key: cord-0970389-0fu2s089 authors: Spits, Hergen; Mjösberg, Jenny title: Heterogeneity of type 2 innate lymphoid cells date: 2022-03-30 journal: Nat Rev Immunol DOI: 10.1038/s41577-022-00704-5 sha: 60475c8c9974a6a8ae94aa6384561d7d0a6716c5 doc_id: 970389 cord_uid: 0fu2s089 More than a decade ago, type 2 innate lymphoid cells (ILC2s) were discovered to be members of a family of innate immune cells consisting of five subsets that form a first line of defence against infections before the recruitment of adaptive immune cells. Initially, ILC2s were implicated in the early immune response to parasitic infections, but it is now clear that ILC2s are highly diverse and have crucial roles in the regulation of tissue homeostasis and repair. ILC2s can also regulate the functions of other type 2 immune cells, including T helper 2 cells, type 2 macrophages and eosinophils. Dysregulation of ILC2s contributes to type 2-mediated pathology in a wide variety of diseases, potentially making ILC2s attractive targets for therapeutic interventions. In this Review, we focus on the spectrum of ILC2 phenotypes that have been described across different tissues and disease states with an emphasis on human ILC2s. We discuss recent insights in ILC2 biology and suggest how this knowledge might be used for novel disease treatments and improved human health. Just over a decade has passed since the first extensive descriptions of type 2 innate lymphoid cells (ILC2s) in mice 1-3 (in 2010) and humans 4, 5 (in 2011), although there had been hints as to the existence of these cells in earlier papers from 2001 (ref. 6 ) and 2006 (ref. 7 ). At the time that ILC2s were discovered, an extensive body of knowledge on the biology of T helper 2 (T H 2) cells was already avail able. T H 2 cells and ILC2s have key transcription factors and cytokine production profiles in common, and these shared features were a major driver of research efforts that have propelled our understanding of the mecha nisms of ILC2 development and function. For some time, it was thought that ILC2s, in contrast to other ILC sub sets, were a homogeneous population of cells because, initially, no phenotypic or functional heterogeneity of ILC2s was noted, in contrast to the subsets of ILC1s and ILC3s that were identified at the time of their discov ery. However, progress in the past couple of years has made clear that ILC2s differ in phenotype and function between and within tissues 8 . Different tissue micro environments imprint the phenotypes and functions of ILC2s, which are, like other ILC subsets, highly plastic, thus facilitating adaptation to their microenvironment 9 . Furthermore, both in mice 10 and humans 11, 12 , regulatory, IL10 producing ILC2s have been identified that can counteract the activities of canonical ILC2s producing IL5 and/or IL13 (ref. 13 ). In addition, whereas ILC2s and the other ILC subsets were previously considered to be exclusively tissue resident, studies in mice 14 and humans 15 have shown that ILC2s can acquire the capac ity to migrate following activation, thereby contributing to systemic immunity. This Review discusses the diverse landscape of ILC2s that is emerging from these recent studies and the consequences of these new data for understanding the roles of ILC2s in cancer, metabolism, inflammation and infection. Initially, human ILC2s were defined by expression of CD127 (the IL7 receptor α subunit), CD161 (encoded by KLRB1) and CRTH2 (the receptor for prostaglandin D2) 4 . CRTH2 functionally regulates the migration of mouse ILC2s to the lung 16 . However, CRTH2 has proved less useful as a definitive marker for human ILC2s 17 because, as discussed below, human peripheral blood contains ILC2 like cells that lack CRTH2 expression 18, 19 . Table 1 shows the ILC2 populations in humans whose lineage identity has been thoroughly confirmed not using transiently expressed markers. In one study, it was observed that peripheral blood CD117 + CRTH2 -ILCs expressing another ILC2 associated marker, KLRG1, could upregulate CRTH2 expression in vitro following incubation with IL7, which suggests that these CD117 + CRTH2cells are precursors of ILC2s or immature CRTH2 -ILC2s 18 (Table 1; fig. 1a ). However, not all CRTH2 + ILC2s are functionally mature because CRTH2 + ILC2s expressing CD5 that are unable to pro duce type 2 cytokines were found in human cord blood and postnatal thymus 20 . Differentiation of CD5 + CRTH2 + ILC2s to cytokine producing ILC2s was accompanied by downregulation of CD5 expression 20 . Furthermore, CD5 is transiently expressed on ILC2s at an early stage during Heterogeneity of type 2 innate lymphoid cells their in vitro differentiation from CD34 + haematopoie tic stem cells in the presence of the Notch ligand Delta like 1 (refs 20,21 ). Together, these observations suggest that CD5 is a marker of functionally immature CRTH2 + ILC2s. Recently, it was found that CD5 + ILCs capable of differentiating to mature ILC2s are present in the intra vascular spaces of immunodeficient mice containing immune cells that have developed from human CD34 + haematopoietic precursors, and it was suggested in this study that CD5 + ILCs follow a developmental path distinct from CD5 -ILCs 22 . Whereas the CRTH2 -KLRG1 + ILC2s seem to include ILC2 precursors 18 , another study has identified ILC2s in human peripheral blood that lack expression of CRTH2 and/or CD127 but express multiple transcripts and proteins typically expressed by conventional mature CRTH2 + ILC2s, including the production of IL5 and IL13 (ref. 19 ). Although their lineage identity has not been precisely confirmed, ILC2s lacking CRTH2 and/or CD127 were enriched in the blood and bronchoalveo lar lavage (BAL) fluid of patients with asthma and could cause airway hyperreactivity when transferred to mice. Supporting these data, single cell RNA sequencing of human lung ILCs has shown the presence of CRTH2 -ILC2 like cells 8 . Such cells could be generated in vitro from blood CRTH2 + ILC2s upon alarmin stimulation, which suggests that lung alarmins induce the downreg ulation of CD127 and CRTH2, the latter likely via endog enous production of prostaglandin D2 (ref. 23 ). Hence, in addition to CRTH2, CD127 and CD117, other markers, including KLRG1, CD30, TNFR2 and CD200R1 (ref. 19 ), might be needed to appropriately assess ILC2 numbers and function in type 2 inflammatory settings. In sum mary, these studies show that ILC2s that lack CRTH2 expression may be either immature cells that have not yet gained CRTH2 expression or functionally mature cells that have downregulated CRTH2 as a result of activation. In addition to the described diversity of CRTH2 -ILC2s, more mature CRTH2 + ILC2s are also highly diverse. Recent data point to CD117 (stem cell factor receptor encoded by KIT) as a surface marker that distinguishes CRTH2 + ILC2s with different functions (Table 1; fig. 1a ). Circulating CRTH2 + CD117 -ILC2s have been described as cells that are committed to the ILC2 lineage with low propensity for transdifferentiation to ILC1s and ILC3s 24 . Expression of CD45RO, which is a well known marker for human memory T cells, seems to define activated CRTH2 + CD117 -ILC2s as they can be generated from ILC2s expressing CD45RA, which is also expressed on naive T cells, by stimulation with IL33 (ref. 15 ) ( fig. 1a ). Whereas a lack of CD117 expres sion seems to be a characteristic of several previously described populations of committed and activated CRTH2 + and CRTH2 -ILC2s 15, 18, 19, 24 , CRTH2 + CD117 + ILC2s have some features of ILC3s, including expression of the transcription factor RORγt (encoded by RORC) and the chemokine receptor CCR6 (refs 24,25 ) (Table 1) . Indeed, CD117 + ILC2s can produce IL17A when stim ulated with cytokines, such as IL23, that also strongly activate ILC3s but, in parallel, they mostly maintain their capacity to produce IL5 and IL13. CD117 + ILC2s are also prone to ILC1 like differentiation in the presence of IL1β and IL12 24 . These observations are in line with accumulating data that strongly suggest that ILC plas ticity can account for flexible ILC responses within a tis sue without the need for recruitment of ILCs from other tissue sources 11, [24] [25] [26] [27] [28] [29] [30] [31] . The plasticity of ILCs has recently been reviewed extensively 9,32 and information on the plasticity of ILC2s in particular is summarized in box 1. In summary, the peripheral blood of healthy human donors contains a spectrum of ILC2 phenotypes, ranging from immature to mature. KLRG1 + CRTH2immature ILC2s and CD5 + CRTH2 + ILC2s seem to be precursors of mature CRTH2 + ILC2s that have at least two phenotypes with distinct functionalities. Whereas CRTH2 + CD117 + ILC2s represent a plastic state with increased propensity for differentiation to an ILC1 like or ILC3 like pheno type, CRTH2 + CD117 -ILC2s are less plastic, being more committed to the ILC2 lineage, and some of these CD117 -ILC2s express CD45RO. CD45RO + ILC2s produce the highest levels of type 2 cytokines among all described ILC2 phenotypes, which indicates that they are in a (pre) activated state. Because ILC subsets have so many features in com mon with T cell subsets, it was long thought that there would be a FOXP3 expressing ILC subset as a coun terpart to FOXP3 + regulatory T (T reg ) Fig. 1 | The plasticity and migration of IlC2s generate phenotypic diversity in tissues. a | KLRG1 + immature type 2 innate lymphoid cells (ILC2s) in humans can generate IL-10-producing regulatory ILC2s as well as IL-5-producing and IL-13-producing ILC2s (CRTH2 + CD117 + ILC2s and CRTH2 + CD117 -ILC2s), depending on the tissue microenvironment and cytokine milieu. It is currently unknown if IL-10-producing ILC2s can lose the capacity for IL-10 production and become IL-5-producing and IL-13-producing ILC2s. Upon activation with IL-33 and thymic stromal lymphopoietin (TSLP), human ILC2s generate CD45RO + ILC2s that are similar to activated gut ILC2s in mice, expressing CD45RO, BATF, IRF4 and increased levels of IL-5 and IL-13. CD117 -ILC2s represent committed ILC2s, but it is currently unclear if such cells are the precursors of CD45RO + ILC2s or if CD45RO + ILC2s can revert to CD45RO -CD117 -ILC2s. ILC2s, preferentially those expressing high levels of CD117 , can generate ILC3-like cells producing IL-17 under the influence of IL-1β, IL-23 and transforming growth factorβ (TGFβ), or ILC1-like cells producing interferonγ (IFNγ) under the influence of IL-1β and IL-12. Such transdifferentiation is prevented, in both cases, by IL-4. It is currently unclear if ILC2-ILC1 plasticity involves an intermediate ILC3-like stage. b | In a mouse model of Nippostrongylus brasiliensis infection, IL-25-activated 'inflammatory' ILC2s in the gut can migrate via the lymphatic system and blood circulation to the lung in a sphingosine 1-phosphate (S1P)-dependent manner, adding to the pool of lung-resident ST2 + ILC2s. Gut-derived ILC2s can also generate ILC3-like cells in the lung in response to type 3 cytokines such as IL-6, IL-23 and TGFβ. Gut-derived ILC2s from the lung likely return to the gut, as such cells cannot be found in the lung after parasite clearance. Additionally, lung-derived, activated ST2 + ILC2s can leave the lung and enter the circulation but it is so far unclear where such cells migrate. IL-10-producing ILC2s can be generated in both the gut and lung but the migratory behaviour of these cells is still unclear. The migratory pattern of human ILC2s is unclear but data support the recirculation of ILC2s between blood and lung in individuals with asthma. NMU, neuromedin U; RA, retinoic acid. pool, regulatory activities have been associated with IL10 production, which was indeed observed in one of the early reports of mouse ILC2s 1 . More recent work in mice has shown that IL10 producing ILC2s can be generated following activation with alarmins, such as IL33, and retinoic acid ( Table 2; fig. 1a ) and that these cells counteract the recruitment of eosinophils in the lung mediated by canonical ILC2s 10 . IL10 producing ILC2s were also found in the mouse intestine, where factors, including IL2, IL4, IL10 and the neuropep tide neuromedin U, could increase IL10 production, whereas TL1A (encoded by TNFSF15) suppressed this 34 . IL10 producing ILC2s generated following activation with IL33 and retinoic acid have now also been iden tified in human peripheral blood and the nasal polyps of patients with chronic rhinosinusitis with nasal polyps (CRSwNP) 12, 13 (Table 1 ). IL10 producing ILC2s from the inflamed nasal tissue of patients with CRSwNP 12 could inhibit the activation of IL5 producing ILC2s and attenuate cytokine production by T H 2 cells 12, 13 . IL10 producing ILC2s in humans are derived from immature (CRTH2 -) and mature (CRTH2 + ) ILC2s expressing KLRG1, as KLRG1 -CRTH2 + ILC2s, which are yet to be properly defined, failed to produce IL10 under stimulation with IL2, IL7, IL33 and retinoic acid 13 . Notably, circulating KLRG1 + ILC2s from individu als with allergic disease were unable to produce IL10, but the capacity of ILC2s to produce IL10 was restored following successful allergen immunotherapy, which suggests that human IL10 producing ILC2s might be involved in the control of allergic responses 13, 35, 36 . However, it has not yet been confirmed whether human IL10 producing ILC2s reside in lung and intestine as well as nasal tissue as is the case in mice. It is also unclear if IL10 production is a feature of a subset of ILC2s or is part of an effector programme that functions to provide negative feedback during activation. How the differen tiation of canonical ILC2s into IL10 producing ILC2s is regulated is just beginning to be unravelled. Recent research has shown that BLIMP1 (encoded by PRDM1) and MAF regulate IL10 production by ILC2s in mice 37 ( Table 2 ) but whether this is also the case in human ILC2s remains to be determined. A landmark paper in 2015 using adult parabiotic mice showed that ILCs, including ILC2s, are stably tissue resident in lung, small intestine and mesenteric lymph nodes over a period of several months, both during homeostatic conditions and in the presence of sys temic inflammation in FOXP3 deficient mice, which lack inflammation inhibiting T reg cells 38 . However, dur ing the chronic phase of Nippostrongylus brasiliensis infection in these parabiotic mice, a fraction of the ILC2 population was found to be recruited from the donor mouse, which, together with local proliferation of host derived ILC2s, generated an enlarged pool of ILC2s for clearance of the infection and tissue healing 38 . Since then, multiple reports have used the gut-lung axis dependent immunity model of N. brasiliensis infection to study ILC2 migration between tissues. This model involves a robust type 2 immune response in both the gut and lung owing to the dynamics of the parasite, which initially infects the lung, then later infects the gut after having been expelled from the lung through cough ing and swallowing. Using this model, studies support a migratory behaviour of gut ILC2s that are activated by the unique tissue microenvironment during N. brasiliensis infection 14, 39 . Gut ILC2s, which express the IL25 receptor component IL17RB (Table 2) , are activated by IL25. Activated gut ILC2s (also referred to as 'inflam matory' ILC2s) then leave the gut in a sphingosine 1 phosphate receptor 1 (S1PR1) dependent manner and migrate to the lungs to support a type 2 immune response in lung tissue, likely via migratory mechanisms involving the integrin LFA1 (ref. 40 ) ( fig. 1b) . Some, if only a small proportion, of the gut ILC2s that enter the lungs upregulate expression of the IL33 receptor ST2 (also known as IL1RL1), thereby taking on the phenotype of lung resident ILC2s (also referred to as 'natural' ILC2s) ( Table 2) . It is currently unclear if gut ILC2s that enter the lung during N. brasiliensis infection also upregulate neuropilin 1 (NRP1), which was recently shown to be an activating receptor specifically expressed by lung ILC2s and upregulated on intestinal ILC2s following transfer to the lungs in mice 41 . In addition to the influx of gut ILC2s, recruited and tissue resident precursors of ILC2s also contribute to the local population of ILC2s in the lung and have a role in the dynamic diversity of ILC2s observed in the lung during N. brasiliensis infection 42 . Whereas some gut derived ST2 + ILC2s stay in the lung, some return to the gut; they enter the circula tion together with activated lung derived ST2 + ILC2s 14,39 , using S1PR1 to exit the lung 39 (fig. 1b ). Gut ILC2s acquire migratory features early in N. brasiliensis infec tion, whereas lung ST2 + ILC2s become migratory at Plasticity can be defined as the capacity of cells to change their destiny during differentiation as well as the capacity of differentiated, functionally mature cells to change their functions and phenotype. Several immune cell types, including T cells 108 and macrophage subsets 109 , have been shown to be capable of plasticity. recent reviews have discussed the plasticity of innate lymphoid cells (iLCs) in general 9, 32 . Concerning type 2 iLCs (iLC2s) in particular, the Lineage - precursor cells that are found in human peripheral blood 18 are transcriptionally more similar to mature iLC2s than to iLC1s or iLC3s, which indicates that they are biased towards the iLC2 lineage. regardless, these iLC2 precursors not only give rise to CrtH2 + iLC2s but also to iLC1-like or iLC3-like cells in the presence of iL-1β and iL-12 or iL-23, respectively. several studies, both in humans and mice, have shown that functionally mature iLC2s are also able to change their cytokine production profile and phenotype in the context of inflammation. Longitudinal single-cell rNa sequencing and assay for transposase-accessible chromatin (ataC) sequencing, together with in vivo fate mapping experiments, have revealed the kinetics and mechanisms of how mouse skin-resident iLCs, including effector iLC2s, acquire iLC3-like features upon induction of iL-23-induced psoriasis-like inflammation 91 . similarly, in humans, skin-resident iLC2s can transition into iLC3-like cells in psoriasis 25 . Moreover, in humans, the transition of iLC2s into iLC1-like cells has been observed in chronic obstructive pulmonary disease 26, 31 and in Crohn's disease 29 . This ILC1-like differentiation process can be mimicked in vitro by the combination of iL-1β and iL-12. Mechanistically, iL-1β induces low levels of expression of the transcription factor t-bet and of iL-12 receptor-β2 on ILC2s, which enables iL-12 to stimulate the conversion of these cells into iLC1s 30 . as iLC2 plasticity seems to be prominent in the context of inflammatory diseases, it may also have a role in other diseases such as cancer, emphasizing the need for further studies in both mouse models and humans. later stages of infection 14, 39 , together suggesting that tissue specific type 2 immunity can be systemically dis seminated as has also been shown recently by others 43 . Further studies will be required to confirm whether ILC2s that migrate from tissues upon local activation indeed contribute to systemic type 2 immunity and to address whether these ILC2s have an increased propen sity for returning to their tissue of origin as has been described for tissue resident T cells 44 . The clinical translation of these findings from mice is complicated by the markedly different distri bution of ILC2s in mice and humans. Humans have IL25 responsive and IL33 responsive, IL13 producing ILC2s in peripheral blood at steady state 4 . By contrast, naive mice, possibly as a result of being kept under spe cific pathogen free or germ free conditions, lack cir culating ILC2s, which are present in peripheral blood only after type 2 stimulation in tissues such as during N. brasiliensis infection as discussed above 39 . Further more, ILC2s are a prominent ILC population in the mouse intestine 14 , whereas CD127 + ILC2s in the intes tine of adult humans are mostly undetectable 8 unless it is severely inflamed 45 . Gut ILC2s increase in num ber in antibiotic treated mice 46 , and thus it is thought that the gut microbiota might suppress ILC2 num bers in human gut; indeed, CRTH2 + ILC2s can be found in human fetal gut 4 and ST2 + ILCs were identi fied in human paediatric gut 47 . However, there are also fundamental differences in the development of innate and adaptive lymphocytes in mice and humans that could explain the divergent distribution of ILC2s in the two species. In mice, fully mature T cells do not appear until the postnatal period 48 , by which time intestinal ILC2s are being maintained and activated through a tuft cell-ILC2 circuit 49 . By contrast, in humans, naive T cells populate the thymus already at 6-9 weeks post concep tion, memory T cells are found in the gut at 14-21 weeks post conception 50 and T cells are the dominant lympho cyte population in the intestine at birth 51 . This raises the possibility that human intestinal ILC2s are regulated not only by microbial factors but also by adaptive lympho cytes, which might suppress intestinal ILC2 numbers by the time of birth. In support of this, it has been reported that ILC3s are inhibited by both regulatory and effec tor CD4 + T cells following weaning in mice 52 . It is also possible that ILC2s are present in human adult gut but at sites that have so far been difficult to sample, or that they lack expression of canonical ILC markers, such as CD127, as has been reported for blood and lung ILC2s 19 , thereby complicating their identification. Hence, the dynamics, regulation, phenotype, location and role of gut ILC2s during the human lifespan require further research. Assessment of BAL fluid of individuals with asthma stimulated with lung allergens provides a unique oppor tunity to gain insight into the kinetics of ILC2 compo sition in human tissues in a clinically relevant setting. Indeed, ILC2s that express ST2 (ref. 53 ) and CRTH2 (ref. 54 ) accumulate in BAL fluid within the first 24 h of allergen challenge. Interestingly, CRTH2 + ILC2s in BAL fluid have increased levels of IL17RB and IL1RL1 (encoding ST2) transcripts as compared to peripheral blood ILC2s 54 . Further analysis revealed that ILC2s in BAL fluid also have increased expression of BATF and IRF4 (ref. 54 ), pointing towards a phenotype equivalent to that of gut ILC2s identified in mice (Table 2) , which can migrate to the lung and acquire ST2 expression 15, 55 ( fig. 1b) . As the accumulation of ILC2s in BAL fluid occurred within 24 h of allergen challenge, likely preceding local proliferation of ILC2s, and correlated with the depletion of peripheral blood ILC2s 54 , it is tempting to speculate that human ILC2s can leave the circulation and enter the lung upon allergen challenge, which is supported by mouse data 14 (fig. 1b) . In line with this, exposure of human peripheral blood ILC2s to alarmins leads to marked phenotypic changes, includ ing increased protein expression of IL17RB and ST2 and decreased expression of CRTH2, which is similar to the phenotype of lung ILC2s both at homeostasis 8 and during allergic inflammation 54 . Of note, similar results were recently obtained in patients with CRSwNP, where it was shown that peripheral blood CD45RA + ILC2s exposed to alarmins undergo conversion to CD45RO + activated 'inflammatory' ILC2 like cells similar to those found in nasal polyps 15 . In addition, as discussed above, CD45RO + cells that have a transcrip tional profile similar to activated and migratory gut ILC2s in mice 15 are found in the circulation of patients with asthma, particularly in steroid resistant disease 15 . Consistent with this, CD45RO + ILC2s are significantly more steroid resistant than CD45RA + ILC2s 15 . Hence, such data support a model in which circulating human CRTH2 + CD45RA + ILC2s that enter type 2 inflamed tissues convert to CD45RO + ILC2 like cells that pro duce large amounts of IL13 and IL5, thereby contrib uting to type 2 pathophysiology. Such CD45RO + ILC2s in humans can later migrate out of inflamed tissues, thereby re entering the circulation and potentially contributing to systemic type 2 immunity as observed in mice 39 (fig. 1b) . However, alternative explanations for the enlarged population of ILC2s seen in asthmatic lungs and nasal tissue from patients with CRSwNP are possible. In BAL fluid, ILC2s may be mobilized from peribron chial areas, which contain IL17RB + ST2 + ILC2s even during homeostasis 8 . Indeed, using intravital micros copy in live mice, ILC2s are seen to have significant CCR8-CCL1 mediated ameboid mobility and preferen tially localize in perivascular and peribronchial spaces upon intranasal IL33 administration 56 . In summary, studies of ILC2s in mice indicate that, under steady state conditions, these cells are tissue resi dent, whereas ILC2s activated by cytokines in the local tis sue environment, such as that induced by N. brasiliensis, can acquire migratory capacities. In humans, ILC2s can be found in the circulation of both healthy individuals and patients with type 2 inflammation but it is currently unclear from which tissues such cells derive and whether populations of tissue resident ILC2s exist. As in mice, human ILC2s can adapt to their microenvironment to generate a diverse repertoire of cells (Table 1; fig. 1 ), with recent studies suggesting that ILC2s have important functions in an increasing variety of disease types, as we discuss below. Previous studies have shown a role for ILC2s in both tumour promotion and tumour rejection, which have been comprehensively summarized elsewhere 57 . However, over the past 2 years, considerable progress has been made in the understanding of how specific pop ulations of ILC2s contribute to immunosurveillance in different types of cancer [58] [59] [60] [61] [62] (fig. 2 ). Most of these mech anistic insights have been generated in mouse models, with supporting but still limited data derived from human cancer tissues. In two studies of human hepatocellular carcinoma (HCC), numbers of intratumoural ILC2s were shown to be increased whereas levels of the ILC2 promoting cytokine IL33 were decreased 58, 59 . In a mouse model of HCC, the reduced level of IL33 was shown to correlate with expansion of a population of KLRG1 -ILC2s with low levels of expression of ST2. To replace the func tion of IL33 signalling through ST2, these cells relied on the lack of KLRG1 mediated inhibition to promote the production of IL13 and of the chemokines CXCL2 and CXCL8; this led to the chemoattraction of neutro phils that, under the influence of IL13, produced the immunosuppressive factor arginase 1 (ARG1), thereby reducing T cell responses and promoting tumour growth 58 (fig. 2a) . Therefore, the authors suggest that the presence of KLRG1 -ILC2s in HCC correlates with reduced survival. However, the other study showed that high intratumoural levels of IL33 and high frequencies of ILC2s are associated with better survival in patients with HCC 59 . Stratifying patients according to their ILC2 to ILC1 ratio revealed that only those patients with increased ILC2s relative to ILC1s had a KLRG1 + ILC2 phenotype driven by IL33 and associated with better survival. Patients with decreased numbers of ILC2s relative to ILC1s had an ILC2 phenotype with low lev els of KLRG1 and high levels of expression of CXCL8, reminiscent of the KLRG1 -CXCL8 producing ILC2s described in the mouse study 58 . This raises the possibility that the two phenotypes of KLRG1and KLRG1 + ILC2s might have opposing functions in HCC, with KLRG1 -ILC2s being pro tumorigenic and KLRG1 + ILC2s being anti tumorigenic (fig. 2a) . Although both HCC studies show the therapeutic potential of targeting ILC2s, we urgently need to better understand the conditions that regulate KLRG1 + versus KLRG1 -ILC2s in this type of cancer and how to selectively target them. The role of the IL33-ILC2 axis in promoting tumour rejection has also been demonstrated in a mouse model of melanoma 60 (fig. 2b ), in which it was shown that IL33 induced ILC2s produce granulocyte-macrophage colony stimulating factor (GMCSF) and thus attract eosinophils to the tumour site, which are crucial for tumour rejection. Unleashing the full capacity of ILC2s through anti PD1 checkpoint blockade further enhanced tumour rejection in this model, involving the recruitment of eosinophils and CD103 + dendritic cells, and the priming of antitumour CD8 + T cells. In humans with melanoma, an intratumoural type 2 gene signature, including KLRG1 expression, was associated with better survival 60 . Similarly, in patients with pan creatic cancer, ILC2 accumulation and IL33 levels are associated with increased survival 61 (fig. 2c) . In a mouse model of pancreatic cancer, IL33 induced expansion of ILC2 populations also led to CD103 + dendritic cell recruitment through CCL5 production and subsequent CD8 + T cell priming. In this model, ILC2s were impor tant for optimal tumour rejection, which was further enhanced by anti PD1 therapy that not only unleashed CD8 + T cell mediated antitumour immunity but also prevented the intrinsic PD1 dependent inhibition of ILC2s 61 (fig. 2c ). These exciting data suggest additional, non T cell driven mechanisms underlying the antitu mour effects of PD1 checkpoint blockade, which pre sents novel opportunities to increase the efficacy of this therapy. The role of ILC2s in colorectal cancer (CRC) is com plex (fig. 2d ). Whereas the total number of ILC2s is low and unaffected in colon tumours of humans 63, 64 , the frequency of intratumoural IL13 + ILC2s is increased in humans 64 and mice 65 with CRC. A recent study high lighted the role of the peroxisome proliferator activated receptor γ (PPARγ) pathway in ILC2s in a mouse model of CRC 64 . Among ILCs, PPARγ is selectively expressed by ILC2s, and in a mouse model of CRC, deletion of PPARγ in ID2 expressing cells, which includes ILC2s (as well as other ILC subsets), reduced the tumour bur den and increased survival. Although the exact mecha nisms were not explored, the pro tumorigenic effects of ILC2s have previously been linked to creating a type 2 cytokine driven immunosuppressive tumour microen vironment involving myeloid derived suppressor cells, T reg cells and the inhibition of natural killer cells [66] [67] [68] . As PPARγ was shown to directly regulate PD1 expression in ILC2s 69 , it is possible that PPARγ inhibition results in www.nature.com/nri 0123456789();: a subset of PD1 -ILC2s with antitumour effects. Indeed, PD1 + ILC2s accumulate in human CRC tumours, and mice engrafted with PD1 + ILC2 containing tumours had, compared with tumours not containing PD1 + ILC2s, an increased tumour mass, which was decreased upon PD1 blockade 70 . By contrast, using the same mouse model of colitis associated CRC, another study showed that selective deletion of ILC2s increased tumour burden and reduced survival 65 . Hence, whereas PD1 + ILC2s seem to be a tumour promoting subset in CRC, the effect of the total ILC2 pool may be anti tumorigenic as it is associated with improved survival 65 . In summary, subsets of ILC2s can have opposing roles within and across different cancers. Notably, whereas the role of IL10 producing T reg cells in sup pressing antitumour immunity is well understood, a role Fig. 2 | IlC2s can contribute to tumour rejection or tumour growth. In mouse models of hepatocellular carcinoma (part a), loss of KLRG1-mediated inhibition in a population of KLRG1type 2 innate lymphoid cells (ILC2s) results in the production of IL-13 as well as of the chemokines CXCL2 and CXCL8. CXCL2 and CXCL8 recruit neutrophils to the tumour site, which, under the influence of IL-13, produce the immunosuppressive factor arginase 1 (ARG1). ARG1 suppresses T cell responses, ultimately promoting tumour growth. In mouse models of melanoma (part b) and pancreatic cancer (part c), IL-33 induces ILC2s that subsequently activate CD103 + dendritic cells (DCs) to prime antitumour CD8 + T cells. In the case of melanoma, the production of granulocyte-macrophage colony-stimulating factor (GM-CSF) by ILC2s also recruits eosinophils to promote tumour rejection. Anti-PD1 therapy blocks cell-intrinsic inhibition of CD8 + T cells and ILC2s through the checkpoint molecule PD1 to further enhance tumour rejection. In mouse models of colorectal cancer (part d), PD1 + ILC2s enhance tumour growth, for example through the inhibition of natural killer (NK) cell responses and enhancing the responses of regulatory T (T reg ) cells and myeloid-derived suppressor cells (MDSCs). Inhibition of PD1 signalling in ILC2s, through either pharmacological or genetic inhibition of the peroxisome proliferator-activated receptorγ (PPARγ) pathway (which reduces PD1 expression) or anti-PD1 therapy promotes tumour rejection in colorectal cancer. These mechanistic insights from mouse models are supported by data derived from human cancer tissues (see inset schematic graphs). Genome-wide transcriptional data indicative of high intratumoural levels of IL33 (in hepatocellular carcinoma (part a) and pancreatic cancer (part c)) or high levels of ILC2-associated gene expression (in melanoma (part b) and colorectal cancer (part d)) predict better survival in humans. Nature reviews | Immunology 0123456789();: for IL10 producing ILC2s in antitumour immunity has yet to be explored. In mice, ILC2s in visceral adipose tissue are ST2 + ARG1 + cells and are transcriptionally, functionally and develop mentally closely related to ILC2s found in the lung 71, 72 ( Table 2 ). Accumulating data in mice point to activated ILC2s as being regulators of adipose tissue homeosta sis, preventing adiposity and promoting beiging of white adipose tissue (WAT) into thermogenic brown fat [73] [74] [75] . In support of these findings, ILC2s in human adipose tis sue are also ST2 + and are reduced in number in adipose tissue from patients with obesity 73 . More recent mouse studies suggest that this function of ILC2s in adipose tis sue homeostasis, and the development of adipose tissue ILC2s 76 , are regulated in particular stromal cell niches, of which at least some are under neuronal control (fig. 3 ). In a model of cold induced beiging of WAT, it was shown that the increased number of ILC2s depended on IL33 produced by a subset of mesenchymal adipose progenitor cells 77 . Mesenchymal adipose progenitor cells responded to β adrenergic receptor signalling via cAMP response element binding protein (CREB), which links neuronal stimulation to adipose tissue plasticity via ILC2s. In line with these observations, it was shown that the sympa thetic nervous system controls mesenchymal adipose progenitor cells through the release of noradrenaline, which acts on β adrenergic receptors on these cells. In turn, these cells produce glial derived neurotropic factor, the receptor for which, RET, is particularly highly expressed on visceral adipose tissue ILC2s. This inter action leads to the production of type 2 cytokines and enkephalins by ILC2s, which promotes beiging of WAT and counteracts the development of obesity following a high fat diet 78 . In addition, it was recently shown that ILC2s regulate the downstream effects of obesity, namely metabolic syndrome, limiting both the onset of insulin resistance and established insulin resistance in a mouse model of obesity 79 . These preclinical data now warrant exploration in humans to determine the clinical relevance. Since their discovery, ILC2s have been shown to medi ate airway inflammation in several mouse models of asthma, and increasing data correlate ILC2 numbers and function with asthma severity in humans (recently excel lently reviewed elsewhere 80 ) . Recent data in mice [81] [82] [83] and humans 84 point towards metabolic factors that influ ence the role of ILC2s in lung homeostasis 84 and airway inflammation [81] [82] [83] . For example, glucose dependent fatty acid uptake and metabolism in lung ILC2s in mice have been shown to fuel airway inflammation through ILC2 proliferation and cytokine production, which could be prevented by a ketogenic diet 82 . In parallel, the short chain fatty acid butyrate has been shown to inhibit ILC2 func tions and ameliorate airway inflammation in mice 83 . Furthermore, bilirubin, the catabolic end product of haem, suppresses ILC2 function and airway inflamma tion in a mouse model of asthma, and human infants with hyperbilirubinaemia have reduced circulating ILC2 frequencies and cytokine production 81 . Hence, ILC2s are clearly not only involved in regulating whole body metabolism through effects on adipose tissue homeo stasis but are also themselves regulated by metabolic factors at a cellular level. The details of how metabolism regulates human ILC2s are beginning to be unravelled 84 , showing a crucial role for oxidative phosphorylation in the maintenance and proliferation of ILC2s, whereas gly colysis is crucial for optimal type 2 cytokine production by ILC2s. Recent studies suggest that neuronal factors also regulate ILC2 functions in the lung in mouse models of airway inflammation. Although this aspect is still under explored in humans, ILC2s in mice express the neuro peptide calcitonin gene related peptide (CGRP; also known as CALCA) and its receptor, which inhibit ILC2 function in vitro 85 . In a mouse model of airway inflam mation induced by intranasal administration of IL33, CGRP receptor deficiency caused worsening of inflam mation, indicating that CGRP might have a role in limiting ILC2 responsiveness in the lung. By contrast, IL25 induced ILC2s in the lung express the neuromedin U receptor (NMUR1) and NMUR1 signalling promotes house dust mite induced airway inflammation 86 . Hence, IL33 induced and IL25 induced lung ILC2s seem to differ in their responsiveness to neuronal factors. In fact, in helminth infection, CGRP finetunes ILC2 responses induced by IL33 and neuromedin U to limit IL13 In mouse adipose tissue, sympathetic nerves and alternatively activated, M2-type macrophages produce noradrenaline that binds β-adrenergic receptors on mesenchymal adipose progenitor cells. This leads to their production of IL-33 and glial cell-derived neurotrophic factor (GDNF), for which the receptors ST2 and RET, respectively, are expressed on adipose tissue type 2 innate lymphoid cells (ILC2s). Activation of adipose tissue ILC2s leads to IL-4 and IL-13 production, which act on mesenchymal adipose cells to promote their production of the eosinophil chemotactic factor CCL11 (also known as eotaxin). Together with ILC2-derived IL-5, this recruits eosinophils to adipose tissue and supports their maintenance. Eosinophils produce IL-4 that supports the differentiation of alternatively activated macrophages, thereby creating a positive-feedback loop for a type 2 environment in the adipose tissue. Adipose tissue ILC2 activation also leads to the production of Met-Enkephalin (MetEnk), which acts on opioid receptors on adipocytes, resulting in a process known as beiging that reduces insulin resistance, a component of the metabolic syndrome. The process by which white adipose tissue is converted to a phenotype more similar to that of brown adipose tissue, thereby increasing energy expenditure by reducing lipids stored within the adipose tissue. Enkephalins small peptides that function as neurotransmitters and that bind to opioid receptors. Metabolic syndrome a combination of conditions, including high blood pressure, high blood glucose levels, high levels of waist body fat, and high cholesterol or triglyceride levels, that predisposes for development of heart disease and type 2 diabetes. Dietary intake of high levels of fat and low levels of carbohydrate. www.nature.com/nri 0123456789();: production while maintaining IL5 production, thereby controlling the magnitude of the ILC2 response 87 . In a mouse model of ovalbumin induced airway inflamma tion, CGRP produced by pulmonary neuroendocrine cells induced IL5 production by ILC2s and promoted inflammation 88 . Although IL13 production was not assessed in this study, based on the helminth infection model, CGRP seems to function as a rheostat of lung ILC2 function, which is regulated by an intricate neu roendocrine network of factors with both activating and inhibitory functions. Studies addressing these aspects in human airway inflammation are now eagerly awaited. In mice, skin ILC2s have a unique phenotype as com pared to those found in lung, adipose tissue and gut, with low levels of expression of ST2 and IL17RB and high levels of expression of the IL18 receptor 71 . In healthy skin of mice, ILC2s provide a homeostatic source of IL13 that, independently of alarmins, con tributes to the differentiation of skin CD11b low type 2 conventional dendritic cells. Upon dermal allergen administration, the presence of these dendritic cells underpins a preferential T H 2 cell response, which is not seen following allergen challenge in the lung 89 . Indeed, atopic dermatitis is associated with an accu mulation of ILC2s, which enhance allergen induced type 2 responses, in both mice and humans 90 . However, the downstream and translational consequences of the type 2 response skewing in skin compared with other tissues remain to be explored in more detail. To the best of our knowledge, there is no evidence for a migratory behaviour of skin ILC2s in mice, unlike that reported for gut and lung ILC2s 14, 39 . Instead, the data support a model whereby flexible ILC2 responses in the skin are achieved through the plasticity of skin resident ILC2s without the need for recruitment of ILCs from other tissue sources. IL23 induced psoriasis like skin inflammation in mice was shown to be independent of the recruitment of cir culating ILCs 91 . Instead, inflammation was mediated by IL23 driven conversion of homeostatic skin ILC2s into a pathogenic ILC3 like state (box 1). These data are supported by a recent single cell RNA sequencing study of human psoriatic skin, which revealed a spectrum of ILCs with mixed features of ILC2s and ILC3s 92 . By con trast, in humans, psoriasis is associated not only with enrichment of dermal IL17 + IL22 + ILC3s 25 but also with an increase in the number of circulating ILC3s 93 and skin homing RORγt + CD117 + ILC2s 25 , which sug gests that progenitors of ILC3s migrate to psoriatic skin and that mature ILC3s are extruded from psori atic skin. Further work will determine whether this flux of skin ILCs in humans can be modelled in mice, enabling the study of the mechanisms involved. One of the first reports of a role of ILC2s in the airways showed that ILC2s contribute to amphiregulin mediated lung tissue repair following influenza virus infection in an IL33 dependent manner 5 . There were also early reports of the depletion of ILCs in individuals infected with HIV1, and it was suggested that such depletion could contribute to the breakdown of gut mucosal bar rier function seen in persons living with HIV1 (ref. 94 ). However, in addition to contributing to tissue repair in viral infection, ILC2s have also been implicated in driving viral pathogenesis and exacerbating chronic lung disease. In mice, respiratory syncytial virus (RSV) infection triggers IL33 production from airway epi thelia, which in turn induces ILC2s to produce IL13 and promotes airway inflammation 95 , through effects on both stromal and immune cells, including goblet cell hyperplasia, B cell IgE isotype switching and the polari zation of alternatively activated, M2 type macrophages. A recent report identified the uric acid induced activa tion of the NLRP3 inflammasome in epithelial cells as being crucial for this process. Inhibition of this path way in RSV infection leads to a reduction not only of uric acid induced IL1β production by myeloid cells but also IL33 production by epithelial cells and, sub sequently, a reduction in the number of ILC2s in the airways of mice 96 (fig. 4a ). Strikingly, a translational study recently revealed the accumulation of ILC2s and the type 2 cytokines IL33, IL4 and IL13 in nasal aspi rates of infants with RSV infection with severe disease as compared to moderate disease 97 . These findings provide clinical relevance to the role of ILC2s in driv ing RSV pathogenesis observed in mice, and suggest that RSV infection of infants might prime for subsequent development of lung diseases later in life 98 . Similarly to RSV infection, rhinovirus infection in mice is also associated with excessive ILC2 activation 99 , thereby contributing to asthma exacerbations ( fig. 4b) . The translational relevance of these findings was recently confirmed as an increased ratio of ILC2s to ILC1s was found in bronchial biopsy samples of individuals with asthma versus without asthma after rhinovirus infection 100 . Of note, the increased ILC2 to ILC1 ratio in individuals with asthma was associated with greater severity of rhinovirus infection, including reduced lung function and increased respiratory symptoms. As ILC2s clearly have a role in respiratory virus infec tions, the recent emergence of severe acute respiratory syndrome coronavirus 2 (SARS CoV2), the causative agent of coronavirus disease 2019 (COVID19), has raised the question of whether ILC2s participate in this disease process (box 2; fig. 4c ). Although there is still little evidence for a role of ILC2s in responding to bacterial infections in humans, recent studies suggest that ILC2s contribute to anti bacterial immunity in mice, specifically against the commensal stomach microbiota as well as the patho gen Helicobacter pylori 101 . Surprisingly, in contrast to IL17RB + 'inflammatory' ILC2s in the small intestine of mice, ILC2s in the stomach are ST2 + and resemble lung ILC2s 14 . IL5 and IL13 production by stomach ILC2s depends on the microbiota 101 . In the context of H. pylori infection, ILC2s accumulate and produce IL5, which drives IgA production. Depletion of ILC2s in this model reduced IgA production and induced mucosal inflam mation and bleeding, suggesting that ILC2 driven IgA limits tissue damage by coating H. pylori. In contrast to mice, there is only a small population of CRTH2 + ILC2s in the adult human intestine and they are absent in the Nature reviews | Immunology 0123456789();: stomach of most healthy human donors 102 . Given that H. pylori causes expansion of stomach ILC2 popula tions in mice, further studies exploring the potential existence of CRTH2 -ILC2s in human stomach and their role in controlling H. pylori infection and subse quent H. pylori associated inflammation and cancer are needed. In contrast to our limited understanding of ILC2s a dec ade ago, today we know that ILC2s are highly dynamic cells that respond, migrate, establish tissue residency and adapt their effector functions depending on the microenvironment. This flexible behaviour resem bles that of T cells and ensures the right response at the right time and place. However, it also means that ILC2s are easily dysregulated to contribute to diseases such as cancer, metabolic disorders, inflammation and infectious diseases. In particular, the recent realization that IL4 and IL13 contribute to epigenetic imprinting of basal stem cells in the nasal epithelium of patients with CRSwNP, with potentially long lasting effects on type 2 skewing, highlights the therapeutic importance of targeting cells producing such cytokines, including ILC2s 103 . Indeed, ILC2s are a well recognized compo nent of a network of immune cells that contribute to allergy and asthma and, as such, are targets of multi ple biologics currently approved or in advanced clin ical trials for adults and, in some cases, adolescents, including those targeting IgE, IL4, IL13, IL5, IL33 and thymic stromal lymphopoietin 104 . Of note, recent data show effects of the anti IL4/anti IL13 drug dupi lumab in ameliorating uncontrolled moderate to severe asthma in 6-11 year old children 105 . By contrast, the pro inflammatory functions of ILC2s are beneficial in certain cancers, where ILC2s can be targets for anti PD1 therapy to improve tumour rejection, at least in mouse models 61 Fig. 4 | IlC2s are involved in virus-induced exacerbations of airway inflammation. a | In a mouse model, respiratory syncytial virus (RSV) infection induces activation of type 2 innate lymphoid cells (ILC2s) in the airways via uric acid-mediated production of IL-33, thymic stromal lymphopoietin (TSLP) and CCL2 by airway epithelial cells, as well as IL-1β production by myeloid cells. IL-13 production by ILC2s mediates mucin production as well as other downstream effects associated with asthma such as the IL-5-mediated maintenance of eosinophils. High levels of IL-33, IL-4, IL-13 and ILC2s in bronchoalveolar lavage (BAL) fluid are associated with more severe airway inflammation in infants with RSV infection. b | In rhinovirus infection in mice, alarmins such as IL-33, produced by airway epithelial cells and myeloid cells, drive ILC2 activation, leading to IL-5 and IL-13 production for the recruitment of eosinophils and mucin production, respectively. In experimental rhinovirus infection in humans, the bronchial ILC2 to ILC1 ratio correlates with asthma symptom score. c | In severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in humans, numbers of CCR6 + ILC2s in the circulation are reduced, which correlates with serum levels of CCL20, indicative of the CCL20-CCR6-dependent recruitment of ILC2s from the circulation to sites of inflammation such as the lungs. Blood ILC2 numbers are inversely correlated with serum markers of organ damage in patients with coronavirus disease 2019 (COVID-19), suggesting a potential detrimental role of ILC2s in inflamed tissues. However, the precise role for ILC2s in the airways, including their production of cytokines, such as amphiregulin, which might be involved in lung tissue repair, in patients with COVID-19 remains unclear. www.nature.com/nri 0123456789();: of ILC2s have been harnessed for third party cellu lar therapy that was well tolerated in a mouse model of graft versus host disease 106 , and co transplanted IL10 producing ILC2s prevented graft rejection in a mouse model of pancreatic islet transplantation 107 . Collectively, these studies present diverse strategies to interfere with ILC2 function depending on the disease type by reducing or enhancing the pro inflammatory functions of ILC2s or enhancing the tissue repair and/or immunosuppressive functions of ILC2s. For such approaches, we need a better understanding of ILC2 diversity across tissues and the factors, including lipids, neurotransmitters, hormones, cytokines and sur face ligands, that finetune the kinetics, magnitude and location of ILC2 responses. In particular, we need to better delineate the origin of and relationships between human ILC2 'subsets' to determine if they rather repre sent states of activation than true subsets with distinct precursors. Furthermore, we need to understand the roles of different ILC2 phenotypes in homeostasis and pathological conditions, such as cancer, obesity, inflam mation and infections, to pave the way for the develop ment of even more effective and specific therapies for such diseases. although the experimental evidence for a role of type 2 innate lymphoid cells (iLC2s) in coronavirus disease 2019 (COviD-19) is still lacking, two observational studies report depletion of peripheral blood iLCs, including iLC2s, in patients with COviD-19 (refs 110,111 ). Notably, peripheral blood iLC2 frequencies were reduced in severe versus moderate COviD-19, and iLC2 frequencies were negatively correlated with clinical markers of disease severity, including markers of tissue damage 110 . these data support a model for COviD-19 pathology in which iLC2s are recruited from the circulation to contribute to lung pathophysiology ( fig. 4c ). in support of that, CCr6 + iLC2s were particularly depleted in the peripheral blood of patients with COviD-19, and their frequency was inversely correlated to the serum concentration of the CCr6 ligand CCL20. indeed, a recent study identified CCr6-CCL20-mediated recruitment of peripheral blood iLC2s to the lung as a driver of excessive collagen production in the airways during cystic fibrosis 112 . the crucial question now is whether iLC2s contribute to tissue repair during COviD-19, as originally described for iLC2s in influenza virus infection 5 , and/or drive harmful pathogenic processes such as lung fibrosis, which is a characteristic feature of COviD-19. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity Innate production of T H 2 cytokines by adipose tissue-associated c-Kit + Sca-1 + lymphoid cells Systemically dispersed innate IL-13-expressing cells in type 2 immunity Human IL-25-and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161 Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion Tissue-specific transcriptional imprinting and heterogeneity in human innate lymphoid cells revealed by full-length single-cell RNA-sequencing Plasticity of innate lymphoid cell subsets Alternative activation generates IL-10 producing type 2 innate lymphoid cells IL-1beta, IL-23, and TGF-beta drive plasticity of human ILC2s towards IL-17-producing ILCs in nasal inflammation Induction of human regulatory innate lymphoid cells from group 2 innate lymphoid cells by retinoic acid This study describes the differentiation of human IL-10-producing 'regulatory' ILC2s and their role in allergen immunotherapy S1P-dependent interorgan trafficking of group 2 innate lymphoid cells supports host defense This study describes activated and steroidresistant CD45R0 + ILC2s in patients with asthma and chronic rhinosinusitis with nasal polyps The prostaglandin D2 receptor CRTH2 promotes IL-33-induced ILC2 accumulation in the lung The prostaglandin D(2) receptor CRTH2 regulates accumulation of group 2 innate lymphoid cells in the inflamed lung KLRG1 and NKp46 discriminate subpopulations of human CD117 + CRTH2 -ILCs biased toward ILC2 or ILC3 Optimal identification of human conventional and nonconventional (CRTH2 -IL7Ralpha -) ILC2s using additional surface markers Human CD5 + innate lymphoid cells are functionally immature and their development from CD34 + progenitor cells is regulated by Id2 An in vitro platform supports generation of human innate lymphoid cells from CD34 + hematopoietic progenitors that recapitulate ex vivo identity CD5 surface expression marks intravascular human innate lymphoid cells that have a distinct ontogeny and migrate to the lung Cytokine-induced endogenous production of prostaglandin D2 is essential for human group 2 innate lymphoid cell activation Expression of c-Kit discriminates between two functionally distinct subsets of human type 2 innate lymphoid cells c-Kit-positive ILC2s exhibit an ILC3-like signature that may contribute to IL-17-mediated pathologies IL-1beta, IL-4 and IL-12 control the fate of group 2 innate lymphoid cells in human airway inflammation in the lungs Interleukin-12 and -23 control plasticity of CD127 + group 1 and group 3 innate lymphoid cells in the intestinal lamina propria Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues IL-12 drives functional plasticity of human group 2 innate lymphoid cells IL-1 is a critical regulator of group 2 innate lymphoid cell function and plasticity Inflammatory triggers associated with exacerbations of COPD orchestrate plasticity of group 2 innate lymphoid cells in the lungs Innate lymphoid cells: models of plasticity for immune homeostasis and rapid responsiveness in protection Searching for the elusive regulatory innate lymphoid cell ILC2s are the predominant source of intestinal ILC-derived IL-10 Increased circulating CRTH2 + Tregs are associated with asthma control and exacerbation IL-10-producing innate lymphoid cells increased in patients with house dust mite allergic rhinitis following immunotherapy IL-10 production by ILC2s requires Blimp-1 and cMaf, modulates cellular metabolism, and ameliorates airway hyperreactivity Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs This study tracks the origin of circulating ILC2s in mice to conclude that tissue-specific activation of Distinct roles of LFA-1 and ICAM-1 on ILC2s control lung infiltration, effector functions, and development of airway hyperreactivity Neuropilin-1 mediates lung tissuespecific control of ILC2s function in type 2 immunity In situ maturation and tissue adaptation of type 2 innate lymphoid cell progenitors ILC2s mediate systemic innate protection by priming mucus production at distal mucosal sites Developmental plasticity allows outside-in immune responses by resident memory T cells Distinct alterations in the composition of mucosal innate lymphoid cells in newly diagnosed and established Crohn's disease and ulcerative colitis Gut microbiota regulate gut-lung axis inflammatory responses by mediating ILC2 compartmental migration Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4 + T cells Kinetics of thymocyte developmental process in fetal and neonatal mice A metabolite-triggered tuft cell-ILC2 circuit drives small intestinal remodeling Memory CD4 + T cells are generated in the human fetal intestine Human fetal TNF-alpha-cytokineproducing CD4 + effector memory T cells promote intestinal development and mediate inflammation early in life Innate and adaptive lymphocytes sequentially shape the gut microbiota and lipid metabolism Allergen-induced increases in sputum levels of group 2 innate lymphoid cells in subjects with asthma This study describes activated ILC2s in blood and BAL fluid following lung segmental allergen provocation in patients with allergic asthma BATF acts as an essential regulator of IL-25-responsive migratory ILC2 cell fate and function Pulmonary environmental cues drive group 2 innate lymphoid cell dynamics in mice and humans ILC2s: new actors in tumor immunity Group-2 innate lymphoid cells promote HCC progression through CXCL2-neutrophil-induced immunosuppression The tumour microenvironment shapes innate lymphoid cells in patients with hepatocellular carcinoma Blockade of the co-inhibitory molecule PD-1 unleashes ILC2-dependent antitumor immunity in melanoma The first description of the role of ILC2s in mediating the effects of anti-PD1 cancer immunotherapy in mice Single-cell transcriptomic landscape reveals tumor specific innate lymphoid cells associated with colorectal cancer progression Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer PPAR drives IL-33-dependent ILC2 pro-tumoral functions Type 2 innate lymphoid cells protect against colorectal cancer progression and predict improved patient survival Tumour-derived PGD2 and NKp30-B7H6 engagement drives an immunosuppressive ILC2-MDSC axis Mesenchymal PGD2 activates an ILC2-Treg axis to promote proliferation of normal and malignant HSPCs ILC2-driven innate immune checkpoint mechanism antagonizes NK cell antimetastatic function in the lung PD-1 expression affects cytokine production by ILC2 and is influenced by peroxisome proliferator-activated receptor-gamma Transdifferentiation of tumor infiltrating innate lymphoid cells during progression of colorectal cancer Tissue signals imprint ILC2 identity with anticipatory function Tissue-resident group 2 innate lymphoid cells differentiate by layered ontogeny and in situ perinatal priming Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity Activated type 2 innate lymphoid cells regulate beige fat biogenesis A stromal cell niche sustains ILC2-mediated type-2 conditioning in adipose tissue Peripheral PDGFRalpha + gp38 + mesenchymal cells support the differentiation of fetal liver-derived ILC2 Cold-responsive adipocyte progenitors couple adrenergic signaling to immune cell activation to promote beige adipocyte accrual Neuro-mesenchymal units control ILC2 and obesity via a brain-adipose circuit Describes ILC2s as effectors in the neuronal regulation of adipose tissue metabolism Costimulation of type-2 innate lymphoid cells by GITR promotes effector function and ameliorates type 2 diabetes Group 2 innate lymphoid cells: team players in regulating asthma Bilirubin represents a negative regulator of ILC2 in allergic airway inflammation Lipid-droplet formation drives pathogenic group 2 innate lymphoid cells in airway inflammation Regulation of type 2 innate lymphoid cell-dependent airway hyperreactivity by butyrate The first report of the metabolic requirements for maintenance, proliferation and effector cytokine production of human ILC2s Calcitonin gene-related peptide negatively regulates alarmin-driven type 2 innate lymphoid cell responses The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation Neuropeptide CGRP limits group 2 innate lymphoid cell responses and constrains type 2 inflammation Pulmonary neuroendocrine cells amplify allergic asthma responses Homeostatic IL-13 in healthy skin directs dendritic cell differentiation to promote TH2 and inhibit TH17 cell polarization TSLP elicits IL-33-independent innate lymphoid cell responses to promote skin inflammation Skin-resident innate lymphoid cells converge on a pathogenic effector state Single-cell analysis reveals innate lymphoid cell lineage infidelity in atopic dermatitis Composition of innate lymphoid cell subsets in the human skin: enrichment of NCR + ILC3 in lesional skin and blood of psoriasis patients Innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression Neutralization of IL-33 modifies the type 2 and type 3 inflammatory signature of viral induced asthma exacerbation Uric acid pathway activation during respiratory virus infection promotes Th2 immune response via innate cytokine production and ILC2 accumulation Elevated levels of type 2 respiratory innate lymphoid cells in human infants with severe respiratory syncytial virus bronchiolitis Role of ILC2 in viral-induced lung pathogenesis Demonstrates that the accumulation of bronchial ILC2s during in vivo rhinovirus infection correlates with respiratory symptoms in humans Pulmonary innate lymphoid cell responses during rhinovirus-induced asthma exacerbations in vivo: a clinical trial Bacteria-induced group 2 innate lymphoid cells in the stomach provide immune protection through induction of IgA Compartment-specific distribution of human intestinal innate lymphoid cells is altered in HIV patients under effective therapy Allergic inflammatory memory in human respiratory epithelial progenitor cells Strategies for choosing a biologic for your patient with allergy or asthma Dupilumab in children with uncontrolled moderate-to-severe asthma Third party type 2 innate lymphoid cells prevent and treat GI tract GvHD IL-10 producing type 2 innate lymphoid cells prolong islet allograft survival Harnessing the plasticity of CD4 + T cells to treat immune-mediated disease Macrophage plasticity and polarization: in vivo veritas Innate lymphoid cell composition associates with COVID-19 disease severity Severe COVID-19 patients exhibit an ILC2 NKG2D + population in their impaired ILC compartment ILC2 lung-homing in cystic fibrosis patients: functional involvement of CCR6 and impact on respiratory failure The authors thank J. Bernink, K. Golabski and R. Stadhouders for helpful input on the manuscript. The authors contributed equally to all aspects of the article. H.S. is a consultant for GlaxoSmithKline. J.M. declares no competing interests. Nature Reviews Immunology thanks G. Eberl, N. Satoh-Takayama and R. Locksley for their contribution to the peer review of this work. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.