Abstract
Background:
Type-2 innate lymphoid cells (ILC2s) are relevant players in type-2 asthma. They initiate eosinophil infiltration and airway hyperreactivity (AHR) through cytokine secretion. Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) is an inhibitory receptor considered as an immune checkpoint in different inflammatory diseases.
Objective:
Here we investigated the expression of LAIR-1 and assessed its role in human and murine ILC2s.
Methods:
WT and LAIR-1 KO mice were intranasally challenged with IL-33 and pulmonary ILC2s were sorted to perform an ex-vivo comparative study, based on RNA sequencing and flow cytometry. We next studied the impact of LAIR-1 deficiency on AHR and lung inflammation using knock-out mice and adoptive transfer experiments in Rag2−/− Il2rg−/− mice. Knockdown antisense strategies and humanized mice were used to assess the role of LAIR-1 in human ILC2s.
Results:
We have demonstrated that LAIR-1 is inducible on activated ILC2s and downregulates cytokine secretion and effector function. LAIR-1 signaling in ILC2s was mediated via inhibitory pathways including SHP1/PI3K/AKT and LAIR-1 deficiency led to exacerbated ILC2-dependent AHR in IL-33 and Alternaria alternata models. In adoptive transfer experiments, we confirmed the LAIR-1-mediated regulation of ILC2s in vivo. Interestingly, LAIR-1 was expressed and inducible in human ILC2s and knockdown approaches of Lair1 resulted in higher cytokine production. Finally, engagement of LAIR-1 by physiological ligand C1q, significantly reduced ILC2-dependent AHR in a humanized ILC2 murine model.
Conclusion:
Our results unravel a novel regulatory axis in ILC2s with the capacity to reduce allergic AHR and lung inflammation.
Keywords: LAIR-1, ILC2, airway hyperreactivity, immune checkpoints, allergic asthma
Graphical Abstract

Capsule summary:
This study demonstrates for the first time the role of LAIR-1 as an inhibitory immune receptor on pulmonary ILC2s and highlights LAIR-1 capacity to downregulate allergic asthma.
Introduction
Type-2 innate lymphoid cells (ILC2s) are newly discovered tissue-resident immune cells belonging to the family of non-cytotoxic ILCs. Unlike B and T cells, ILC2s lack the expression of lineage markers and specific antigen receptors1,2. Mainly residing in adventitial cuffs within collagen-rich regions, ILC2s are in tight interaction with resident structural cells that release non-specific alarmins including IL-33, IL-25 and thymic stromal lymphopoietin (TSLP), to trigger immune activation under stress conditions3–6. In particular, necrosis and active necroptosis are the principal mechanisms for IL-33 secretion that is associated with increased pulmonary epithelial permeability7,8. Pulmonary ILC2s are associated with adventitial stromal cells, known as a fibroblast-like subset implicated in alarmin secretion and matrix remodeling4. Due to their location, ILC2s are among the first immune responders to these endogenous danger signals, promoting their activation and proliferation. Activated ILC2s mimic T-helper 2 (Th2) cells in secreting copious amounts of classical Th2 effector cytokines including IL-5, IL-9 and IL-132,9. During the last decade, numerous studies have evidenced the role of ILC2s in different diseases such as type-2 diabetes mellitus10,11, chronic rhinosinusitis12, liver fibrosis13, but mainly in allergic asthma type-2 inflammation14–16.
Allergic asthma is the most frequent type of asthma characterized by airway hyperreactivity (AHR) and Th2-mediated immune responses. Exposure to exogenous stimuli, including inhaled allergens, triggers specific symptoms ranging from chest pain, cough and wheezing to acute bronchoconstriction in life-threatening attacks17,18. The inflammatory cascade is initiated with alarmin secretion in the lungs, which instructs a type-2 response associated with massive recruitment of eosinophils in response to IL-519,20. Representing key upstream players, ILC2s have become potential therapeutic target in asthma21. Novel strategies are considering the modulation of inhibitory and co-stimulatory receptors defining activated ILC2s, such as CD200R22, TIGIT23, TNFR224, ICOS and ICOS-L25. In this context, we have recently demonstrated the therapeutic potential of a novel programmed cell death protein 1 (PD-1) agonist in allergic asthma through the downregulation of ILC2s26. Other inhibitory receptors are yet to be explored in ILC2s, such as the Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1).
LAIR-1, also known as CD305, was first discovered in 1997 and described as a novel inhibitory receptor having a single immunoglobulin-like domain and a cytoplasmic tail, equipped with two immuno-receptor tyrosine-based inhibitory motifs (ITIMs)27. LAIR-1 is broadly expressed and inhibits the activation of different immune populations including B and T lymphocytes, Natural killers (NKs), dendritic cells (DCs) and monocytes27–31. It was reported that LAIR-1 binds to collagens with high affinity through the conserved sequence of Gly-Pro-Hyp collagen repeats32 and a single residue arginine 6533. Moreover, collagen was reported to induce and enhance the expression of LAIR-134. The first component of complement C1q also contains a collagen-like region (CLR) and is therefore considered as a functional ligand for LAIR-1 among other immune receptors35–37. As an ITIM-containing receptor, LAIR-1 engagement mainly leads to Src homology phosphatase-1 (SHP1) recruitment, promoting the transduction of negative signaling27,30,38. To date, studies have demonstrated a critical role for LAIR-1 in the immune imbalance defining autoimmune diseases and cancers. LAIR-1 crosslinking limits rheumatoid arthritis (RA)34 and systematic lupus erythematosus (SLE) progression31, while it decreases immune cell cytotoxicity and improves oncogenic resistance in cancer39,40.
In the present study, we highlighted for the first time two major findings. First, we demonstrated that LAIR-1 is inducible on pulmonary ILC2s and controls their activation mainly through signal transduction downstream of SHP-1. Second, we used the knockout mouse to assess the protective anti-inflammatory role of LAIR-1 in ILC2-dependent AHR. In a translational approach, we also demonstrated a major role for LAIR-1 in the regulation of human ILC2s both in vitro and in vivo utilizing a humanized mouse model. Therefore, our data clearly introduced LAIR-1 as an inhibitory receptor in ILC2s that could serve as a potential therapeutic target to treat ILC2-related disorders including asthma and allergic diseases.
Methods
Mouse experiments
All mice were bred in the animal facility of the Keck School of Medicine, University of Southern California (USC). Experimentation protocols were approved by the USC Institutional Animal Care and Use Committee and conducted in accordance with the principles of the Declaration of Helsinki. Wild-type (WT) C57BL/6, Lair1 knock-out (LAIR-1 KO) and Rag2/Il2rg double knockout (Rag2−/− Il2rg−/−) mice were purchased from Jackson Laboratory (Bar Harbor, ME). Six to eight-week-old aged- and sexed-matched mice were used in the study.
Tissue digestion and pulmonary ILC2 sorting
Mice were intranasally (i.n.) challenged on 3 consecutive days with 0.5 or 1 μg per mouse in 50 μL of carrier-free recombinant mouse (rm)-IL-33 (ThermoFisher) or PBS. On day 4, lungs were collected and processed as described previously26,41. Briefly, lung tissue was cut into small pieces and incubated in type IV collagenase (400 U/ml; Worthington Biochemicals) at 37 °C for 60 min. To obtain single-cell suspension, lung tissue digest was passed through a 70-μm cell strainer (Falcon). In some experiments, blood, bone marrow, ear skin and visceral adipose tissue were collected and prepared for flow cytometry, as previously described11,42. The following antibodies were used to identify and sort pulmonary ILC2s: APC-Cy7 anti-mouse CD45 (clone 30-F11), PE-Cy7 anti-mouse CD127 (clone A7R34) (both from BioLegend), PerCP-eFluor710 anti-mouse ST2 (clone RMST2-2; eBioscience), PE anti-mouse LAIR-1 (clone 113; ThermoFisher Scientific), and FITC anti-mouse lineage cocktail including TCR-γδ (clone eBioGL3; eBioscience), TCR-β (clone H57-597), CD3e (clone 145-2C11), CD45R (clone RA3-6B2), Gr-1 (clone RB6-8C5), CD11c (clone N418), CD11b (clone M1/70), Ter119 (clone TER-119), FcɛRI (clone MAR-1), CD5 (clone 53–7.3) and NKp46 (clone 29A1.4) (all from BioLegend). APC anti-mouse KLRG1 (clone 2F1/KLRG1; BioLegend), PE anti-mouse MAIR-V (clone TX70; BioLegend), APC anti-mouse CD200R (clone OX110; Invitrogen) and APC anti-mouse PD-L1 (clone 10F.9G2; BioLegend) were added for immunophenotyping experiments. When indicated, BV421 anti-mouse CD135 (Flt3; clone A2F10), APC anti-mouse LPAM-1 (Integrin α4β7; clone DATK32) and BV510 anti-mouse CD25 (clone PC61) (all from BioLegend) were used to identify ILC2 precursors in the bone marrow. Fluorescent live/dead fixable stains (ThermoFisher) were used in all experiments to exclude dead cells, according to manufacturer’s instructions. Pulmonary ILC2s were sorted on a FACSARIA III system (Becton Dickinson). The sort purity was always ≥95% and the percentage of LAIR-1+ ILC2s from WT mice was ~25–35%. Cells were cultured in RPMI (Gibco) supplemented with 10% heat-inactivated fetal bovine serum and 100 units per mL penicillin–streptomycin (GenClone).
RNA-sequencing (RNAseq)
Sorted ILC2s were incubated (5 × 104/mL) with rm-IL-2 (10 ng/mL) and rm-IL-7 (10 ng/mL) for 24 h. Total RNA was isolated using MicroRNAeasy (Qiagen). In total, 10 ng of input RNA was used to produce cDNA for downstream library preparation. Samples were sequenced on a NextSeq 500 (Illumina) system. Raw reads were aligned, normalized and further analyzed using Partek Genomics Suite software, version 7.0 Copyright; Partek Inc. Normalized read counts were tested for differential expression using Partek’s gene-specific analysis (GSA) algorithm. RNA sequence data that support the findings of this study have been deposited in Genbank with the primary accession code GSE171819.
Intracellular staining and cytokine quantification
ILC2s were cultured in the presence of rm-IL-2 (10 ng/mL) and rm-IL-7 (10 ng/mL). When indicated, ILC2s were incubated with purified mouse C1q (10 μg/mL, Complement Technology) or with 15 μM of the SHP-1 inhibitor, known as Tyrosine Phosphatase Inhibitor 1 (TPI-1, MedChemExpress)39,43. The BD Biosciences Cytofix/Cytoperm kit was used and followed by intracellular staining with PE anti-p65 (clone 532301, R&D Systems), BV421 anti-AKT (pS473) (clone M89-61, BD Biosciences), PE-Cy7 anti-BCL-2 (clone 10C4, eBioscience) and unconjugated anti-SHP-1 (clone SR41-02, ThermoFisher) followed by Alexa Fluor 647 goat anti-rabbit IgG secondary antibody (Jackson ImmunoResearh). Intranuclear staining was performed using the Foxp3 Transcription Factor Staining Kit (ThermoFisher) according to the manufacturer’s instructions and APC anti-mouse Ki67 (SolA15, ThermoFisher) was used to assess proliferation. The CellTrace Violet Cell Proliferation Kit (ThermoFisher) was also used in some experiments, according to manufacturer’s instructions. In parallel, the levels of IL-5, IL-6, and IL-13 were measured in supernatants using Legendplex multiplex kits (BioLegend) and data were analyzed via the LEGENDplex data analysis software v8.0. GM-CSF was quantified using ELISA kit from BioLegend.
Induction and assessment of AHR
Mice were intranasally (i.n.) challenged for 3 days with mouse rm-IL-33 (0.5 μg per mouse in 50 μL, BioLegend) to induce AHR or for 5 consecutive days with 100 μg of Alternaria alternata extracts (Greer Laboratories). Control mice were challenged with PBS and lung function was evaluated on the day following the last challenge. In some experiments, AHR was induced in Rag2−/− Il2rg−/− mice after an intravenous adoptive transfer of total pulmonary ILC2s (50 × 103) sorted from IL-33-challenged WT and LAIR-1 KO mice. Lung function was assessed by direct measurement of lung resistance and dynamic compliance (cDyn) in restrained, tracheostomized, and mechanically ventilated mice using the FinePointe RC system (Buxco Research Systems) under general anesthesia. Mice were sequentially challenged with aerosolized PBS (baseline), followed by increasing doses of methacholine ranging from 5 to 40 mg/mL. Maximum lung resistance and minimum compliance values were recorded during a 3-min period after each methacholine challenge. AHR data were analyzed by repeated measurements of a general linear model.
Assessment of lung inflammation
To collect the bronchoalveolar lavage (BAL) fluid, lungs were washed with 3 ml ice-cold PBS. BAL supernatant was collected to quantify cytokines using Legendplex multiplex kits (BioLegend). Principal immune populations were identified in the BAL using the following antibodies: PE-Cy7 anti-mouse CD45 (clone 30-F11; BioLegend), APC-Cy7 anti-mouse CD11c (clone N418; BioLegend), PE anti-mouse Siglec-F (clone E50-2440; BD Biosciences), BV421 anti-mouse CD11b (clone M1/70; BD Biosciences), APC anti-mouse Gr-1 (clone RB6-8C5; BioLegend), FITC anti-mouse CD19 (clone 6D5; BioLegend), PerCP-Cy5.5 anti-mouse CD3 (clone 17A2; BioLegend). Lung tissue was processed and ILC2s were identified as described above, while the expression of C1q in CD11c+ cells was assessed using APC-Cy7 anti-mouse CD11c (clone N418; BioLegend) and FITC anti-C1q (clone JL-1; Invitrogen). When indicated, intracellular staining was performed to evaluate cytokine production using APC anti-mouse/human IL-5 (clone TRFK5; BioLegend) and PE anti-mouse IL-13 (clone eBio13A; eBioscience) after 4 h of in vitro incubation with 50 μg/mL PMA (Sigma-Aldrich), 500 μg/mL ionomycin (Sigma-Aldrich), and 1 μg/mL Golgi plug. Fluorescent live/dead fixable stains (ThermoFisher Scientific) were used to exclude dead cells, according to manufacturer’s instructions. CountBright Absolute Count Beads were used to count lung and BAL immune cells (Invitrogen). Acquisition was performed on a BD FACSCanto II (BD Biosciences) using the BD FACSDiva software v8.0.1. Data were analyzed with FlowJo software (TreeStar) version 10. In some experiments, one lobe per lung was collected for histology and stored in paraformaldehyde 4% buffered in PBS. Lungs were embedded in paraffin and cut into 4-μm sections for hematoxylin and eosin (H&E) staining or for trichrome staining. Histology pictures were acquired on a Leica DME microscope and Leica ICC50HD camera (Leica) and ImageJ was used for analysis.
Culture of human ILC2s and adoptive transfer
All human studies were approved by USC Institutional review board and conducted in accordance with the principles of the Declaration of Helsinki. Informed consent was obtained from all human participants. Human peripheral blood ILC2s were isolated from total PBMCs as previously described26. Briefly, human fresh blood was first diluted 1:1 in PBS and transferred to SepMateTM-50 separation tubes (Stemcell Technologies) prefilled with 15 mL LymphoprepTM (Axis-Shield). Samples were centrifuged at 1200 × g for 10 min to collect PBMCs. The CRTH2 MicroBead kit (Miltenyi Biotec) was then used according to the manufacturer’s conditions in order to isolate CRTH2+ cells. The following antibodies were used to identify and sort human ILC2s: FITC anti-lineage, FITC anti-CD235a (clone HI264), FITC anti-FceRIa (clone AER-37), FITC anti-CD1a (clone HI149), FITC anti-CD123 (clone 6H6), FITC anti-CD5 (clone L17F12), APC-Cy7 anti-CD45 (clone HI30), PE anti-CD294 (CRTH2) (clone BM16), PE-Cy7 anti-CD127 (clone A019D5) (all from BioLegend). Purified ILC2s were cultured in complete RPMI media with recombinant human (rh)-IL-2 (20 ng/mL), rh-IL-7 (20 ng/mL) and rh-IL-33 (20 ng/mL) for 3 days. For knock-down experiments, 5 μM of Lair-1 antisense morpholino oligonucleotides AGACATGGCCCAGGTCCCAGCAGT or random control oligonucleotides (Gene Tools, LLC, Philomath, Ore) were added to the culture as free uptake oligonucleotides at T=0h and T=36h. When indicated LAIR-1 expression was assessed on freshly isolated or cultured ILC2s using Alexa Fluor 647 anti-human LAIR-1 (clone NKTA255, BioLegend). Intranuclear staining with eFluor450 anti-human Ki67 (clone 20Raj1, eBioscience) was performed to assess proliferation. Human IL-5, IL-6 and IL-13 were quantified in supernatants using Legendplex multiplex kits from BioLegend.
For adoptive transfer experiments, in vitro-cultured ILC2s (50 × 103) were intravenously (i.v.) injected in Rag2−/− Il2rg−/− mice. To induce AHR, mice were intranasally challenged with 1 μg of rh-IL-33 for three consecutive days. On days 4 and 5, mice received 50 μg of purified human C1q (Complement Technology) or PBS via the intraperitoneal route (i.p.) as described before44. The i.p route was chosen since C1q is a high molecular weight protein (410–462 kDa), and therefore has a limited diffusion via the intranasal route45,46. On day 6, lung function was measured before BAL and lung collection for flow cytometry analysis.
Statistical analysis
Data are representative of at least two independent experiments and are presented as means ± SEM (except for RNAseq). Two-tailed Student’s t test was applied for comparisons between 2 groups. For multigroup comparisons, we used one-way ANOVA with the Tukey post hoc test. Data were analyzed with Prism Software (GraphPad Software Inc.). Error bars represent standard error of the mean. p value < 0.05 was considered to denote statistical significance (*p <0.05, **p < 0.01, ***p < 0.001).
Results
LAIR-1 is inducible on pulmonary ILC2s
LAIR-1 is broadly and constitutively expressed on different immune populations including monocytes, NK and B/T cells47. To elucidate the expression of LAIR-1 on ILC2s, C57BL/6 WT mice were challenged for three consecutive days with rm-IL-33 or with PBS and lungs were collected at day 4 (Fig. 1A). Pulmonary ILC2 expression was assessed utilizing flow cytometry by gating on CD45+ lineage− ST2+ CD127+ as described before26(Fig. 1B). LAIR-1 was not expressed on naïve pulmonary ILC2s, while intranasal stimulation with IL-33 induced a LAIR-1+ ILC2 population (Fig. 1C, D; Fig E1, A48). Similarly, LAIR-1 was not expressed on skin and adipose tissue resident ILC2s at the basal state however it was weakly expressed on bone marrow and blood ILC2s (Fig E1, B–H). To explore the particularities of this induced subset, we sorted both LAIR-1− and LAIR-1+ ILC2s and established a differential expression profiling using RNAseq analysis. LAIR-1+ ILC2s presented a different transcriptional profile, as revealed by principal component analysis (PCA) (Fig. 1E). This analysis provides unsupervised information on the clustering of LAIR-1− and LAIR-1+ ILC2 samples based on the highest variability in the overall dataset. LAIR-1 expression was associated with the upregulation of 950 genes and the downregulation of 875 genes (Fig. 1F). In particular, genes related to inflammation and ILC2 activation such as Il13, Il9 and Nfkbia were downregulated in LAIR-1+ ILC2s. Along with Lair1 gene induction, a battery of genes related to tissue remodeling, such as Loxl2, Col18a1, Cd9 and Ecm1, were among the most upregulated (Fig. 1G; Fig E1, I). In parallel, the trichrome histological staining suggested lung tissue remodeling and collagen deposition following repeated IL-33 challenges (Fig E2, A). C1q was reported as a potential natural ligand that engages LAIR-1 through a non-complement mechanism36. Furthermore, both flow cytometry and transcriptional data suggested that C1q is mainly and constitutively expressed by antigen presenting cells (DCs and macrophages) in mouse and human lungs (Fig E2, B–E49). To confirm the regulatory role of LAIR-1 axis, we treated both ILC2 subsets with C1q or PBS and assessed cytokine production (Fig. 1H). In line with the transcriptional data, LAIR-1+ ILC2s secreted less IL-5 and IL-13 in vitro. In addition, C1q significantly decreased IL-5 and IL-13 production in LAIR-1+ ILC2s, but not in LAIR-1− ILC2s (Fig. 1I, J). Altogether, our data indicate that LAIR-1 is inducible on ILC2s, is functional, and participates in ILC2 downregulation.
Figure 1: IL-33 induces a LAIR-1+ population in pulmonary ILC2s.

(A-J) C57BL/6 WT mice were intranasally challenged for three consecutive days with 0.5 μg of rm-IL-33 or with PBS, and lungs were collected on day 4.
(B) Pulmonary ILC2s identified as CD45+ lineage− CD127+ ST2+ cells.
(C) Representative flow cytometry plots showing LAIR-1 expression on ILC2s from PBS- and IL-33-challenged mice.
(D) LAIR-1 expression kinetics on ILC2s in response to in vivo IL-33 challenges, represented as percentages of LAIR-1+ ILC2s.
(E-J) LAIR-1− and LAIR-1+ ILC2s were sorted from IL-33-challenged mice and incubated with rm-IL-2 (10 ng/mL) and rm-IL-7 (10 ng/mL) for 24 h.
(E) PCA plot showing the clustering of LAIR-1− and LAIR-1+ ILC2 samples based on uncorrelated variables.
(F) Heatmap representation of total differentially regulated genes, generated with Gene-specific analysis (GSA) algorithm (counts <20; p-value ≤ 0.05; n = 3).
(G) Focused volcanoplot representing a list of relevant and significantly regulated genes in LAIR-1+ ILC2s as compared to LAIR-1− ILC2s.
(H-J) LAIR-1− and LAIR-1+ ILC2s were cultured with or without C1q at 10 μg/ml and supernatants were collected after 24 hours.
(I, J) Levels of IL-5 (I) and IL-13 (J) quantified using LEGENDplex immunoassay.
Data are presented as means ± SEM (one-way ANOVA; *p <0.05, **p < 0.01, ***p < 0.001).
Lack of LAIR-1 on pulmonary ILC2 increases effector function and Th2 cytokine secretion
To better assess the implication of LAIR-1 on ILC2s, we introduced the previously characterized LAIR-1 KO mouse50, which has a targeted deletion of Lair1 gene. WT and LAIR-1 KO mice were intranasally challenged with rm-IL-33 for 3 consecutive days in order to compare the transcriptional profile of FACS-sorted ILC2s (Fig. 2A). The absence of LAIR-1 resulted in 1181 significantly modulated genes (counts <20; p-value ≤ 0.05), displayed as a heatmap for differential gene expression (Fig. 2B). Interestingly, 326 genes were consistently and commonly modulated in our 2 transcriptional analyses: i) LAIR-1− vs LAIR-1+ ILC2s from WT mice and ii) LAIR-1 KO vs WT ILC2s. Relevant inflammatory genes including Il13, Il9, Tnfrsf8, Stat5b were upregulated in LAIR-1− and LAIR-1 KO ILC2s, while mainly antioxidant and anti-inflammatory genes, such as Gclm, Prdx6, Nqo1, Dok2 and CD200r1 were consistently downregulated in comparison to LAIR-1+ and WT ILC2s (Fig. 2C). This further highlights a central anti-inflammatory role for LAIR-1 in activated ILC2s. To better explore the LAIR-1 mechanism of action, we performed pathway analysis using gene-level statistics to compute pathway-level scores. Interestingly, PI3K/AKT signaling, Th2 pathway and NF-κB activation were among the highly affected pathways (Fig. 2D). As ILC2s functionally mimic Th2 cells, we first compared the expression of cytokine related genes in activated WT and LAIR-1 KO ILC2s. A list of genes encoding Th2 cytokines, including Il5, Il13, Il9, and Csf2 were significantly upregulated in the absence of LAIR-1 (Fig. 2E). At the protein level, ILC2s from LAIR-1 KO mice exhibited higher production of IL-5, IL-13, IL-6 and GM-CSF (Fig. 2F–I), confirming the transcriptional data. Taken together, these results indicate that LAIR-1 axis can negatively regulate relevant inflammatory pathways and dampen cytokine production in activated ILC2s.
Figure 2: LAIR-1 deficiency increases ILC2 activation and cytokine production.

(A-I) WT and LAIR-1 KO mice were challenged intranasally for 3 consecutive days with 0.5 μg rm-IL-33. On day 4, pulmonary ILC2s were sorted and cultured with rm-IL-2 (10 ng/mL) and rm-IL-7 (10 ng/mL) for 24 h.
(B) Heatmap representation of total differentially regulated genes, generated with GSA algorithm (counts <20; p-value ≤ 0.05; n = 3).
(C) Venn diagram highlighting a list of commonly regulated genes in two bulk RNAseq: LAIR-1− vs LAIR-1+ from WT mice and LAIR-1 KO vs WT ILC2s. Green list represents downregulated genes and red list represents upregulated genes in LAIR-1− and LAIR KO ILC2s.
(D) Pathway enrichment analysis enriched for relevant up-regulated genes in LAIR-1 KO ILC2s.
(E) Differentially expressed cytokine and cytokine receptor genes plotted as the normalized counts in WT compared to LAIR-1 KO ILC2s. Red dots represent significantly upregulated relevant genes (p-value ≤ 0.05; n = 3).
(F-H) Levels of IL-5 (F), IL-13 (G), and IL-6 (H) quantified using LEGENDplex bead-based immunoassay.
(I) Level of GM-CSF quantified by ELISA.
Data are presented as means ± SEM (unpaired two-tailed Student’s t test; *p <0.05, **p < 0.01, ***p < 0.001).
Cell, mouse and human images are provided with permission from Servier Medical Art.
LAIR-1 mediates regulation of ILC2s utilizing SHP-1/PI3K/AKT signaling circuits
The PI3K/AKT pathway is a principal regulator of cell activation, differentiation, proliferation and survival51. According to our pathway analysis, PI3K/AKT was one of the mainly affected pathways in activated LAIR-1 deficient ILC2s. Consistently, several studies have highlighted a constitutive interaction between LAIR-1 and SHP-138,39, a phosphatase that directly inhibits the PI3K/AKT downstream signaling as well as NF-κB52 (Fig. 3A). To investigate these interactions in ILC2s, we assessed the effect of LAIR-1 on the expression of relevant downstream players at the transcriptional and the protein level. Interestingly, a list of genes related to PI3K pathway, NFκB activation, apoptosis and proliferation were significantly upregulated in LAIR-1 KO activated ILC2s compared to WT, while Ptpn6 that encodes SHP-1 was significantly downregulated (Fig. 3B). Using flow cytometry, we also observed a significantly lower expression of SHP-1 in LAIR-1 KO ILC2s as compared to WT ILC2s (Fig. 3C). The lack of LAIR-1 was also associated with higher expression of AKT, p65 which is a principal component of canonical NF-κB pathway, and the anti-apoptotic factor BCL-2 (Fig. 3D–F). Moreover, proliferation was improved as revealed by the ki67 intranuclear staining (Fig. 3G) and the CellTrace Violet staining (Fig E3, A). To further confirm the implication of SHP-1 in ILC2 activation, we used the specific inhibitor TPI-1 at 15 μM to modulate SHP-1 activity in vitro. Interestingly, SHP-1 inhibition in ILC2s led to an increased in vitro proliferation and enhanced IL-5 production (Fig E3, B, C). Moreover, the LAIR-1 ligand C1q increases the expression of SHP-1 in ILC2s (Fig E3, D). Altogether, these results support the involvement of LAIR-1/SHP-1 interactions in the control of ILC2 activation.
Figure 3: LAIR-1 signaling involves PI3K/AKT and NF-κB pathways.

(A-G) WT and LAIR-1 KO mice were challenged intranasally for 3 consecutive days with 0.5 μg rm-IL-33. On day 4, pulmonary ILC2s were sorted and cultured with rm-IL-2 (10 ng/mL) and rm-IL-7 (10 ng/mL) for 24 h.
(A) Predicted LAIR-1 downstream signaling cascade.
(B) Heat map representation of differentially regulated genes involved in LAIR-1 signaling pathways. GSA algorithm was used to test for differential expression of genes in WT vs LAIR-1 KO ILC2s (counts <20; p-value < 0.05; n = 3).
(C-F) Representative histogram (left) with the corresponding MFI quantification (right) of SHP-1 (C); p65 (D); AKT (E) and BCL-2 (F) expression in WT and LAIR-1 KO ILC2s.
(G) Representative flow cytometry plots of Ki67 intranuclear staining and corresponding quantification (right) presented as the percentage of proliferative ILC2s.
Data are presented as means ± SEM (unpaired two-tailed Student’s t test; *p <0.05, **p < 0.01).
LAIR-1 regulates lung function and inflammation in ILC2-dependent AHR
Given the regulatory role of LAIR-1 in pulmonary ILC2s, we next hypothesized that this receptor may reduce AHR and lung inflammation. WT and LAIR-1 KO mice received via the intranasal route either IL-33 or PBS for 3 consecutive days. One day after the last challenge, lung function was assessed by direct measurement of lung resistance and dynamic compliance (cDyn) in anesthetized tracheostomized mice using the FinePointe RC system (Buxco Research Systems), followed by BAL collection and lung tissue sample analysis. Although IL-33 significantly increased lung resistance in both genotypes; lung resistance in LAIR-1 KO mice was significantly higher than in WT mice (Fig. 4A). Consistently, results of dynamic compliance showed lower response in LAIR-1 KO compared to WT mice (Fig. 4B). This suggested that LAIR-1 reduces IL-33-induced AHR. The principal immune populations in the BAL were identified and quantified by flow cytometry (Fig E4, A–E). In agreement with AHR, the total number of immune cells (CD45+) in the BAL of IL-33-challenged mice was significantly higher in LAIR-1 KO compared to WT mice (Fig. 4C). Eosinophilia, known as a main feature in type-2 asthma, was significantly higher in LAIR-1 KO mice than in WT mice, as well as the number of inflammatory CD11b+ DCs (Fig. 4D, E; Fig E4, A). In the lungs, ILC2 count was higher in LAIR-1 KO mice following IL-33 challenge, as was the percentage of IL-5+ IL-13+ ILC2s (Fig. 4F, G; Fig E5, A). The exacerbated lung inflammation in LAIR-1 deficient mice was also confirmed by histology (Fig. 4H). Expectedly, the lack of LAIR-1 improved ILC2s viability confirming our previous in vitro data (Fig E5, B), however it had no significant effect on the expression of other receptors including MAIR-V, KLRG1, PD-L1 and CD200R (Fig E5, C–H). Altogether, these results reveal that LAIR-1 dampens IL-33-induced lung inflammation and reduces AHR.
Figure 4: LAIR-1 deficiency exacerbates ILC2-dependent AHR and lung inflammation.

(A-H) WT and LAIR-1 KO mice were challenged for 3 consecutive days with 0.5 μg rm-IL-33. On day 4, AHR and lung inflammation were assessed.
(A) Lung resistance and (B) dynamic compliance; n = 5.
(C) Total number of CD45+ cells in the BAL.
(D, E) Total number of eosinophils (D) and CD11b+ DCs (E) in the BAL.
(F) Total number of pulmonary ILC2s gated as CD45+ lineage− ST2+ CD127+ cells.
(G) Percentage of IL-5+ and IL-13+ ILC2s identified by intracellular staining.
(H) H&E staining of lung sections (scale bars 50 μm).
(I-M) WT and LAIR-1 KO mice were challenged for 3 consecutive days with 0.5 μg rm-IL-33. On day 4, pulmonary ILC2s were sorted and adoptively transferred into Rag2−/− Il2rg−/− mice via the intravenous route (i.v.). Recipient mice were then challenged intranasally for 3 consecutive days with 0.5 μg rm-IL-33 before the assessment of AHR and lung inflammation.
(J) Lung resistance; n = 4.
(K) Total number of eosinophils in the BAL.
(L) Gating of lineage− cells from live CD45+ cells.
(M) Total number of pulmonary ILC2s gated as CD45+ lineage− ST2+ CD127+ cells.
Data are presented as means ± SEM (one-way ANOVA; *p <0.05, **p < 0.01, ***p < 0.001).
To address LAIR-1 specific implication in ILC2-dependent asthma, we adoptively transferred activated WT or LAIR-1 KO ILC2s in Rag2−/−Il2rg−/− mice that lack all lymphoid cells. Recipient mice were then challenged with IL-33 for 3 consecutive days. On day 4, AHR, BAL eosinophils and pulmonary ILC2 numbers were assessed (Fig. 4I). As expected, in the absence of any adoptive transfer, Rag2−/−Il2rg−/− mice did not develop AHR in response to IL-33. Consistent with the previous observations in LAIR-1 KO mice, we observed here that mice receiving LAIR-1 KO ILC2s displayed significantly higher lung resistance and increased eosinophil recruitment as compared to those receiving WT ILC2s (Fig. 4J, K). In addition, the percentage of CD45+ lineage negative cells and consequently the number of LAIR-1 KO ILC2s in the lungs were significantly higher (Fig. 4L, M). Therefore, these models overall confirm our previous results and underline the protective role of LAIR-1 in IL-33-induced AHR and pulmonary inflammation through the regulation of ILC2s.
LAIR-1 downregulates Alternaria alternata-induced AHR and limits lung inflammation
We next investigated whether LAIR-1 conserves its immunoregulatory role in a clinically relevant AHR model induced by the fungal allergen Alternaria alternata, known to be associated with type-2 inflammation53,54. WT and LAIR-1 KO mice were intranasally challenged with Alternaria alternata extracts (100 μg per mouse) for 5 consecutive days, followed by AHR and lung inflammation assessment (Fig. 5A). Consistent with the IL-33 model, lung resistance was higher in LAIR-1 KO mice as compared to WT mice (Fig. 5B). This was associated with an increased number of CD45+ cells in the BAL (Fig. 5C). In particular, LAIR-1 KO mice displayed significantly higher numbers of eosinophils, T cells and B cells, but similar recruitment of neutrophils and CD11c+ cells (Fig. 5D–H). We further addressed whether LAIR-1 is inducible in ILC2s and regulates their activation in response to a widespread fungal allergen. Interestingly, our results revealed that mice exposure to Alternaria alternata induces LAIR-1 expression on about 30–40% of pulmonary ILC2s (Fig. 5I). In parallel, the lack of LAIR-1 was associated with an increased number of pulmonary ILC2s as well as an increased percentage of IL-5+IL-13+ ILC2s (Fig. 5J, K). Taken together, these results confirm the inhibitory role of LAIR-1 in a clinically relevant asthma model and demonstrate LAIR-1 capacity to control type-2 immune response.
Figure 5: LAIR-1 deficiency significantly increases Alternaria alternata-induced pulmonary inflammation.

(A-K) WT and LAIR-1 KO mice were challenged intranasally for 5 consecutive days with 100 μg of Alternaria alternata extracts. On day 6, AHR and lung inflammation were assessed.
(B) Lung resistance measured in response to increasing concentrations of methacholine, n=4.
(C) Total number of CD45+ cells in the BAL.
(D-H) Total number of eosinophils (D), neutrophils (E), CD11c+ cells (F), T cells (G) and B cells (H) in the BAL.
(I) Percentage of LAIR-1+ ILC2s in the lungs.
(J) Total number of pulmonary ILC2s.
(K) Percentage of IL-5+ and IL-13+ ILC2s.
Data are presented as means ± SEM (unpaired two-tailed Student’s t test; *p <0.05, **p < 0.01).
LAIR-1 is inducible and controls the effector function of human ILC2s
ILC2s are now considered as relevant immune players in human type-2 asthma55–57, despite their low percentage among PBMCs (Fig E6). To assess the translational potential of our data, we explored whether LAIR-1 is expressed and if it modulated human ILC2 activation, as demonstrated in mouse. To this end, we assessed the expression of LAIR-1 on freshly sorted and on cultured ILC2s. Human ILC2s were identified and sorted as CD45+ lineage− CD127+ CRTH2+ cells (Fig. 6A, B). Interestingly, we first observed that LAIR-1 was expressed on circulating ILC2s at the basal state (T=0h) and was significantly induced in response to rh-IL-33 (T=72h) (Fig. 6C). Since LAIR-1 was remarkably expressed on human ILC2s, we performed functional tests using specific LAIR-1 antisense morpholino oligonucleotides to address the effect of a transient knockdown of Lair1 gene on human ILC2 activation in response to IL-33. As a validation of our approach, we observed a significant decrease in LAIR-1 expression on morpholino-treated ILC2s (Fig. 6D). Then, we demonstrated that morpholino treatment along with IL-33 stimulation increases ILC2 proliferation, as revealed after 72 hours of culture using the ki67 intranuclear staining (Fig. 6E). In parallel, the quantification of cytokine levels in the supernatant showed that specific Lair1 antisense morpholino is able to induce a significant upregulation in IL-5, IL-6 and IL-13 production as compared to control sequences (Fig. 6F–H). We lastly tested the effect of LAIR-1 engagement on human ILC2s in vivo using a humanized mouse model. Briefly, Rag2−/−Il2rg−/− mice that lack lymphoid cells were reconstituted with human peripheral blood ILC2s and intranasally challenged for three consecutive days with PBS or rh-IL-33 to induce AHR. On days 4 and 5, mice received PBS or human C1q, a potential physiological ligand for LAIR-1, via the intraperitoneal route (i.p.). On day 6, we assessed AHR and lung inflammation (Fig. 6I). Strikingly, lung resistance in C1q-treated mice was significantly reduced as compared to control mice (Fig. 6J). In line with lung resistance, C1q-treated mice showed abrogated lung inflammation associated with decreased eosinophilia and fewer number of human ILC2s (Fig. 6K, L). Taken together, these results indicate that LAIR-1 is highly implicated in the control of human ILC2 activation, suggesting a promising therapeutic approach against type-2 asthma.
Figure 6: LAIR-1 is expressed and functional on human ILC2s.

(A-H) Human ILC2s were sorted from PBMCs and cultured in the presence of rh-IL-2, rh-IL-7 and rh-IL-33, with LAIR-1 morpholino antisense oligonucleotides or control oligonucleotides.
(B) Gating strategy of human ILC2s identified as CD45+ lineage− CD127+ CRTH2+ cells.
(C) Representative histogram of LAIR-1 expression on human ILC2s and corresponding quantification (right).
(D, E) Effect of morpholino treatment on LAIR-1 expression presented as MFI (D) and Ki67+ ILC2s percentage (E).
(F-H) Levels of IL-5 (F), IL-6 (G) and IL-13 (H).
(I-L) In vitro cultured human ILC2s were adoptively transferred into Rag2−/− Il2rg−/− mice. Recipient mice were intranasally challenged with 1 μg of rh-IL-33 or PBS for 3 consecutive days and intraperitoneally (i.p) injected with human C1q or PBS on days 4 and 5. Measurement of lung function and inflammation followed on day 6.
(J) Lung resistance; n = 4.
(K) Total number of eosinophils in the BAL gated as CD45+ SiglecF+ CD11c− cells.
(L) Total number of human ILC2s in lungs gated as CRTH2+ CD127+ cells.
Data are presented as means ± SEM (paired two-tailed Student’s t test in D to H; one-way ANOVA test for multiple comparisons; *p <0.05, **p < 0.01, ***p < 0.001).
Discussion
The present study is the first to elucidate the expression, the signaling pathways and the inhibitory potential of LAIR-1 axis in ILC2s in the context of allergic asthma. We demonstrated that the transmembrane glycoprotein inhibitory receptor LAIR-1, is inducible on mouse ILC2s upon activation. LAIR-1 deficiency increases type-2 cytokine production in association with an upregulation of PI3K/AKT signaling. We further evidenced the protective role of LAIR-1 in ILC2-dependent asthma. Interestingly, LAIR-1 is expressed and inducible on human ILC2s and downregulates their activation both in vitro and in vivo, suggesting a new potential target in allergic asthma.
LAIR-1 is a broadly expressed immune receptor, playing an inhibitory role on the majority of immune cells27,47,58. In neutrophils, LAIR-1 is lost during differentiation and is only expressed upon stimulation on mature cells30. No study has investigated yet the conditions under which ILC2s could express LAIR-1. Here we demonstrated that ILC2s precursors in the bone marrow as well as circulating ILC2s display a weak expression of LAIR-1, while tissue resident ILC2s do not express it. In agreement with recent studies highlighting the flexible nature of activated ILC2s1,59,60, pulmonary ILC2s could express LAIR-1 only upon stimulation. This suggests that LAIR-1 is induced on highly activated ILC2s to dampen their functions and its expression could necessitate a special inflammatory microenvironment. Besides the downregulation of genes related to inflammation, LAIR-1+ ILC2s highly express genes related to extracellular matrix generation and tissue remodeling. For instance, the gene encoding LOXL2, an enzyme that promotes collagen crosslinking39,61, was highly upregulated in LAIR-1+ ILC2s. In parallel, several studies have reported airway remodeling and collagen deposition in different asthma models62–64.Therefore, ILC2s may help providing an optimal microenvironment for LAIR-1 expression while sustaining bidirectional modulatory interactions with collagen-producing lung cells6.
Lacking antigen specific receptors, secretion of type-2 cytokine represents the main contribution for ILC2s in the orchestration of the immune response. Our data demonstrated, at the transcriptional and the protein levels, that LAIR-1 controls Th2 cytokine production in ILC2s, along with SHP-1 activation and the downregulation of PI3K/AKT as well as NF-κB signaling pathways. Therefore, we suggest here that the recruitment of SHP-1 phosphatase by LAIR-1 intracellular domain mediates an inhibitory cascade in ILC2s, leading to decreased cytokine production and proliferation. Previous studies showed that following LAIR-1 engagement, ITIM phosphorylation is a key event driving the suppression of main immune pathways, including interferon-α production in plasmacytoid DCs (pDCs)31,35 and TCR signaling in T cells65. Interestingly, a recent study has also demonstrated that LAIR-1/SHP-1 interaction triggers CD8+ T cell exhaustion in lung tumors39. Therefore, our findings support previous studies and decrypt the relevant mechanisms associated with LAIR-1 activity in pulmonary ILC2s.
A balance between activating and inhibitory signals is required to avoid inappropriate immune responses. Due to its broad inhibitory role, LAIR-1 acts as an immune checkpoint and is involved in cancer, autoimmune and infectious diseases. In particular, the development of RA is associated with an imbalanced expression of LAIR-1 on T cells and osteoclasts66,67. Furthermore, a decreased LAIR-1 expression on B cells and pDCs worsens SLE progression31,47. LAIR-1 also suppresses neutrophil inflammatory functions in viral bronchiolitis68,69. Although LAIR-1 crosslinking has become a promising therapeutic strategy against several diseases, only one study has investigated LAIR-1 involvement in allergic contexts and reported that LAIR-1 reduces cytokine production by DCs and limits T cell proliferation in allergic contact dermatitis model70. Here we investigated the implication of LAIR-1 in allergic asthma for the first time, using IL-33 and Alternaria alternata models. Our data support a protective role for LAIR-1 in allergic airway inflammation, as LAIR-1 deficiency is associated with exacerbated AHR and eosinophilia. Using adoptive transfer experiments, we demonstrated that LAIR-1 can suppress ILC2-dependent asthma primarily through ILC2 inhibition.
Human cohorts have confirmed the involvement of ILC2s in asthma progression and severity71,72. Although mouse asthma models are very useful to elucidate immunological mechanisms, we further validate our data in human ILC2s to be more clinically relevant. Interestingly, circulating ILC2s from healthy donors express LAIR-1. Similarly to mice, this expression is significantly inducible in response to IL-33, suggesting a critical role for LAIR-1 in ILC2 homeostasis and regulation. Due to the absence of specific LAIR-1 inhibitors and agonists to date, we used the morpholino gene silencing technique as well as C1q, in order to modulate LAIR-1 activity in vitro and in vivo, respectively. A transient knockdown of Lair1 in ILC2s led to increased proliferation and secretion of type-2 cytokines in response to IL-33, revealing a remarkable inhibitory role in human ILC2s. C1q was used in different human studies for its capacity to mediate immunosuppressive effect through LAIR-1 engagement35,65,73. It was also reported as a potent inhibitor in ovalbumin-induced allergic asthma model44. Since C1q is one of the main components of the total protein serum content37,74, it is very plausible that circulating ILC2s are highly exposed and may be regulated by C1q. To further develop our translational approach, we developed here a humanized mouse model reconstituted with human ILC2s and treated with human C1q after IL-33 challenge. Interestingly, C1q significantly reduces AHR and eosinophilia mainly through the regulation of ILC2s. Existing single cell RNAseq49 shows that luminal macrophages and activated DCs are the major sources of C1q in lungs of healthy and asthmatic patients (Fig E2, E). As ILC2s localize with DC subsets in lungs4,75, this might suggest the existence of unrecognized regulatory axis mediated by C1q/LAIR-1 interactions in allergic asthma.
Based on diverse experimental strategies, this study provides evidence for the implication of LAIR-1 in mouse and human ILC2s, and consequently in ILC2-dependent asthma. Therefore, our findings could be associated with clinical implications via the therapeutic modulation of LAIR-1 in allergic asthma.
Supplementary Material
Clinical implication:
LAIR-1 engagement may serve as potential therapeutic target for the treatment of ILC2-related diseases such as allergic asthma.
Acknowledgements
We are grateful to USC Libraries Bioinformatics Service for assisting with data analysis, in particular Dr. Yong-Hwee E Loh, Meng Li and Dr. Yibu Chen. The bioinformatics software and computing resources used in the analysis are funded by the USC Office of Research and the Norris Medical Library.
This article was financially supported by National Institutes of Health Public Health Service grants R01 ES025786, R01 ES021801, R01 HL144790, R01 HL151493, R01 AI145813 and R01 HL151769 (O.A.).
Abbreviations
- AHR
Airway hyperreactivity
- BAL
Bronchoalveolar lavage
- DC
Dendritic cell
- GSA
Gene specific analysis
- IL-
Interleukin-
- ILC2
Type-2 innate lymphoid cell
- ITIM
Immuno-receptor tyrosine-based inhibitory motif
- KO
Knock-out
- LAIR-1
Leukocyte-Associated Ig-like Receptor-1
- NK
Natural killer
- PBMC
Peripheral blood mononuclear cell
- pDC
Plasmacytoid dendritic cell
- RA
Rheumatoid arthritis
- RNAseq
RNA-sequencing
- SHP-1
Src homology phosphatase-1
- SLE
Systematic lupus erythematosus
- Th2
T-helper 2
- WT
Wild-type
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosure statement: The authors declare no competing interests.
References
- [1].Bal SM, Golebski K, Spits H. Plasticity of innate lymphoid cell subsets. Nat Rev Immunol 2020;20:552–65. [DOI] [PubMed] [Google Scholar]
- [2].Klose CSN, Artis D. Innate lymphoid cells control signaling circuits to regulate tissue-specific immunity. Cell Res 2020;30:475–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 2013;502:245–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Dahlgren MW, Jones SW, Cautivo KM, Dubinin A, Ortiz-Carpena JF, Farhat S, et al. Adventitial Stromal Cells Define Group 2 Innate Lymphoid Cell Tissue Niches. Immunity 2019;50:707–722.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Mindt BC, Fritz JH, Duerr CU. Group 2 Innate Lymphoid Cells in Pulmonary Immunity and Tissue Homeostasis. Front Immunol 2018;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Puttur F, Denney L, Gregory LG, Vuononvirta J, Oliver R, Entwistle LJ, et al. Pulmonary environmental cues drive group 2 innate lymphoid cell dynamics in mice and humans. Sci Immunol 2019;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Molofsky AB, Savage A, Locksley RM. Interleukin-33 in tissue homeostasis, injury and inflammation. Immunity 2015;42:1005–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Kaczmarek A, Vandenabeele P, Krysko DV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 2013;38:209–23. [DOI] [PubMed] [Google Scholar]
- [9].Maazi H, Akbari O. Type two innate lymphoid cells: the Janus cells in health and disease. Immunol Rev 2017;278:192–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Galle-Treger L, Sankaranarayanan I, Hurrell BP, Howard E, Lo R, Maazi H, et al. Costimulation of type-2 innate lymphoid cells by GITR promotes effector function and ameliorates type 2 diabetes. Nat Commun 2019;10:713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Shafiei-Jahani P, Hurrell BP, Galle-Treger L, Helou DG, Howard E, Painter J, et al. DR3 stimulation of adipose resident ILC2s ameliorates type 2 diabetes mellitus. Nat Commun 2020;11:4718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Poposki JA, Klingler AI, Tan BK, Soroosh P, Banie H, Lewis G, et al. Group 2 innate lymphoid cells are elevated and activated in chronic rhinosinusitis with nasal polyps. Immun Inflamm Dis 2017;5:233–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Gonzalez-Polo V, Pucci-Molineris M, Cervera V, Gambaro S, Yantorno SE, Descalzi V, et al. Group 2 innate lymphoid cells exhibit progressively higher levels of activation during worsening of liver fibrosis. Ann Hepatol 2019;18:366–72. [DOI] [PubMed] [Google Scholar]
- [14].Aron JL, Akbari O. Regulatory T cells and type 2 innate lymphoid cell-dependent asthma. Allergy 2017;72:1148–55. [DOI] [PubMed] [Google Scholar]
- [15].Howard E, Lewis G, Galle-Treger L, Hurrell BP, Helou DG, Shafiei-Jahani P, et al. IL-10 production by ILC2s requires Blimp-1 and cMaf, modulates cellular metabolism and ameliorates airway hyperreactivity. J Allergy Clin Immunol 2020. [DOI] [PubMed] [Google Scholar]
- [16].Hurrell BP, Howard E, Galle-Treger L, Helou DG, Shafiei-Jahani P, Painter JD, et al. Distinct Roles of LFA-1 and ICAM-1 on ILC2s Control Lung Infiltration, Effector Functions, and Development of Airway Hyperreactivity. Front Immunol 2020;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Bush A Pathophysiological Mechanisms of Asthma. Front Pediatr 2019;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Kuruvilla ME, Lee FE-H, Lee GB. Understanding Asthma Phenotypes, Endotypes, and Mechanisms of Disease. Clin Rev Allergy Immunol 2019;56:219–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Lambrecht BN, Hammad H, Fahy JV. The Cytokines of Asthma. Immunity 2019;50:975–91. [DOI] [PubMed] [Google Scholar]
- [20].Porsbjerg CM, Sverrild A, Lloyd CM, Menzies-Gow AN, Bel EH. Anti-alarmins in asthma: targeting the airway epithelium with next-generation biologics. Eur Respir J 2020;56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Group Kato A. 2 Innate Lymphoid Cells in Airway Diseases. Chest 2019;156:141–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Shafiei-Jahani P, Helou DG, Hurrell BP, Howard E, Quach C, Painter JD, et al. CD200–CD200R immune checkpoint engagement regulates ILC2 effector function and ameliorates lung inflammation in asthma. Nat Commun. 2021;12:2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Miyamoto C, Kojo S, Yamashita M, Moro K, Lacaud G, Shiroguchi K, et al. Runx/Cbfβ complexes protect group 2 innate lymphoid cells from exhausted-like hyporesponsiveness during allergic airway inflammation. Nat Commun 2019;10:447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].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;29:4509–4524.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Maazi H, Patel N, Sankaranarayanan I, Suzuki Y, Rigas D, Soroosh P, et al. ICOS:ICOS-Ligand interaction is required for type 2 innate lymphoid cell function, homeostasis and induction of airway hyperreactivity. Immunity 2015;42:538–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Helou DG, Shafiei-Jahani P, Lo R, Howard E, Hurrell BP, Galle-Treger L, et al. PD-1 pathway regulates ILC2 metabolism and PD-1 agonist treatment ameliorates airway hyperreactivity. Nat Commun 2020;11:3998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Meyaard L, Adema GJ, Chang C, Woollatt E, Sutherland GR, Lanier LL, et al. LAIR-1, a Novel Inhibitory Receptor Expressed on Human Mononuclear Leukocytes. Immunity 1997;7:283–90. [DOI] [PubMed] [Google Scholar]
- [28].Jansen CA, Cruijsen CWA, de Ruiter T, Nanlohy N, Willems N, Janssens-Korpela P-L, et al. Regulated expression of the inhibitory receptor LAIR-1 on human peripheral T cells during T cell activation and differentiation. Eur J Immunol 2007;37:914–24. [DOI] [PubMed] [Google Scholar]
- [29].Vries A-RV der V de, Clevers H, Logtenberg T, Meyaard L. Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is differentially expressed during human B cell differentiation and inhibits B cell receptor-mediated signaling. Eur J Immunol 1999;29:3160–7. [DOI] [PubMed] [Google Scholar]
- [30].Verbrugge A, Ruiter T de, Geest C, Coffer PJ, Meyaard L. Differential expression of leukocyte-associated Ig-like receptor-1 during neutrophil differentiation and activation. J Leukoc Biol 2006;79:828–36. [DOI] [PubMed] [Google Scholar]
- [31].Bonaccorsi I, Cantoni C, Carrega P, Oliveri D, Lui G, Conte R, et al. The Immune Inhibitory Receptor LAIR-1 Is Highly Expressed by Plasmacytoid Dendritic Cells and Acts Complementary with NKp44 to Control IFNα Production. PLOS ONE 2010;5:e15080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Lebbink RJ, de Ruiter T, Adelmeijer J, Brenkman AB, van Helvoort JM, Koch M, et al. Collagens are functional, high affinity ligands for the inhibitory immune receptor LAIR-1. J Exp Med 2006;203:1419–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Tang X, Narayanan S, Peruzzi G, Apara A, Natarajan K, Margulies DH, et al. A single residue, arginine 65, is critical for the functional interaction of leukocyte-associated inhibitory receptor-1 with collagens. J Immunol Baltim Md 1950 2009;182:5446–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Kim S, Easterling ER, Price LC, Smith SL, Coligan JE, Park J-E, et al. The Role of Leukocyte Associated Immunoglobulin-Like Receptor-1 (LAIR-1) in Suppressing Collagen-Induced Arthritis. J Immunol Baltim Md 1950 2017;199:2692–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Son M, Santiago-Schwarz F, Al-Abed Y, Diamond B. C1q limits dendritic cell differentiation and activation by engaging LAIR-1. Proc Natl Acad Sci 2012;109:E3160–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Son M, Diamond B, Volpe BT, Aranow CB, Mackay MC, Santiago-Schwarz F. Evidence for C1q-mediated crosslinking of CD33/LAIR-1 inhibitory immunoreceptors and biological control of CD33/LAIR-1 expression. Sci Rep 2017;7:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Reid KBM. Complement Component C1q: Historical Perspective of a Functionally Versatile, and Structurally Unusual, Serum Protein. Front Immunol 2018;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Sathish JG, Johnson KG, Fuller KJ, LeRoy FG, Meyaard L, Sims MJ, et al. Constitutive association of SHP-1 with leukocyte-associated Ig-like receptor-1 in human T cells. J Immunol Baltim Md 1950 2001;166:1763–70. [DOI] [PubMed] [Google Scholar]
- [39].Peng DH, Rodriguez BL, Diao L, Chen L, Wang J, Byers LA, et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8 + T cell exhaustion. Nat Commun 2020;11:4520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Wu X, Zhang L, Zhou J, Liu L, Fu Q, Fu A, et al. Clinicopathologic significance of LAIR-1 expression in hepatocellular carcinoma. Curr Probl Cancer 2019;43:18–26. [DOI] [PubMed] [Google Scholar]
- [41].Shafiei-Jahani P, Helou DG, Hurrell BP, Galle-Treger L, Howard E, Quach C, et al. CD52-targeted depletion by Alemtuzumab ameliorates allergic airway hyperreactivity and lung inflammation. Mucosal Immunol 2021:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Helou DG, Noël B, Gaudin F, Groux H, Ali ZE, Pallardy M, et al. Cutting Edge: Nrf2 Regulates Neutrophil Recruitment and Accumulation in Skin during Contact Hypersensitivity. J Immunol 2019:ji1801065. [DOI] [PubMed] [Google Scholar]
- [43].Kundu S, Fan K, Cao M, Lindner DJ, Zhao ZJ, Borden E, et al. Novel SHP-1 inhibitors TPI-1 and analogs with pre-clinical anti-tumor activities as tolerated oral agents. J Immunol Baltim Md 1950 2010;184:6529–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Mascarell L, Airouche S, Berjont N, Gary C, Gueguen C, Fourcade G, et al. The regulatory dendritic cell marker C1q is a potent inhibitor of allergic inflammation. Mucosal Immunol 2017;10:695–704. [DOI] [PubMed] [Google Scholar]
- [45].Bally I, Ancelet S, Moriscot C, Gonnet F, Mantovani A, Daniel R, et al. Expression of recombinant human complement C1q allows identification of the C1r/C1s-binding sites. Proc Natl Acad Sci 2013;110:8650–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Ozsoy Y, Gungor S, Cevher E. Nasal Delivery of High Molecular Weight Drugs. Molecules 2009;14:3754–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Guo N, Zhang K, Gao X, Lv M, Luan J, Hu Z, et al. Role and mechanism of LAIR-1 in the development of autoimmune diseases, tumors, and malaria: A review. Curr Res Transl Med 2020;68:119–24. [DOI] [PubMed] [Google Scholar]
- [48].Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour R-EE, Nyman J, Dionne D, et al. The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 2017;549:351–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Vieira Braga FA, Kar G, Berg M, Carpaij OA, Polanski K, Simon LM, et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat Med 2019;25:1153–63. [DOI] [PubMed] [Google Scholar]
- [50].Tang X, Tian L, Esteso G, Choi S-C, Barrow AD, Colonna M, et al. Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) deficient mice have an altered immune cell phenotype. J Immunol Baltim Md 1950 2012;188:548–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Hoxhaj G, Manning BD. The PI3K–AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer 2020;20:74–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Wen L-Z, Ding K, Wang Z-R, Ding C-H, Lei S-J, Liu J-P, et al. SHP-1 Acts as a Tumor Suppressor in Hepatocarcinogenesis and HCC Progression. Cancer Res 2018;78:4680–91. [DOI] [PubMed] [Google Scholar]
- [53].Snelgrove RJ, Gregory LG, Peiró T, Akthar S, Campbell GA, Walker SA, et al. Alternaria-derived serine protease activity drives IL-33–mediated asthma exacerbations. J Allergy Clin Immunol 2014;134:583–592.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Kim H-K, Lund S, Baum R, Rosenthal P, Khorram N, Doherty TA. Innate type 2 response to Alternaria extract enhances ryegrass-induced lung inflammation. Int Arch Allergy Immunol 2014;163:92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Doherty TA, Broide DH. Airway innate lymphoid cells in the induction and regulation of allergy. Allergol Int 2019;68:9–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Helfrich S, Mindt BC, Fritz JH, Duerr CU. Group 2 Innate Lymphoid Cells in Respiratory Allergic Inflammation. Front Immunol 2019;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Winkler C, Hochdörfer T, Israelsson E, Hasselberg A, Cavallin A, Thörn K, et al. Activation of group 2 innate lymphoid cells after allergen challenge in asthmatic patients. J Allergy Clin Immunol 2019;144:61–69.e7. [DOI] [PubMed] [Google Scholar]
- [58].Carvalheiro T, Garcia S, Pascoal Ramos MI, Giovannone B, Radstake TRDJ, Marut W, et al. Leukocyte Associated Immunoglobulin Like Receptor 1 Regulation and Function on Monocytes and Dendritic Cells During Inflammation. Front Immunol 2020;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Entwistle LJ, Gregory LG, Oliver RA, Branchett WJ, Puttur F, Lloyd CM. Pulmonary Group 2 Innate Lymphoid Cell Phenotype Is Context Specific: Determining the Effect of Strain, Location, and Stimuli. Front Immunol 2020;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].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;213:569–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Chopra V, Sangarappillai RM, Romero-Canelón I, Jones AM. Lysyl Oxidase Like-2 (LOXL2): An Emerging Oncology Target. Adv Ther 2020;3:1900119. [Google Scholar]
- [62].Liu G, Cooley MA, Nair PM, Donovan C, Hsu AC, Jarnicki AG, et al. Airway remodelling and inflammation in asthma are dependent on the extracellular matrix protein fibulin-1c. J Pathol 2017;243:510–23. [DOI] [PubMed] [Google Scholar]
- [63].An G, Zhang X, Wang W, Huang Q, Li Y, Shan S, et al. The effects of interleukin 33 on airways collagen deposition and matrix metalloproteinase expression in a murine surrogate of asthma. Immunology 2018;154:637–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Osei ET, B. Mostaço-Guidolin L, Hsieh A, Warner SM, AL-Fouadi M, Wang M, et al. Epithelial-interleukin-1 inhibits collagen formation by airway fibroblasts: Implications for asthma. Sci Rep 2020;10:8721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Park J-E, Brand DD, Rosloniec EF, Yi A-K, Stuart JM, Kang AH, et al. Leukocyte-associated immunoglobulin-like receptor 1 inhibits T-cell signaling by decreasing protein phosphorylation in the T-cell signaling pathway. J Biol Chem 2020;295:2239–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Zhang Y, Ding Y, Huang Y, Zhang C, Boquan J, Ran Z. Expression of leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) on osteoclasts and its potential role in rheumatoid arthritis. Clinics 2013;68:475–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Zhang Y, Lv K, Zhang CM, Jin BQ, Zhuang R, Ding Y. The role of LAIR-1 (CD305) in T cells and monocytes/macrophages in patients with rheumatoid arthritis. Cell Immunol 2014;287:46–52. [DOI] [PubMed] [Google Scholar]
- [68].Geerdink RJ, Hennus MP, Westerlaken GHA, Abrahams AC, Albers KI, Walk J, et al. LAIR-1 limits neutrophil extracellular trap formation in viral bronchiolitis. J Allergy Clin Immunol 2018;141:811–4. [DOI] [PubMed] [Google Scholar]
- [69].Kumawat K, Geerdink RJ, Hennus MP, Roda MA, van Ark I, Leusink-Muis T, et al. LAIR-1 Limits Neutrophilic Airway Inflammation. Front Immunol 2019;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Omiya R, Tsushima F, Narazaki H, Sakoda Y, Kuramasu A, Kim Y, et al. Leucocyte-associated immunoglobulin-like receptor-1 is an inhibitory regulator of contact hypersensitivity. Immunology 2009;128:543–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Smith SG, Chen R, Kjarsgaard M, Huang C, Oliveria J-P, O’Byrne PM, et al. Increased numbers of activated group 2 innate lymphoid cells in the airways of patients with severe asthma and persistent airway eosinophilia. J Allergy Clin Immunol 2016;137:75–86.e8. [DOI] [PubMed] [Google Scholar]
- [72].Silberbrandt A, Sverrild A, Jørgensen GØ, Hvidtfeldt M, Frøssing L, Pfluger LE, et al. The fraction of circulating type 2 innate lymphoid cells in peripheral blood is associated with asthma severity. Eur Respir J 2019;54. [Google Scholar]
- [73].Son M, Diamond B. C1q-mediated repression of human monocytes is regulated by leukocyte-associated Ig-like receptor 1 (LAIR-1). Mol Med Camb Mass 2015;20:559–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Fante MA, Decking S-M, Bruss C, Schreml S, Siska PJ, Kreutz M, et al. Heat-Inactivation of Human Serum Destroys C1 Inhibitor, Pro-motes Immune Complex Formation, and Improves Human T Cell Function. Int J Mol Sci 2021;22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Maazi H, Banie H, Aleman Muench GR, Patel N, Wang B, Sankaranarayanan I, et al. Activated plasmacytoid dendritic cells regulate type 2 innate lymphoid cell-mediated airway hyperreactivity. J Allergy Clin Immunol 2018;141:893–905.e6. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
