Skip to main content

Group 3 innate lymphoid cells secret neutrophil chemoattractants and are insensitive to glucocorticoid via aberrant GR phosphorylation

Abstract

Background

Patients with neutrophil-mediated asthma have poor response to glucocorticoids. The roles and mechanisms of group 3 innate lymphoid cells (ILC3s) in inducing neutrophilic airway inflammation and glucocorticoid resistance in asthma have not been fully clarified.

Methods

ILC3s in peripheral blood were measured by flow cytometry in patients with eosinophilic asthma (EA) and non-eosinophilic asthma (NEA). ILC3s were sorted and cultured in vitro for RNA sequencing. Cytokines production and signaling pathways in ILC3s after IL-1β stimulation and dexamethasone treatment were determined by real-time PCR, flow cytometry, ELISA and western blot.

Results

The percentage and numbers of ILC3s in peripheral blood was higher in patients with NEA compared with EA, and negatively correlated with blood eosinophils. IL-1β stimulation significantly enhanced CXCL8 and CXCL1 production in ILC3s via activation of p65 NF-κB and p38/JNK MAPK signaling pathways. The expression of neutrophil chemoattractants from ILC3s was insensitive to dexamethasone treatment. Dexamethasone significantly increased phosphorylation of glucocorticoid receptor (GR) at Ser226 but only with a weak induction at Ser211 residues in ILC3s. Compared to human bronchial epithelial cell line (16HBE cells), the ratio of p-GR S226 to p-GR S211 (p-GR S226/S211) was significantly higher in ILC3s at baseline and after dexamethasone treatment. In addition, IL-1β could induce Ser226 phosphorylation and had a crosstalk effect to dexamethasone via NF-κB pathway.

Conclusions

ILC3s were elevated in patients with NEA, and associated with neutrophil inflammation by release of neutrophil chemoattractants and were glucocorticoid (GC) resistant. This paper provides a novel cellular and molecular mechanisms of neutrophil inflammation and GC-resistance in asthma.

Trial registration The study has been prospectively registered in the World Health Organization International Clinical Trials Registry Platform (ChiCTR1900027125)

Background

Group 3 innate lymphoid cells (ILC3s) have crucial roles in immunity and tissue homeostasis [1, 2], which are widely distributed throughout the body and are constitutively present at mucosal barrier sites such as the lung, liver, gut, spleen, skin and secondary lymphoid tissues [3]. ILC3s lack the diversified antigen receptors expressed on T cells and B cells and are defined by expression of the Retinoid-related orphan receptorγt (RORγt) and the secretion of IL-17, IL-22 in response to IL-23 and IL1-β [4, 5]. A recent study found ILC3 (IL-17+RORγt+) cells were responsible for the development of airway hyperreactivity (AHR) induced by IL-1β in obese mice asthma model, and blockade of IL-1β abolished the AHR and reduced the number of ILC3 cells [6]. Meanwhile, these ILC3s were found to be increased in non-allergic neutrophilic asthma mice [7] and a similar IL-17+ ILCs were also found in bronchoalveolar lavage fluid from human patients with severe asthma[6]. However, the roles of ILC3 in asthmatic patients with airway neutrophilic inflammation are still not clear.

Asthma is a heterogeneous disease with two broad inflammatory phenotypes: eosinophilic asthma (EA) and non-eosinophilic asthma (mainly mediated by neutrophil) (NEA) [8, 9]. Neutrophil mediated asthma occurs in about 50% of corticosteroid resistant/insensitive asthma cases [10]. Evidence suggests IL-17 produced by Th17 cells appears to drive neutrophil-predominant steroid-resistant asthma [11, 12] and increases the production and release of chemokine IL-8 via airway epithelial cells, further propagating the chemotactic neutrophil response [13]. Since ILC3s can mimic the function of T helper type 17 (Th17) cells [1], they may also contribute to corticosteroid resistance in asthma, which is supported by a recent study that found the proportions of lung ILC3s in a murine neutrophilic asthma model was not altered by fluticasone propionate treatment [7]. However, the mechanisms behind the ILC3 associated steroid resistance are unknown. Therefore, in this study we investigated the relationship between ILC3s and neutrophilic inflammation in asthma patients and the mechanisms of corticosteroid resistance, which will provide a better understanding of the innate immunity and treatment target for NEA.

Methods

Human subjects

Adult patients with asthma were consecutively recruited from Respiratory Outpatient Department of West China Hospital, Sichuan University from June 2019 to December 2020, according to diagnosis of Global Initiative for Asthma (GINA) recommendations [14]. Patients were stratified by baseline blood eosinophil counts 300/μL or greater and less than 300 cells/μL as eosinophilic asthma (EA) and non-eosinophilic asthma (NEA), respectively [15, 16]. Detailed inclusion and exclusion criteria for subjects were described as previously reported [17]. The healthy control population enrolled healthy volunteers from West China Hospital. The study was approved by the Institutional Review Board (IRB) at West China Hospital, Sichuan University (Chengdu, China) (No. 2019–856). All participants provided written informed consent.

Flow cytometry analysis of peripheral blood

Fresh blood was collected in EDTA-treated tubes and peripheral blood mononuclear cells (PBMCs) were isolated as previously reported [18, 19] and stained with Zombie Aqua (Live/Dead) together with antibody panel 1(Additional file 4: Table S1). Lineage (Lin) markers included CD19/CD56/CD14/CD11b/CD11c/CD123/FcεRI. Group 2 innate lymphoid cell (ILC2) was defined as LinCD45+CD3CD4CD8CD127+CRTH2+, Group 1 innate lymphoid cell (ILC1) was defined as LinCD45+CD3CD4CD8CD127+CRTH2CD117 and ILC3 was defined as Lin-CD45+CD3CD4CD8CD127+CRTH2CD117+ [20,21,22] (Additional file 1: Fig. S1).

Cell lines and reagents

Human bronchial epithelial cell line (16HBE cells) were received as a gift from Tianfu Life Science Center of Sichuan University West China Hospital. Human IL-2, recombinant human IL-1β and IL-23 were purchased from Peprotech. NF-κB inhibitor (TPCA-1), p38 inhibitor (SB203580) and JNK inhibitor (SP600125) were purchased from Selleck. Dexamethasone was purchased from Sigma.

ILC3s sorting and culture

PBMCs from healthy donors were labeled with the antibody panel 2 (Additional file 4: Table S1) after CD3 depletion by using CD3 Microbeads (Miltenyi Biotech). ILC3 cells were sorted with the FACSAria III (BD, Franklin Lakes, NJ).

The cells were cultured in RPMI with 10% human serum, 1× l-glutamine, 1× penicillin/streptomycin, 1× sodium pyruvate, 1× nonessential amino acids, and 250 U/mL IL-2. Details were described in our previously published study [23].

Intracellular staining

For RORγt staining, cultured ILC3s were incubated with antibody for RORγt after surface staining with Zombie Aqua (Live/Dead) together with antibody panel 3 (Additional file 4: Table S1) and fixation/permeabilization with FOXP3/Transcription Factor Staining Buffer set (eBioscience) [24]. ILC3 was defined as CD3-RORγt+. Cells with purity ≥ 90% were used for subsequent experiments.

For staining of IL-22, IL-17A and CXCL8, cultured ILC3s were stimulated with Phorbol 12-myristate 13-acetate (PMA) cocktail in the presence of Brefeldin A (Cell Activation Cocktail with Brefeldin, Biolegend) for 6 h at 37 ℃. After staining with Zombie Aqua (Live/Dead) and fixation with IC Fixation Buffer (eBioscience), cells were permeabilized with a permeabilization buffer (Invitrogen) and labeled with corresponding antibodies (Additional file 4: Table S1). The above samples were acquired on a BD LSR II flow cytometer.

RNA extraction and quantitative real-time polymerase chain reaction (PCR)

Total RNA was extracted from ILC3 cells using RNeasy Mini Kit (QIAGEN) following the manufacturer’s instructions and reverse-transcribed to cDNA using HiScript III RT SuperMix for qPCR (+gDNA wiper) Kit (Vazyme, China). Primers sequences were presented in Additional file 4: Table S2. Data correction was performed using Bio-Rad CFX Maestro and analyzed by 2−ΔΔCt method.

RNA sequencing (RNA-seq)

ILC3s were stimulated with or without IL-1β + IL-23 for 48 h before sending to Beijing Novogene Bioinformation Technology Co., Ltd. (Beijing, China) in TRIzol Reagent for RNA-Seq. Standard methods were used to extract RNA, and RNA integrity was assessed using the RNA Nano 6000 Assay Kit of the Bioanalyzer. 1 μg RNA was used for cDNA library preparation after purification using Poly-T oligo-attached magnetic beads. Sequencing was performed on the Illumina Novaseq 6000 (Novogene Bioinformatics Technology Co., Ltd., Tianjin, China) platform [25].

Enzyme-linked immunosorbent assay (ELISA)

Cell supernatants were analyzed by Human CXCL8 Uncoated ELISA kit (Thermo Fisher Scientific) and human CXCL1 ELISA kit (Elabscience, China) following manufacturer’s recommendations.

Western blot

Proteins of cells were extracted with radioimmunoprecipitation (RIPA) lysis buffer (Beyotime, China) with phenylmethanesulfonyl fluoride (PMSF) and protease inhibitor cocktail and quantified by BCA Protein Assay Kit (Beyotime, China). The process was performed as previously reported [26]. Antibody and antigen complexes were detected using ECL chemiluminescent kit (Beyotime, China) by ChemiDoc™ MP Imaging System (Bio-Rad, USA). Antibodies used in western blot were presented in Additional file 4: Table S3.

Statistical analysis

Continuous variables are expressed as mean ± standard deviation (SD) or median (interquartile range, IQR). Categorical variables are summarized as frequencies and proportions. Differences between groups were analyzed with Student’s t test or Mann–Whitney test. And for three or more groups, Analysis of Variance or the Kruskal–Wallis test were performed, Tukey HSD (Tukey Honest Significant Differences) were used for multiple comparisons as post hoc test. Categorical data were compared using Chi-square test or Fisher exact test. Test of Spearman was performed for evaluating correlations. Differences were considered statistically significant at *P < 0.05, **P < 0.01, ***P < 0.001 and ****P < 0.0001. Data was analyzed by Prism 6.0 (GraphPad) and flow cytometry data was analyzed by FlowJo 9.7.6 software (Tree Star).

For sequencing data, differential gene expression analysis was performed using the DESeq2 R package (1.20.0). The P-value was adjusted using the Benjamini & Hochberg method for controlling the false discovery rate. Genes with adjusted P-value < 0.05 and absolute value of log2(FoldChange) ≥ 1 were assigned as differentially expressed genes. Gene Ontology (GO) enrichment analysis and KEGG pathways analysis of differentially expressed genes were conducted by the clusterProfiler R package, and terms with adjusted P-value < 0.05 were considered significantly enriched.

Results

ILC3 counts are higher in patients with NEA and they show a negative correlation with blood eosinophils

Twenty patients with NEA and eleven patients with EA were enrolled in this study, respectively. And eleven healthy donors were included as healthy control (HC). In the NEA group, blood eosinophils count (170.00 ± 87.12 × 106/L vs 570.00 ± 201.15 × 106/L, P < 0.001) and percentage (2.95 [1.30, 4.23]% vs 9.70 [5.70, 13.50]%, P < 0.001), fractional exhaled nitric oxide (FeNO) (54.00 [27.25, 68.25] ppb vs 87.00 [60.00, 153.0]ppb, P = 0.032) were lower compared to the EA group (Table 1, Fig. 1A), and the percentage of blood neutrophils was higher (Additional file 2: Fig. S2A). To determine the difference of ILC subsets between EA and NEA, the proportions of ILC subsets in peripheral blood were measured (Additional file 1: Fig. S1). Due to the imbalance of age between HC and patients with asthma, we firstly checked the relationship between age and ILC subsets and found a negative correlation with ILC3 (r = − 0.29, P = 0.067) and positive correlation with ILC2 (r = 0.35, P = 0.023) but not ILC1(Fig. 1B). After adjusting for age, the percentage (18.33 ± 5.64% vs 9.30 ± 8.16%, P = 0.0212) and numbers (139.00 [89.50, 224.50] vs 45.00 [2.00, 135.00], P = 0.021) of ILC3s in NEA group was higher than EA group (Fig. 1C). The percentage of ILC3s was negatively correlated with blood eosinophils counts (r = − 0.57, P < 0.001) and percentage (r = − 0.55, P = 0.0013) (Fig. 1D), but was not correlated with blood neutrophils counts and percentage (Additional file 2: Fig. S2B).

Table 1 Demographic and clinical characteristics of study subjects
Fig. 1
figure 1

ILC3s elevated in patients with NEA compared to EA. A Blood eosinophils count and percentage among healthy control (HC) and patients with NEA and EA. B Correlation between subsets of ILCs and age. C Percentage and numbers (in every 50 × 105 recorded lymphocyte) of ILC1, ILC2 and ILC3 in HC, NEA and EA patients. D Correlation between ILC subsets and blood eosinophils count and percentage. ****p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05, ns p > 0.05

IL-1β induces neutrophil chemoattractants expression in ILC3s

In order to explore relationship between ILC3s and neutrophilic inflammation, ILC3s were sorted and cultured in vitro. Because of plasticity in vitro [27, 28], ILC3s were first identified by flow cytometry based on the transcription factor RORγt [4], and only cells with RORγt+ILC3 purity ≥ 90% were used for further experiments. Meanwhile, the cytokines IL-17A and IL-22 secreted by in vitro cultured cells were explored for further confirmation [5]. So, intracellular staining of transcription factor (RORγt) and cytokines (IL-17A and IL-22) by flow cytometry was performed, and results showed the expression of RORγt and production of IL-17A and IL-22 upon PMA stimulation in the in vitro cultured cells (Fig. 2A), confirming these cells were ILC3s.

Fig. 2
figure 2

Flow cytometry gating strategy for ILC3 identification and cytokines production profiles of ILC3s. A ILC3s were identified by transcription factor RORγt and the secretion of IL-17, IL-22 and CXCL8. Cytokines-producing cells of ILC3s were detected by Flow cytometry. Blue, ILC3s without stimulation. Red, ILC3s stimulated with PMA + ionomycin. Numbers represent percentages of cells producing IL-17A or IL-22 or CXCL8 in ILC3s. B Time course of CXCL8, CXCL1, TNF-α and GM-CSF mRNA expression in ILC3s after IL-1β (50 ng/mL) stimulation. C Protein level of CXCL8 and CXCL1 in culture supernatant of ILC3s with or without IL-1β (50 ng/mL) stimulation after 24 h. D, E Effect of IL-1β and IL-23 on CXCL8 expression in ILC3s

Next, we measured the capability of release of neutrophil chemoattractants on ILC3s. CXCL8, one of the most potent chemokines for neutrophil migration [29, 30], was significantly enhanced after PMA or IL-1β stimulation in ILC3s at both transcriptome and protein level, peaked at 30 min and 4 h, respectively (Fig. 2A–C). In addition, IL-1β also induced production of other neutrophil chemoattractants in ILC3s including CXCL1, Tumor necrosis factor (TNF-α) and Granulocyte–macrophage colony-stimulating factor (GM-CSF) (Fig. 2B, C). Previous study reported the stimulation of ILC3s by IL-1β plus IL-23 [31,32,33], therefore, we sought to determine whether there is any additive or synergistic effect of IL-1β and IL-23. We found that IL-1β + IL-23 significantly increased the production of CXCL8 in ILC3s in a similar extent as IL-1β alone, and IL-23 alone did not induce CXCL8 production, indicating no additive or synergistic stimulation effect of IL-1β and IL-23 in ILC3s (Fig. 2D, E).

ILC3s are involved in inflammation response via NF-κB and MAPK pathways

To investigate the pathways that regulate the production of neutrophil chemoattractants in ILC3s, RNA sequencing was performed on in vitro cultured ILC3s with or without IL-1β + IL-23 stimulation. IL-1β + IL-23 stimulation was associated with 760 up-regulated and 450 down-regulated genes, and the top five overexpressed genes were IL-22, CXCL8, CCR7, IGFBP4, and CD22 (Fig. 3A). IL-1β + IL-23 induced expression of cytokines and chemokines such as CSF2, TNFSF4, IL-22, CXCL8 and IL-17(Fig. 3B), which have been previous identified to be effector cytokines of ILC3s [5]. The GO gene set enrichment analyses showed response to inflammation was an important biological process of ILC3s (Fig. 3C, left panel), and CXCL8 was involved in “inflammatory response” during cell activation (Fig. 3D).

Fig. 3
figure 3

RNA sequencing of in vitro cultured ILC3s with or without IL-1β + IL-23 stimulation. A Volcano plots of differential expression genes (DEGs) in ILC3s stimulated by IL-1β + IL-23. The top five differential expressed genes were IL-22, CCR7, IGFBP4, CD22 and CXCL8. B Heatmap analysis of effector cytokines in ILC3s. In the heatmap, the redder the color is, the higher the expression is, and the greener the expression is, the lower the expression is. C Function terms of DEGs by Gene Ontology (GO) enrichment analysis and KEGG pathway analysis of DEGs in ILC3s upon IL-1β + IL-23(50 ng/mL) stimulation. D Heatmap analysis of DEGs in inflammation response. E The clustering heat map of DEGs involved in NF-κB signal pathway. F, G Western blot analysis by antiphospho-p65 Ab, antiphospho-p38 Ab, antiphospho-JNK Ab and antiphospho-Erk Ab. Cell extracts were prepared after stimulation with IL-1β (50 ng/mL) for indicated times. H Phosphorylation and inhibition of p65, p38 and JNK by specific inhibitors in ILC3s. Western blot analysis of p-p65, p-p38 and p-JNK were performed with cell extracts from ILC3s, preincubated for 1 h with p65 NF-κB inhibitor TPCA-1, p38-MAPK inhibitor SB203580 or JNK-MAPK inhibitor SP600125 and then stimulated with IL-1β (50 ng/mL) for 10 min. I Expression of CXCL8 protein in culture supernatant of ILC3 cells after IL-1β stimulation with or without corresponding inhibitors. ILC3s were preincubated for 1 h with indicated inhibitors and then stimulated with IL-1β (50 ng/mL) for 24 h

The KEGG pathway analysis showed the NF-κB (P = 0.006) (Fig. 3C, right panel) and MAPK signaling pathway (P = 0.071) (data not shown) were participated in ILC3s upon IL-1β + IL-23 stimulation. Further enrichment analysis also showed CXCL8 was involved in the NF-κB signaling pathway (Fig. 3E). To verify these signaling pathways, we detected the activation of key proteins in NF-κB and MAPK pathways in ILC3s by western blotting, and found IL-1β stimulation induced phosphorylation of p65, p38 and JNK, but had no effect on the activation of Erk (Fig. 3F, G). These increased phosphorylation were significantly inverted by the corresponding inhibitors (Fig. 3H). The enhancement of CXCL8 level by IL-1β was completely inhibited by p65 NF-κB inhibitor (TPCA-1) and partially inhibited by p38-MAPK inhibitor (SB203580) and JNK-MAPK inhibitor (SP600125) (Fig. 3I).

IL-1β-induced neutrophil chemoattractants expression in ILC3s is insensitive to dexamethasone due to aberrant GR phosphorylation

Patients with neutrophilic asthma usually have poor response to corticosteroids [34,35,36], and our previous study has found that the numbers of ILC3s in asthma patients were not reduced after treatment with prednisolone [23]. Therefore, we speculated that ILC3s might contribute to the steroid resistance. To test this hypothesis, in vitro cultured ILC3s were treated with dexamethasone (Dex) and the expression levels of CXCL8 and CXCL1 were detected. 16HBEs were selected as reference cells, which were sensitive to dexamethasone treatment, as Dex significantly reduced CXCL8 production induced by IL-1β (Fig. 4A). We found that the CXCL8 and CXCL1 production of ILC3s in response to IL-1β was not inhibited by Dex at protein level, but partially reduced by Dex at transcriptome level (Fig. 4B, C). These findings suggest the steroid resistance of ILC3s in terms of neutrophil chemoattractants expression.

Fig. 4
figure 4

Effect of dexamethasone (Dex) on IL-1β-induced CXCL8 and CXCL1 production and phosphorylation of glucocorticoids receptor (GR) at S226 and S211 in ILC3s. A Expression of CXCL8 in 16HBEs with and without Dex treatment. B, C, Expression of CXCL8 and CXCL1 in ILC3s with Dex treatment by indicated concentrations. Cells were pre-treated with Dex (10 to 1000 ng/mL) for 1 h before 24 h stimulation with IL-1β (50 ng/mL), supernatants were collected and assayed for CXCL8 and CXCL1 release by ELISA, and ILC3 cells were extracted for qPCR analysis. D Western blotting analysis of GR in 16HBEs and ILC3s after stimulation with Dex (1000 ng/mL) for 15 min. E, F Western blotting analysis of phosphorylation of GR S226 and S211 in 16HBEs (E) and ILC3s (F) after stimulation with Dex (1000 ng/mL) for 15 min. G Ratio of p-GR S226/S211 in 16HBEs and ILC3s at baseline and after dexamethasone treatment. H Western blotting analysis of phosphorylation of MAPK in ILC3s with Dex stimulation (1000 ng/mL) for 15 min. I Effects of Dex and corresponding inhibitors on phosphorylated GR at S226 and S211 in ILC3s. J GR phosphorylation at S226 and S211 in ILC3s with indicated stimulation. Cells were stimulated with IL-1β (50 ng/mL) or dexamethasone (1000 ng/mL) alone or combination for 15 min. ILC3s were preincubated for 1 h with corresponding inhibitors then stimulated with dexamethasone (1000 ng/mL) for 15 min, then lysed and assessed for phosphorylation of GR S226 or GR S211 by Western blotting

Next, we investigated the mechanisms that mediate steroids resistance in ILC3s. The effects of glucocorticoids (GCs) are mediated by their binding and activation of GC receptor (GR) [37], and phosphorylation status and sites of GR can dictate cells respond to GCs [38]. Therefore, we firstly compared the levels of GR between ILC3s and 16HBEs, and no difference was found between ILC3s and 16HBEs at baseline or after treatment of dexamethasone (Fig. 4D). Then, we measured the phosphorylation levels of two sites of GR, i.e. p-GR S226 and p-GR S211, and the results showed that the level of p-GR S226 was significantly up-regulated by Dex in both 16HBEs and ILC3s. However, there was a significant increase in the phosphorylation of GR S211 in 16HBEs and only a weak increase in ILC3s in response to Dex treatment (Fig. 4E, F and Additional file 3: Fig. S3). In addition, we also found that the ratio of p-GR S226 to p-GR S211 (p-GR S226/S211) was significantly higher in ILC3s than 16HBEs at baseline and after dexamethasone treatment (Fig. 4G). Taken together, these data indicate the steroid resistance of ILC3s is mediated by the enhanced phosphorylation at Ser226 and weak phosphorylation of GR at Ser211, with a higher ratio of p-GR S226/S211 than that of 16HBEs.

Finally, we explored the potential signaling pathways that regulate phosphorylation of GR in ILC3s. MAPK signaling pathway has been found to directly phosphorylate GR [38], and in ILC3s we found Dex treatment increased JNK MAPK phosphorylation, but not phosphorylation of p38 MAPK or Erk MAPK (Fig. 4H), which was further confirmed by the inhibitory experiment showing that the GR phosphorylation at S226 in response to dexamethasone was significantly inhibited by JNK-MAPK inhibitor SP600125 (Fig. 4I). Interestingly, we also found p65 NF-κB inhibitor (TPCA-1) also abolished the effect of Dex on phosphorylation of S226 (Fig. 4I), indicating a role of NF-κB signaling pathway in GR activation. Indeed, evidence has shown GR and NF-κB interacted with each other, dexamethasone treatment could reduce the binding of NF-kB chromatin and high levels of NF-κB attenuated GR function [39]. Since IL-1β significantly increased activity of NF-κB and JNK MAPK (Fig. 3F, G), we then sought to explore the crosstalk effects of dexamethasone and IL-1β in ILC3s. As expected, the level of GR phosphorylation at S226 was also significantly elevated by IL-1β treatment alone or combination with dexamethasone for 15 min (Fig. 4J), suggesting the crosstalk effect between dexamethasone and IL-1β in corticosteroids-insensitivity in ILC3s.

Discussion

The pro-inflammatory roles of ILC3 in asthma AHR and inflammation have been known for several years [6], but the underlying mechanisms have not been fully illustrated. Previous studies have reported the production of IL-17 by ILC3 might be a key contributor to the airway neutrophilic inflammation through neutrophil recruitment in patients with asthma [40,41,42]. Here, we illustrated that percentage of ILC3s in peripheral blood of NEA patients was increased compared with EA patients and were negatively correlated with blood eosinophils. However, a phase II study with brodalumab (a human anti-IL-17 receptor monoclonal antibody) failed to show the treatment effect in subjects with moderate to severe asthma [43], which suggests other mechanisms might mediate neutrophilic inflammation in asthma. In our study, we found ILC3s produced neutrophil chemoattractants including CXCL8, CXCL1, TNF-α and GM-CSF after IL-1β stimulation, suggesting ILC3s may mediate airway neutrophilia in asthma by release of neutrophil chemoattractants. Meanwhile, we noticed a relatively high level of CXCL8 and CXCL1 released by ILC3s without stimulation, this might suggest the contribution of ILC3s in the recruitment of neutrophils in physiological condition but we could not exclude the possibility that they were due to the effect of rIL-2 in the culture medium [44, 45].

One of the clinical characteristics of patients with NEA is the poor response to corticosteroids, leading to a higher severity of disease and difficult-to-control asthma [46]. In this study, the expression of CXCL8 and CXCL1 in ILC3s upon IL-1β stimulation was insensitive to dexamethasone, suggesting that neutrophilic inflammation mediated by IL-1β-ILC3-CXCL8/CXCL1 axis may be involved in the development of steroid-resistant of asthma. To find out the mechanisms that mediate this insensitivity, we focused on the GR, which is the key receptor responsible for the physiological and pharmacological effects of glucocorticoids. GR can be phosphorylated at over 20 sites, while Ser211 and Ser226 are the two well-characterized sites. Phosphorylation at Ser211 promotes nuclear translocation and enhances transcriptional activity, while phosphorylation at Ser226 inhibits transcriptional activity and promotes nuclear export and has an inhibitory effect on GR function [38, 47]. The lack of Ser211 phosphorylation has been suggested to be a possible mechanism for GC resistance of human lymphoid cells [48]. Evidences have also shown the expression of GR Ser226 in PBMCs from severe asthma patients was significantly higher than that from non-severe asthma patients after dexamethasone treatment [49, 50]. In our study, we found the enhanced Ser226 phosphorylation while weak Ser211 phosphorylation upon dexamethasone treatment in ILC3s, and compared to GC-sensitive 16HBEs, the ratio of p-GR S226 to p-GR S211 (p-GR S226/S211) was significantly higher in ILC3s at baseline and after dexamethasone treatment, which well explained the insensitivity of ILC3s to dexamethasone treatment.

The phosphorylation of GR has been reported to be controlled by the p38 MAPK and Erk/JNK MAPK signaling pathways, of which p38 MAPK promotes the Ser211 phosphorylation while Erk/JNK MAPK down-regulates Ser211 phosphorylation and increases Ser226 phosphorylation [48, 51, 52]. In our study, we confirmed the regulatory effect of JNK MAPK on Ser226 phosphorylation by using its inhibitor SP600125, and the JNK MAPK activity was enhanced by dexamethasone treatment but not p38 MAPK activity, which reveals the intrinsic regulatory mechanisms associated with insensitivity of ILC3 to corticosteroids. Furthermore, phosphorylation of GR usually happens after binding to its ligand (GC), but a ligand-independent fashion as part of the crosstalk with other signaling pathways has also been reported [38]. A study of ligand-independent phosphorylation of GR showed TNF-α induced phosphorylation of the unliganded human GR at Ser-226 not Ser-211 [53]. In our study, we found IL-1β could induce Ser226 phosphorylation and the NF-κB inhibitor TPCA-1 demolished this effect in ILC3s. Meanwhile, IL-1β and dexamethasone could activate the same signaling pathway of JNK MAPK in ILC3s. This evidence implies that elevated IL-1β in neutrophil dominant asthma may have a crosstalk effect on dexamethasone [54], which aggravates corticosteroids insensitivity in neutrophilic asthma [55]. However, we could not exclude the possibility that the IL-1β induced phosphorylation at Ser226 might be through TNF-α, which has been shown to impair glucocorticoid receptor phosphorylation at Ser211 [56, 57] and directly phosphorylate GR at Ser226 by activating JNK MAPK [58], because in our study, TNF-α was enhanced by IL-1β in ILC3s.

However, there are several limitations that need to be addressed in this study. Firstly, due to the COVID-19 pandemic, we were unable to obtain induced sputum samples from asthma patients, thus unable to detect ILC3 in airways and distinguish neutrophilic asthma from NEA; therefore, EA and NEA were classified according to blood eosinophil counts 300/μL. Secondly, different subgroups of ILC3 have not been investigated, which could provide more information of ILC3 heterogeneity. Third, we did not directly compare the stimulation responses of ILC3 from healthy control and different asthma phenotypes/severities, which might imply the functional differences of ILC3 in the context of disease and asthma phenotypes. Fourth, we did not investigate other potential mechanisms that could be involved in the regulation of glucocorticosteroids such as GR isoforms (GRβ) and the lack of GRE in CXCL8 gene. Finally, we focused on patients with asthma, however, it would be interesting to investigate the roles of ILC3 in a broader spectrum of diseases such as patients with chronic obstructive pulmonary disease (COPD) as they are generally less sensitive to inhaled steroids and age dependent, and evidence showed ILC3s were notably increased in lung tissue from patients with COPD [59].

In conclusion, ILC3s are elevated in patients with NEA compared to EA and are crucial contributors to the neutrophilic airway inflammation via production of neutrophil chemoattractants through p65 NF-κB and p38/JNK MAPK signaling pathways. Specifically, the expression of neutrophil chemoattractants induced by IL-1β is insensitive to dexamethasone, which is due to aberrant GR phosphorylation with increased phosphorylation at Ser226 and weak phosphorylation at Ser211. Our findings provide evidence of the roles of innate immunity in neutrophilic inflammation and steroids insensitivity, which help to discover novel treatment targets for future drug development and better management of neutrophilic phenotype of asthma.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

ACT:

Asthma Control Test

AHR:

Airway hyperreactivity

BAL:

Bronchoalveolar lavage

BCA:

Bicinchoninic acid

BMI:

Body mass index

BP:

Biological process

CC:

Cellular component

CXCL:

C-X-C motif ligand

Dex:

Dexamethasone

DNA:

Deoxyribonucleic acid

EA:

Eosinophilic asthma

ELISA:

Enzyme-linked immunosorbent assay

Erk:

Extracellular-signal-regulated kinase

FeNO:

Fractional exhaled nitric oxide

GC:

Glucocorticoid

GINA:

Global Initiative for Asthma

GM-CSF:

Granulocyte–macrophage colony-stimulating factor

GO:

Gene Ontology

GR:

Glucocorticoid receptor

16HBE:

Human bronchial epithelial cell line

HC:

Healthy control

IFN:

Interferon

IgE:

Immunoglobulin E

IL:

Interleukin

ILC:

Innate lymphoid cell

ILC1:

Group 1 innate lymphoid cell

ILC2:

Group 2 innate lymphoid cell

ILC3:

Group 3 innate lymphoid cell

IRB:

Institutional Review Board

JNK:

C-jun N-terminal kinase

MAPK:

Mitogen-activated protein kinase

MF:

Molecular function

MMP-9:

Matrix metalloprotease 9

mRNA:

Messenger Ribose Nucleic Acid

NE:

Neutrophil elastase

NEA:

Non-eosinophilic asthma

NF-kB:

Nuclear factor kB

PBMC:

Peripheral blood mononuclear cell

PCR:

Polymerase chain reaction

PMA:

Phorbol 12-myristate 13-acetate

PMSF:

Phenylmethanesulfonyl fluoride

RIPA:

Radio immunoprecipitation

RNA:

Ribose Nucleic Acid

RORγt:

Retinoid-related orphan receptorγt

RT-PCR:

Real-time polymerase chain reaction

SD:

Standard deviation

Th17 cells:

T helper 17 cells

TNF:

Tumor necrosis factor

References

  1. Spits H, Artis D, Colonna M, et al. Innate lymphoid cells–a proposal for uniform nomenclature. Nat Rev Immunol. 2013;13(2):145–9.

    Article  CAS  PubMed  Google Scholar 

  2. Vivier E, Artis D, Colonna M, et al. Innate Lymphoid Cells: 10 Years On. Cell. 2018;174(5):1054–66.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ardain A, Porterfield JZ, Kløverpris HN, et al. Type 3 ILCs in lung disease. Front Immunol. 2019;10:92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  7. Jonckheere AC, Seys SF, Steelant B, et al. Innate lymphoid cells are required to induce airway hyperreactivity in a murine neutrophilic asthma model. Front Immunol. 2022;13: 849155.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gibson PG, Simpson JL, Saltos N. Heterogeneity of airway inflammation in persistent asthma: evidence of neutrophilic inflammation and increased sputum interleukin-8. Chest. 2001;119(5):1329–36.

    Article  CAS  PubMed  Google Scholar 

  9. Douwes J, Gibson P, Pekkanen J, et al. Non-eosinophilic asthma: importance and possible mechanisms. Thorax. 2002;57(7):643–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ray A, Kolls JK. Neutrophilic inflammation in asthma and association with disease severity. Trends Immunol. 2017;38(12):942–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chesné J, Braza F, Mahay G, et al. IL-17 in severe asthma. Where do we stand? Am J Respir Crit Care Med. 2014;190(10):1094–101.

    Article  PubMed  Google Scholar 

  12. McKinley L, Alcorn JF, Peterson A, et al. TH17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice. J Immunol. 2008;181(6):4089–97.

    Article  CAS  PubMed  Google Scholar 

  13. Ramakrishnan RK, Al HS, Hamid Q. Role of IL-17 in asthma pathogenesis and its implications for the clinic. Expert Rev Respir Med. 2019;13(11):1057–68.

    Article  CAS  PubMed  Google Scholar 

  14. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention, 2020.  www.ginasthma.org.

  15. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet. 2012;380(9842):651–9.

    Article  CAS  PubMed  Google Scholar 

  16. FitzGerald JM, Bleecker ER, Nair P, et al. Benralizumab, an anti-interleukin-5 receptor α monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2016;388(10056):2128–41.

    Article  CAS  PubMed  Google Scholar 

  17. Wang G, Wang F, Gibson PG, et al. Severe and uncontrolled asthma in China: a cross-sectional survey from the Australasian Severe Asthma Network. J Thorac Dis. 2017;9(5):1333–44.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Cosío BG, Mann B, Ito K, et al. Histone acetylase and deacetylase activity in alveolar macrophages and blood mononocytes in asthma. Am J Respir Crit Care Med. 2004;170(2):141–7.

    Article  PubMed  Google Scholar 

  19. Björklund ÅK, Forkel M, Picelli S, et al. The heterogeneity of human CD127(+) innate lymphoid cells revealed by single-cell RNA sequencing. Nat Immunol. 2016;17(4):451–60.

    Article  PubMed  Google Scholar 

  20. Mazzurana L, Bonfiglio F, Forkel M, et al. Crohn’s Disease Is Associated With Activation of Circulating Innate Lymphoid Cells. Inflamm Bowel Dis. 2021;27(7):1128–38.

    Article  PubMed  Google Scholar 

  21. Mazzurana L, Forkel M, Rao A, et al. Suppression of Aiolos and Ikaros expression by lenalidomide reduces human ILC3-ILC1/NK cell transdifferentiation. Eur J Immunol. 2019;49(9):1344–55.

    Article  CAS  PubMed  Google Scholar 

  22. Mjösberg JM, Trifari S, Crellin NK, et al. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol. 2011;12(11):1055–62.

    Article  PubMed  Google Scholar 

  23. Luo J, Wu Y, Shrimanker R, et al. Resistance to apoptosis underpins the corticosteroid insensitivity of group 2 innate lymphoid cells. J Allergy Clin Immunol. 2019;144(6):1722–6.

    Article  PubMed  Google Scholar 

  24. Law JP, Hirschkorn DF, Owen RE, et al. The importance of Foxp3 antibody and fixation/permeabilization buffer combinations in identifying CD4+CD25+Foxp3+ regulatory T cells. Cytometry A. 2009;75(12):1040–50.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wei J, Shi Q, Xiong L, et al. Transcriptome profiling of cells exposed to particular and intense electromagnetic radiation emitted by the “SG-III” prototype laser facility. Sci Rep. 2021;11(1):2017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li X, Wang S, Zhu R, et al. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J Hematol Oncol. 2016;9:42.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Klose CS, Kiss EA, Schwierzeck V, et al. A T-bet gradient controls the fate and function of CCR6-RORγt+ innate lymphoid cells. Nature. 2013;494(7436):261–5.

    Article  CAS  PubMed  Google Scholar 

  28. Crellin NK, Trifari S, Kaplan CD, et al. Regulation of cytokine secretion in human CD127(+) LTi-like innate lymphoid cells by Toll-like receptor 2. Immunity. 2010;33(5):752–64.

    Article  CAS  PubMed  Google Scholar 

  29. Loos T, Opdenakker G, Van Damme J, et al. Citrullination of CXCL8 increases this chemokine’s ability to mobilize neutrophils into the blood circulation. Haematologica. 2009;94(10):1346–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hammond ME, Lapointe GR, Feucht PH, et al. IL-8 induces neutrophil chemotaxis predominantly via type I IL-8 receptors. J Immunol. 1995;155(3):1428–33.

    Article  CAS  PubMed  Google Scholar 

  31. Teunissen M, Munneke JM, Bernink JH, et al. Composition of innate lymphoid cell subsets in the human skin: enrichment of NCR(+) ILC3 in lesional skin and blood of psoriasis patients. J Invest Dermatol. 2014;134(9):2351–60.

    Article  CAS  PubMed  Google Scholar 

  32. Ahn YO, Weeres MA, Neulen ML, et al. Human group3 innate lymphoid cells express DR3 and respond to TL1A with enhanced IL-22 production and IL-2-dependent proliferation. Eur J Immunol. 2015;45(8):2335–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sutton CE, Lalor SJ, Sweeney CM, et al. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity. 2009;31(2):331–41.

    Article  CAS  PubMed  Google Scholar 

  34. Chambers ES, Nanzer AM, Pfeffer PE, et al. Distinct endotypes of steroid-resistant asthma characterized by IL-17A(high) and IFN-γ(high) immunophenotypes: Potential benefits of calcitriol. J Allergy Clin Immunol. 2015;136(3):628–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Choi JS, Jang AS, Park JS, et al. Role of neutrophils in persistent airway obstruction due to refractory asthma. Respirology. 2012;17(2):322–9.

    Article  PubMed  Google Scholar 

  36. Shaw DE, Berry MA, Hargadon B, et al. Association between neutrophilic airway inflammation and airflow limitation in adults with asthma. Chest. 2007;132(6):1871–5.

    Article  CAS  PubMed  Google Scholar 

  37. Timmermans S, Souffriau J, Libert C. A general introduction to glucocorticoid biology. Front Immunol. 2019;10:1545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kino T. GR-regulating serine/threonine kinases: new physiologic and pathologic implications. Trends Endocrinol Metab. 2018;29(4):260–70.

    Article  CAS  PubMed  Google Scholar 

  39. Fasolino M, Vahedi G. GR extinguishes inflamed chromatin, and NF-κB evacuates. Immunity. 2017;47(2):214–6.

    Article  CAS  PubMed  Google Scholar 

  40. Everaere L, Ait-Yahia S, Molendi-Coste O, et al. Innate lymphoid cells contribute to allergic airway disease exacerbation by obesity. J Allergy Clin Immunol. 2016;138(5):1309–18.

    Article  CAS  PubMed  Google Scholar 

  41. Wu Y, Yue J, Wu J, et al. Obesity may provide Pro-ILC3 development inflammatory environment in asthmatic children. J Immunol Res. 2018;2018:1628620.

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Mao JT, Huang M, Wang J, et al. Cocaine down-regulates IL-2-induced peripheral blood lymphocyte IL-8 and IFN-gamma production. Cell Immunol. 1996;172(2):217–23.

    Article  CAS  PubMed  Google Scholar 

  45. Gusella GL, Musso T, Bosco MC, et al. IL-2 up-regulates but IFN-gamma suppresses IL-8 expression in human monocytes. J Immunol. 1993;151(5):2725–32.

    Article  CAS  PubMed  Google Scholar 

  46. Esteban-Gorgojo I, Antolín-Amérigo D, Domínguez-Ortega J, et al. Non-eosinophilic asthma: current perspectives. J Asthma Allergy. 2018;11:267–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Galliher-Beckley AJ, Cidlowski JA. Emerging roles of glucocorticoid receptor phosphorylation in modulating glucocorticoid hormone action in health and disease. IUBMB Life. 2009;61(10):979–86.

    Article  CAS  PubMed  Google Scholar 

  48. Miller AL, Garza AS, Johnson BH, et al. Pathway interactions between MAPKs, mTOR, PKA, and the glucocorticoid receptor in lymphoid cells. Cancer Cell Int. 2007;7:3.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Wu D, Gu B, Qian Y, et al. Long non-coding RNA growth arrest specific-5: a potential biomarker for early diagnosis of severe asthma. J Thorac Dis. 2020;12(5):1960–71.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kobayashi Y, Mercado N, Barnes PJ, et al. Defects of protein phosphatase 2A causes corticosteroid insensitivity in severe asthma. PLoS ONE. 2011;6(12): e27627.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Miller AL, Webb MS, Copik AJ, et al. p38 Mitogen-activated protein kinase (MAPK) is a key mediator in glucocorticoid-induced apoptosis of lymphoid cells: correlation between p38 MAPK activation and site-specific phosphorylation of the human glucocorticoid receptor at serine 211. Mol Endocrinol. 2005;19(6):1569–83.

    Article  CAS  PubMed  Google Scholar 

  52. Itoh M, Adachi M, Yasui H, et al. Nuclear export of glucocorticoid receptor is enhanced by c-Jun N-terminal kinase-mediated phosphorylation. Mol Endocrinol. 2002;16(10):2382–92.

    Article  CAS  PubMed  Google Scholar 

  53. Verhoog NJ, Du Toit A, Avenant C, et al. Glucocorticoid-independent repression of tumor necrosis factor (TNF) alpha-stimulated interleukin (IL)-6 expression by the glucocorticoid receptor: a potential mechanism for protection against an excessive inflammatory response. J Biol Chem. 2011;286(22):19297–310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Adcock IM, Caramori G. Cross-talk between pro-inflammatory transcription factors and glucocorticoids. Immunol Cell Biol. 2001;79(4):376–84.

    Article  CAS  PubMed  Google Scholar 

  55. Clarke DL, Davis NH, Majithiya JB, et al. Development of a mouse model mimicking key aspects of a viral asthma exacerbation. Clin Sci. 2014;126(8):567–80.

    Article  CAS  Google Scholar 

  56. Chachi L, Shikotra A, Duffy SM, et al. Functional KCa31 channels regulate steroid insensitivity in bronchial smooth muscle cells. J Immunol. 2013;191(5):2624–36.

    Article  CAS  PubMed  Google Scholar 

  57. Bouazza B, Krytska K, Debba-Pavard M, et al. Cytokines alter glucocorticoid receptor phosphorylation in airway cells: role of phosphatases. Am J Respir Cell Mol Biol. 2012;47(4):464–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ismaili N, Garabedian MJ. Modulation of glucocorticoid receptor function via phosphorylation. Ann N Y Acad Sci. 2004;1024:86–101.

    Article  CAS  PubMed  Google Scholar 

  59. De Grove KC, Provoost S, Verhamme FM, et al. Characterization and quantification of innate lymphoid cell subsets in human lung. PLoS ONE. 2016;11(1): e145961.

    Article  Google Scholar 

Download references

Acknowledgements

We wish to thank Huifang Li and Mengli Zhu (Core Facility of West China Hospital) for their help with the flow cytometry measurements.

Funding

This study was supported by the National Natural Science Foundation of China (No. 8770035).

Author information

Authors and Affiliations

Authors

Contributions

CTL, JL, LXH and LY contributed to the conception and design of the study. All authors recruited the patients and obtained the samples. CTL and JL supervised and advised the overall development of this study. LXH and LY performed experiments, analyzed data, and wrote the manuscript. CTL, JL, LXH and LY contributed to interpretation of results. All authors revised the manuscript critically for intellectual content and provided approval of the version to be published. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jian Luo or Chun Tao Liu.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Institutional Review Board (IRB) at West China Hospital, Sichuan University (Chengdu, China) (No. 2019–856). All participants provided written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1: Figure S1.

Flow cytometric gating strategy for ILCs in PBMCs. ILC2 populations were gated as LinCD45+CD3CD4CD8 CD127+CRTH2+, ILC1 populations were gated as LinCD45+CD3CD4CD8 CD127+CRTH2CD117, and ILC3 populations were gated as LinCD45+CD3CD4CD8CRTH2CD127+CD117+ Lineage markers contained CD11b, CD11c, CD14, CD19, CD56, CD123, and FcεRI. ILCs, innate lymphoid cells; ILC1, group 1 innate lymphoid cell; ILC2, group 2 innate lymphoid cell; ILC3, group 3 innate lymphoid cell.

Additional file 2: Figure S2.

A, Blood neutrophils count and percentage among healthy control (HC) and patients with NEA and EA. B, Correlation between ILC subsets and blood neutrophils count and percentage. **** p < 0.0001, *** p < 0.001, ** p < 0.01, * p < 0.05, ns p > 0.05.

Additional file 3: Figure S3.

A, Level of p-GR S226 and p-GR S211 in HBEs with or without dexamethasone treatment. B, Level of p-GR S226 and p-GR S211 in ILC3s with or without dexamethasone treatment.

Additional file 4: Table S1.

Antibodies used in flow cytometry and cell sorting. Table S2. Primer sequences used in RT-PCR. Table S3. Antibodies used for western blotting.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, L.X., Yang, L., Liu, T. et al. Group 3 innate lymphoid cells secret neutrophil chemoattractants and are insensitive to glucocorticoid via aberrant GR phosphorylation. Respir Res 24, 90 (2023). https://doi.org/10.1186/s12931-023-02395-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-023-02395-5

Keywords