–206.

    CAS  PubMed  Google Scholar 

  121. Urboniene D, Babusyte A, Lotvall J, Sakalauskas R, Sitkauskiene B. Distribution of gammadelta and other T-lymphocyte subsets in patients with chronic obstructive pulmonary disease and asthma. Respir Med. 2013;107(3):413–23.

    Article  PubMed  Google Scholar 

  122. de-Oliveira MG, Lira AAL, Sgnotto FR, Inoue AHS, Santos LS, Nakamatsu BY, et al. Maternal IgG impairs the maturation of offspring intrathymic IL-17-producing gammadeltaT cells: implications for murine and human allergies. Clin Exp Allergy. 2019;49(7):1000–12.

    Article  CAS  PubMed  Google Scholar 

  123. Huang Y, Aydintug MK, Loomis J, Macleod MK, McKee AS, Kirchenbaum G, et al. Antigen-specific regulation of IgE antibodies by non-antigen-specific gammadelta T cells. J Immunol. 2013;190(3):913–21.

    Article  CAS  PubMed  Google Scholar 

  124. Zhao Y, Yang J, Gao YD. Altered expressions of helper T cell (Th)1, Th2, and Th17 cytokines in CD8(+) and gammadelta T cells in patients with allergic asthma. J Asthma. 2011;48(5):429–36.

    Article  CAS  PubMed  Google Scholar 

  125. Krug N, Erpenbeck VJ, Balke K, Petschallies J, Tschernig T, Hohlfeld JM, et al. Cytokine profile of bronchoalveolar lavage-derived CD4(+), CD8(+), and gammadelta T cells in people with asthma after segmental allergen challenge. Am J Respir Cell Mol Biol. 2001;25(1):125–31.

    Article  CAS  PubMed  Google Scholar 

  126. Hinks TS, Zhou X, Staples KJ, Dimitrov BD, Manta A, Petrossian T, et al. Innate and adaptive T cells in asthmatic patients: relationship to severity and disease mechanisms. J Allergy Clin Immunol. 2015;136(2):323–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ishimori A, Harada N, Chiba A, Harada S, Matsuno K, Makino F, et al. Circulating activated innate lymphoid cells and mucosal-associated invariant T cells are associated with airflow limitation in patients with asthma. Allergol Int. 2017;66(2):302–9.

    Article  CAS  PubMed  Google Scholar 

  128. Lezmi G, Abou-Taam R, Garcelon N, Dietrich C, Machavoine F, Delacourt C, et al. Evidence for a MAIT-17-high phenotype in children with severe asthma. J Allergy Clin Immunol. 2019;144(6):1714-1716 e6.

    Article  PubMed  Google Scholar 

  129. Lezmi G, Abou Taam R, Dietrich C, Chatenoud L, de Blic J, Leite-de-Moraes M. Circulating IL-17-producing mucosal-associated invariant T cells (MAIT) are associated with symptoms in children with asthma. Clin Immunol. 2018;188:7–11.

    Article  CAS  PubMed  Google Scholar 

  130. Tastan C, Karhan E, Zhou W, Fleming E, Voigt AY, Yao X, et al. Tuning of human MAIT cell activation by commensal bacteria species and MR1-dependent T-cell presentation. Mucosal Immunol. 2018;11(6):1591–605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Kabata H, Moro K, Fukunaga K, Suzuki Y, Miyata J, Masaki K, et al. Thymic stromal lymphopoietin induces corticosteroid resistance in natural helper cells during airway inflammation. Nat Commun. 2013;4:2675.

    Article  PubMed  Google Scholar 

  132. Li Y, Lan F, Yang Y, Xu Y, Chen Y, Qin X, et al. The absence of IL-9 reduces allergic airway inflammation by reducing ILC2, Th2 and mast cells in murine model of asthma. BMC Pulm Med. 2022;22(1):180.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Motomura Y, Morita H, Moro K, Nakae S, Artis D, Endo TA, et al. Basophil-derived interleukin-4 controls the function of natural helper cells, a member of ILC2s, in lung inflammation. Immunity. 2014;40(5):758–71.

    Article  CAS  PubMed  Google Scholar 

  134. Meylan F, Hawley ET, Barron L, Barlow JL, Penumetcha P, Pelletier M, et al. The TNF-family cytokine TL1A promotes allergic immunopathology through group 2 innate lymphoid cells. Mucosal Immunol. 2014;7(4):958–68.

    Article  CAS  PubMed  Google Scholar 

  135. Hurrell BP, Galle-Treger L, Jahani PS, Howard E, Helou DG, Banie H, et al. TNFR2 signaling enhances ILC2 survival, function, and induction of airway hyperreactivity. Cell Rep. 2019;29(13):4509-24 e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Thio CL, Lai AC, Chi PY, Webster G, Chang YJ. Toll-like receptor 9-dependent interferon production prevents group 2 innate lymphoid cell-driven airway hyperreactivity. J Allergy Clin Immunol. 2019;144(3):682-97 e9.

    Article  CAS  PubMed  Google Scholar 

  137. Moro K, Kabata H, Tanabe M, Koga S, Takeno N, Mochizuki M, et al. Interferon and IL-27 antagonize the function of group 2 innate lymphoid cells and type 2 innate immune responses. Nat Immunol. 2016;17(1):76–86.

    Article  CAS  PubMed  Google Scholar 

  138. Ogasawara N, Poposki JA, Klingler AI, Tan BK, Weibman AR, Hulse KE, et al. IL-10, TGF-beta, and glucocorticoid prevent the production of type 2 cytokines in human group 2 innate lymphoid cells. J Allergy Clin Immunol. 2018;141(3):1147-1151 e8.

    Article  CAS  PubMed  Google Scholar 

  139. Silver JS, Kearley J, Copenhaver AM, Sanden C, Mori M, Yu L, et al. Inflammatory triggers associated with exacerbations of COPD orchestrate plasticity of group 2 innate lymphoid cells in the lungs. Nat Immunol. 2016;17(6):626–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Golebski K, Ros XR, Nagasawa M, van Tol S, Heesters BA, Aglmous H, et al. IL-1beta, IL-23, and TGF-beta drive plasticity of human ILC2s towards IL-17-producing ILCs in nasal inflammation. Nat Commun. 2019;10(1):2162.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Higham A, Cadden P, Southworth T, Rossall M, Kolsum U, Lea S, et al. Leukotriene B4 levels in sputum from asthma patients. ERJ Open Res. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/23120541.00088-2015.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Mansour M, Farouk N, El Maragy A, Radwan I, El-Ahmady O. Elevated plasma level of leukotrienes in bronchial asthma patients: a possible clinical relevance. Dis Mark. 1995;12(2):117–22.

    Article  CAS  Google Scholar 

  143. von Moltke J, O’Leary CE, Barrett NA, Kanaoka Y, Austen KF, Locksley RM. Leukotrienes provide an NFAT-dependent signal that synergizes with IL-33 to activate ILC2s. J Exp Med. 2017;214(1):27–37.

    Article  Google Scholar 

  144. Zhou W, Toki S, Zhang J, Goleniewksa K, Newcomb DC, Cephus JY, et al. Prostaglandin I2 signaling and inhibition of group 2 innate lymphoid cell responses. Am J Respir Crit Care Med. 2016;193(1):31–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhou Y, Wang W, Zhao C, Wang Y, Wu H, Sun X, et al. Prostaglandin E(2) inhibits group 2 innate lymphoid cell activation and allergic airway inflammation through E-prostanoid 4-cyclic adenosine monophosphate signaling. Front Immunol. 2018;9:501.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Xue L, Salimi M, Panse I, Mjosberg JM, McKenzie AN, Spits H, et al. Prostaglandin D2 activates group 2 innate lymphoid cells through chemoattractant receptor-homologous molecule expressed on TH2 cells. J Allergy Clin Immunol. 2014;133(4):1184–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Talbot S, Abdulnour RE, Burkett PR, Lee S, Cronin SJ, Pascal MA, et al. Silencing nociceptor neurons reduces allergic airway inflammation. Neuron. 2015;87(2):341–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour RE, Nyman J, Dionne D, et al. The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature. 2017;549(7672):351–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Wallrapp A, Burkett PR, Riesenfeld SJ, Kim SJ, Christian E, Abdulnour RE, et al. Calcitonin gene-related peptide negatively regulates alarmin-driven type 2 innate lymphoid cell responses. Immunity. 2019;51(4):709-723 e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Cao Y, Li Y, Wang X, Liu S, Zhang Y, Liu G, et al. Dopamine inhibits group 2 innate lymphoid cell-driven allergic lung inflammation by dampening mitochondrial activity. Immunity. 2023;56(2):320-335 e9.

    Article  CAS  PubMed  Google Scholar 

  151. Howard E, Hurrell BP, Helou DG, Shafiei-Jahani P, Hasiakos S, Painter J, et al. Orai inhibition modulates pulmonary ILC2 metabolism and alleviates airway hyperreactivity in murine and humanized models. Nat Commun. 2023;14(1):5989.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Hurrell BP, Sakano Y, Shen S, Helou DG, Li M, Shafiei-Jahani P, et al. Iron controls the development of airway hyperreactivity by regulating ILC2 metabolism and effector function. Sci Transl Med. 2024;16(746):eadk4728.

    Article  CAS  PubMed  Google Scholar 

  153. Thio CL, Chi PY, Lai AC, Chang YJ. Regulation of type 2 innate lymphoid cell-dependent airway hyperreactivity by butyrate. J Allergy Clin Immunol. 2018;142(6):1867-1883 e12.

    Article  CAS  PubMed  Google Scholar 

  154. Luo CH, Lai AC, Chang YJ. Butyrate inhibits Staphylococcus aureus-aggravated dermal IL-33 expression and skin inflammation through histone deacetylase inhibition. Front Immunol. 2023;14:1114699.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Lewis G, Wang B, Shafiei Jahani P, Hurrell BP, Banie H, Aleman Muench GR, et al. Dietary fiber-induced microbial short chain fatty acids suppress ILC2-dependent airway inflammation. Front Immunol. 2019;10:2051.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Karagiannis F, Masouleh SK, Wunderling K, Surendar J, Schmitt V, Kazakov A, et al. Lipid-droplet formation drives pathogenic group 2 innate lymphoid cells in airway inflammation. Immunity. 2020;52(4):620-634 e6.

    Article  CAS  PubMed  Google Scholar 

  157. Newman JC, Verdin E. Ketone bodies as signaling metabolites. Trends Endocrinol Metab. 2014;25(1):42–52.

    Article  CAS  PubMed  Google Scholar 

  158. Thio CL, Lai AC, Ting YT, Chi PY, Chang YJ. The ketone body beta-hydroxybutyrate mitigates ILC2-driven airway inflammation by regulating mast cell function. Cell Rep. 2022;40(13): 111437.

    Article  CAS  PubMed  Google Scholar 

  159. Halim TY, Hwang YY, Scanlon ST, Zaghouani H, Garbi N, Fallon PG, et al. Group 2 innate lymphoid cells license dendritic cells to potentiate memory TH2 cell responses. Nat Immunol. 2016;17(1):57–64.

    Article  CAS  PubMed  Google Scholar 

  160. Frech M, Omata Y, Schmalzl A, Wirtz S, Taher L, Schett G, et al. Btn2a2 regulates ILC2-T cell cross talk in type 2 immune responses. Front Immunol. 2022;13: 757436.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Halim TYF, Rana BMJ, Walker JA, Kerscher B, Knolle MD, Jolin HE, et al. Tissue-restricted adaptive type 2 immunity is orchestrated by expression of the costimulatory molecule OX40L on group 2 innate lymphoid cells. Immunity. 2018;48(6):1195-1207 e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Schwartz C, Khan AR, Floudas A, Saunders SP, Hams E, Rodewald HR, et al. ILC2s regulate adaptive Th2 cell functions via PD-L1 checkpoint control. J Exp Med. 2017;214(9):2507–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Stockis J, Yip T, Moreno-Vicente J, Burton O, Samarakoon Y, Schuijs MJ, et al. Cross-talk between ILC2 and Gata3(high) T(regs) locally constrains adaptive type 2 immunity. Sci Immunol. 2024;9(97):eadl1903.

    Article  CAS  PubMed  Google Scholar 

  164. Wang Y, Quan Y, He J, Chen S, Dong Z. SLAM-family receptors promote resolution of ILC2-mediated inflammation. Nat Commun. 2024;15(1):5056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Yamaguchi M, Samuchiwal SK, Quehenberger O, Boyce JA, Balestrieri B. Macrophages regulate lung ILC2 activation via Pla2g5-dependent mechanisms. Mucosal Immunol. 2018;11(3):615–26.

    Article  CAS  PubMed  Google Scholar 

  166. LeSuer WE, Kienzl M, Ochkur SI, Schicho R, Doyle AD, Wright BL, et al. Eosinophils promote effector functions of lung group 2 innate lymphoid cells in allergic airway inflammation in mice. J Allergy Clin Immunol. 2023;152(2):469-485 e10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Rigas D, Lewis G, Aron JL, Wang B, Banie H, Sankaranarayanan I, et al. Type 2 innate lymphoid cell suppression by regulatory T cells attenuates airway hyperreactivity and requires inducible T-cell costimulator-inducible T-cell costimulator ligand interaction. J Allergy Clin Immunol. 2017;139(5):1468-1477 e2.

    Article  CAS  PubMed  Google Scholar 

  168. Haworth O, Cernadas M, Levy BD. NK cells are effectors for resolvin E1 in the timely resolution of allergic airway inflammation. J Immunol. 2011;186(11):6129–35.

    Article  CAS  PubMed  Google Scholar 

  169. Simons B, Ferrini ME, Carvalho S, Bassett DJ, Jaffar Z, Roberts K. PGI2 controls pulmonary NK cells that prevent airway sensitization to house dust mite allergen. J Immunol. 2017;198(1):461–71.

    Article  CAS  PubMed  Google Scholar 

  170. Ferrini ME, Hong S, Stierle A, Stierle D, Stella N, Roberts K, et al. CB2 receptors regulate natural killer cells that limit allergic airway inflammation in a murine model of asthma. Allergy. 2017;72(6):937–47.

    Article  CAS  PubMed  Google Scholar 

  171. Lunding LP, Webering S, Vock C, Behrends J, Wagner C, Holscher C, et al. Poly(inosinic-cytidylic) acid-triggered exacerbation of experimental asthma depends on IL-17A produced by NK cells. J Immunol. 2015;194(12):5615–25.

    Article  CAS  PubMed  Google Scholar 

  172. Yang D, Li Y, Liu T, Yang L, He L, Huang T, et al. IL-1beta promotes IL-17A production of ILC3s to aggravate neutrophilic airway inflammation in mice. Immunology. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/imm.13644.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Ham J, Kim J, Choi S, Park J, Baek MG, Kim YC, et al. Interactions between NCR(+)ILC3s and the microbiome in the airways shape asthma severity. Immune Netw. 2021;21(4): e25.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Teng F, Tacho-Pinot R, Sung B, Farber DL, Worgall S, Hammad H, et al. ILC3s control airway inflammation by limiting T cell responses to allergens and microbes. Cell Rep. 2021;37(8): 110051.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Du Z, Huang L, Dai X, Yang D, Niu L, Miller H, et al. Progranulin regulates the development and function of NKT2 cells through EZH2 and PLZF. Cell Death Differ. 2022;29(10):1901–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Tumes D, Hirahara K, Papadopoulos M, Shinoda K, Onodera A, Kumagai J, et al. Ezh2 controls development of natural killer T cells, which cause spontaneous asthma-like pathology. J Allergy Clin Immunol. 2019;144(2):549-560 e10.

    Article  CAS  PubMed  Google Scholar 

  177. Koh J, Woo YD, Yoo HJ, Choi JP, Kim SH, Chang YS, et al. De novo fatty-acid synthesis protects invariant NKT cells from cell death, thereby promoting their homeostasis and pathogenic roles in airway hyperresponsiveness. Elife. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.87536.

    Article  PubMed  PubMed Central  Google Scholar 

  178. He Q, Liu L, Yang Q, Wang A, Chen S, Li R, et al. Invariant natural killer T cells promote immunogenic maturation of lung dendritic cells in mouse models of asthma. Am J Physiol Lung Cell Mol Physiol. 2017;313(6):L973–90.

    Article  PubMed  Google Scholar 

  179. Deng N, Chen Q, Guo X, Liu L, Chen S, Wang A, et al. Blockade of CD40L inhibits immunogenic maturation of lung dendritic cells: implications for the role of lung iNKT cells in mouse models of asthma. Mol Immunol. 2020;121:167–85.

    Article  CAS  PubMed  Google Scholar 

  180. Chuang YT, Leung K, Chang YJ, DeKruyff RH, Savage PB, Cruse R, et al. A natural killer T-cell subset that protects against airway hyperreactivity. J Allergy Clin Immunol. 2019;143(2):565-576 e7.

    Article  CAS  PubMed  Google Scholar 

  181. Thio CL, Lai AC, Wang JC, Chi PY, Chang YL, Ting YT, et al. Identification of a PD-L1+Tim-1+ iNKT subset that protects against fine particulate matter-induced airway inflammation. JCI Insight. 2022;7(23): e164157.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Feng X, Zhao C, Li L, Feng J, He W, Shi T, et al. iNKT cells with high PLZF expression are recruited into the lung via CCL21-CCR7 signaling to facilitate the development of asthma tolerance in mice. Eur J Immunol. 2021;51(2):414–32.

    Article  CAS  PubMed  Google Scholar 

  183. Caccamo N, Battistini L, Bonneville M, Poccia F, Fournie JJ, Meraviglia S, et al. CXCR5 identifies a subset of Vgamma9Vdelta2 T cells which secrete IL-4 and IL-10 and help B cells for antibody production. J Immunol. 2006;177(8):5290–5.

    Article  CAS  PubMed  Google Scholar 

  184. Qi Q, Xia M, Hu J, Hicks E, Iyer A, Xiong N, et al. Enhanced development of CD4+ gammadelta T cells in the absence of Itk results in elevated IgE production. Blood. 2009;114(3):564–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Huang Y, Jin N, Roark CL, Aydintug MK, Wands JM, Huang H, et al. The influence of IgE-enhancing and IgE-suppressive gammadelta T cells changes with exposure to inhaled ovalbumin. J Immunol. 2009;183(2):849–55.

    Article  CAS  PubMed  Google Scholar 

  186. Shimizu Y, Horigane-Konakai Y, Ishii Y, Sugimoto C, Wakao H. Mucosal-associated invariant T cells repress group 2 innate lymphoid cells in Alternaria alternata-induced model of allergic airway inflammation. Front Immunol. 2022;13:1005226.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Yu QN, Guo YB, Li X, Li CL, Tan WP, Fan XL, et al. ILC2 frequency and activity are inhibited by glucocorticoid treatment via STAT pathway in patients with asthma. Allergy. 2018;73(9):1860–70.

    Article  CAS  PubMed  Google Scholar 

  188. Rabe KF, Martinez FJ, Bhatt SP, Kawayama T, Cosio BG, Mroz RM, et al. AERIFY-1/2: two phase 3, randomised, controlled trials of itepekimab in former smokers with moderate-to-severe COPD. ERJ Open Res. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/23120541.00718-2023.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Wechsler ME, Ruddy MK, Pavord ID, Israel E, Rabe KF, Ford LB, et al. Efficacy and safety of itepekimab in patients with moderate-to-severe asthma. N Engl J Med. 2021;385(18):1656–68.

    Article  CAS  PubMed  Google Scholar 

  190. Kelsen SG, Agache IO, Soong W, Israel E, Chupp GL, Cheung DS, et al. Astegolimab (anti-ST2) efficacy and safety in adults with severe asthma: a randomized clinical trial. J Allergy Clin Immunol. 2021;148(3):790–8.

    Article  CAS  PubMed  Google Scholar 

  191. Maurer M, Cheung DS, Theess W, Yang X, Dolton M, Guttman A, et al. Phase 2 randomized clinical trial of astegolimab in patients with moderate to severe atopic dermatitis. J Allergy Clin Immunol. 2022;150(6):1517–24.

    Article  CAS  PubMed  Google Scholar 

  192. Reid F, Singh D, Albayaty M, Moate R, Jimenez E, Sadiq MW, et al. A Randomized phase I study of the anti-interleukin-33 antibody tozorakimab in healthy adults and patients with chronic obstructive pulmonary disease. Clin Pharmacol Ther. 2024;115(3):565–75.

    Article  CAS  PubMed  Google Scholar 

  193. Wilkinson T, De Soyza A, Carroll M, Chalmers JD, Crooks MG, Griffiths G, et al. A randomised phase 2a study to investigate the effects of blocking interleukin-33 with tozorakimab in patients hospitalised with COVID-19: ACCORD-2. ERJ Open Res. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/23120541.00249-2023.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Menzies-Gow A, Corren J, Bourdin A, Chupp G, Israel E, Wechsler ME, et al. Tezepelumab in adults and adolescents with severe, uncontrolled asthma. N Engl J Med. 2021;384(19):1800–9.

    Article  CAS  PubMed  Google Scholar 

  195. Corren J, Menzies-Gow A, Chupp G, Israel E, Korn S, Cook B, et al. Efficacy of tezepelumab in severe, uncontrolled asthma: pooled analysis of the PATHWAY and NAVIGATOR clinical trials. Am J Respir Crit Care Med. 2023;208(1):13–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Brightling CE, Nair P, Cousins DJ, Louis R, Singh D. Risankizumab in severe asthma—a phase 2a, placebo-controlled trial. N Engl J Med. 2021;385(18):1669–79.

    Article  CAS  PubMed  Google Scholar 

  197. Castro M, Corren J, Pavord ID, Maspero J, Wenzel S, Rabe KF, et al. Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N Engl J Med. 2018;378(26):2486–96.

    Article  CAS  PubMed  Google Scholar 

  198. Simpson EL, Bieber T, Guttman-Yassky E, Beck LA, Blauvelt A, Cork MJ, et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N Engl J Med. 2016;375(24):2335–48.

    Article  CAS  PubMed  Google Scholar 

  199. Bhatt SP, Rabe KF, Hanania NA, Vogelmeier CF, Bafadhel M, Christenson SA, et al. Dupilumab for COPD with blood eosinophil evidence of type 2 inflammation. N Engl J Med. 2024;390(24):2274–83.

    Article  CAS  PubMed  Google Scholar 

  200. Pavord ID, Chanez P, Criner GJ, Kerstjens HAM, Korn S, Lugogo N, et al. Mepolizumab for eosinophilic chronic obstructive pulmonary disease. N Engl J Med. 2017;377(17):1613–29.

    Article  CAS  PubMed  Google Scholar 

  201. Ortega HG, Liu MC, Pavord ID, Brusselle GG, FitzGerald JM, Chetta A, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med. 2014;371(13):1198–207.

    Article  PubMed  Google Scholar 

  202. Castro M, Zangrilli J, Wechsler ME, Bateman ED, Brusselle GG, Bardin P, et al. Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet Respir Med. 2015;3(5):355–66.

    Article  CAS  PubMed  Google Scholar 

  203. Ramakrishnan S, Russell REK, Mahmood HR, Krassowska K, Melhorn J, Mwasuku C, et al. Treating eosinophilic exacerbations of asthma and COPD with benralizumab (ABRA): a double-blind, double-dummy, active placebo-controlled randomised trial. Lancet Respir Med. 2025;13(1):59–68.

    Article  PubMed  Google Scholar 

  204. Nair P, O’Byrne PM. The interleukin-13 paradox in asthma: effective biology, ineffective biologicals. Eur Respir J. 2019;53(2):1802250.

    Article  CAS  PubMed  Google Scholar 

  205. Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, et al. Secukinumab in plaque psoriasis–results of two phase 3 trials. N Engl J Med. 2014;371(4):326–38.

    Article  PubMed  Google Scholar 

  206. Gordon KB, Blauvelt A, Papp KA, Langley RG, Luger T, Ohtsuki M, et al. Phase 3 trials of ixekizumab in moderate-to-severe plaque psoriasis. N Engl J Med. 2016;375(4):345–56.

    Article  CAS  PubMed  Google Scholar 

  207. Busse WW, Holgate S, Kerwin E, Chon Y, Feng J, Lin J, et al. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med. 2013;188(11):1294–302.

    Article  CAS  PubMed  Google Scholar 

  208. Moriyama S, Brestoff JR, Flamar AL, Moeller JB, Klose CSN, Rankin LC, et al. beta(2)-adrenergic receptor-mediated negative regulation of group 2 innate lymphoid cell responses. Science. 2018;359(6379):1056–61.

    Article  CAS  PubMed  Google Scholar 

  209. Matsuyama T, Machida K, Motomura Y, Takagi K, Doutake Y, Tanoue-Hamu A, et al. Long-acting muscarinic antagonist regulates group 2 innate lymphoid cell-dependent airway eosinophilic inflammation. Allergy. 2021;76(9):2785–96.

    Article  CAS  PubMed  Google Scholar 

  210. Chu C, Parkhurst CN, Zhang W, Zhou L, Yano H, Arifuzzaman M, et al. The ChAT-acetylcholine pathway promotes group 2 innate lymphoid cell responses and anti-helminth immunity. Sci Immunol. 2021;6(57):eabe3218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Salimi M, Stoger L, Liu W, Go S, Pavord I, Klenerman P, et al. Cysteinyl leukotriene E(4) activates human group 2 innate lymphoid cells and enhances the effect of prostaglandin D(2) and epithelial cytokines. J Allergy Clin Immunol. 2017;140(4):1090-1100 e11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Leff JA, Busse WW, Pearlman D, Bronsky EA, Kemp J, Hendeles L, et al. Montelukast, a leukotriene-receptor antagonist, for the treatment of mild asthma and exercise-induced bronchoconstriction. N Engl J Med. 1998;339(3):147–52.

    Article  CAS  PubMed  Google Scholar 

  213. Calhoun WJ. Summary of clinical trials with zafirlukast. Am J Respir Crit Care Med. 1998;157(6 Pt 1):S238–46.

    Article  PubMed  Google Scholar 

  214. Kim J, Ham J, Kang HR, Bae YS, Kim T, Kim HY. JAK3 inhibitor suppresses multipotent ILC2s and attenuates steroid-resistant asthma. Sci Adv. 2023;9(51):eadi3770.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Zhang X, Liu J, Li X, Zheng G, Wang T, Sun H, et al. Blocking the HIF-1alpha/glycolysis axis inhibits allergic airway inflammation by reducing ILC2 metabolism and function. Allergy. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/all.16361.

    Article  PubMed  Google Scholar 

  216. Albacker LA, Chaudhary V, Chang YJ, Kim HY, Chuang YT, Pichavant M, et al. Invariant natural killer T cells recognize a fungal glycosphingolipid that can induce airway hyperreactivity. Nat Med. 2013;19(10):1297–304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Meyer EH, Goya S, Akbari O, Berry GJ, Savage PB, Kronenberg M, et al. Glycolipid activation of invariant T cell receptor+ NK T cells is sufficient to induce airway hyperreactivity independent of conventional CD4+ T cells. Proc Natl Acad Sci U S A. 2006;103(8):2782–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Hachem P, Lisbonne M, Michel ML, Diem S, Roongapinun S, Lefort J, et al. Alpha-galactosylceramide-induced iNKT cells suppress experimental allergic asthma in sensitized mice: role of IFN-gamma. Eur J Immunol. 2005;35(10):2793–802.

    Article  CAS  PubMed  Google Scholar 

  219. Lai AC, Chi PY, Thio CL, Han YC, Kao HN, Hsieh HW, et al. alpha-lactosylceramide protects against iNKT-mediated murine airway hyperreactivity and liver injury through competitive inhibition of cd1d binding. Front Chem. 2019;7:811.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Gonzalez-Cuesta M, Lai AC, Chi PY, Hsu IL, Liu NT, Wu KC, et al. Serine-/cysteine-based sp(2)-iminoglycolipids as NOVEL TLR4 agonists: evaluation of their adjuvancy and immunotherapeutic properties in a murine model of asthma. J Med Chem. 2023;66(7):4768–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Lai AC, Gonzalez-Cuesta M, Ho CH, Chi PY, Wu KC, Rocha G, et al. alpha-GalCer sp(2)-iminoglycolipid analogs as CD1d-dependent iNKT modulators: Evaluation of their immunotherapeutic potential in murine models of asthma and autoimmune hepatitis. Eur J Med Chem. 2025;282: 117060.

    Article  CAS  PubMed  Google Scholar 

  222. Grund JC, Krammer S, Yang Z, Mitlander H, Rauh M, Zirlik S, et al. Vitamin D(3) resolved human and experimental asthma via B lymphocyte-induced maturation protein 1 in T cells and innate lymphoid cells. J Allergy Clin Immunol Glob. 2023;2(3): 100099.

    Article  PubMed  PubMed Central  Google Scholar 

  223. Hurrell BP, Shen S, Li X, Sakano Y, Kazemi MH, Quach C, et al. Piezo1 channels restrain ILC2s and regulate the development of airway hyperreactivity. J Exp Med. 2024;221(5): e20231835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Sakano Y, Sakano K, Hurrell BP, Helou DG, Shafiei-Jahani P, Kazemi MH, et al. Blocking CD226 regulates type 2 innate lymphoid cell effector function and alleviates airway hyperreactivity. J Allergy Clin Immunol. 2024;153(5):1406–22.

    Article  CAS  PubMed  Google Scholar 

  225. Sakano Y, Sakano K, Hurrell BP, Shafiei-Jahani P, Kazemi MH, Li X, et al. SIRPalpha engagement regulates ILC2 effector function and alleviates airway hyperreactivity via modulating energy metabolism. Cell Mol Immunol. 2024;21(10):1158–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by National Science and Technology Council (grant 111-2320-B-001-025-MY3 and 113-2320-B-001-018-MY3 to Y.J.C.), Academia Sinica Investigator Award (AS-IA-110-L04 to Y.J.C.) and Academia Sinica Grand Challenge Award (AS-GC-110-05 to Y.J.C) in Taiwan. All figures were created with Biorender.com.

Author information

Authors and Affiliations

Authors

Contributions

CLPT: conceptualization; writing—original draft; writing—review and editing. JSS: writing—original draft. CHL: writing—original draft. YJC: conceptualization, writing—review & editing; supervision; funding acquisition.

Corresponding author

Correspondence to Ya-Jen Chang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Thio, C.LP., Shao, JS., Luo, CH. et al. Decoding innate lymphoid cells and innate-like lymphocytes in asthma: pathways to mechanisms and therapies. J Biomed Sci 32, 48 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-025-01142-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12929-025-01142-w

Keywords