IL-33 and its receptor ST2 are contributing factors to airway inflammation and asthma exacerbation. The IL-33/ST2 signaling pathway is involved in both the onset and the acute exacerbations of asthma. In this study, we address the role of endogenous IL-33 and its autoamplification of the IL-33/ST2 pathway in Ag-dependent and Ag-independent asthma-like models. Wild-type, IL-33 knockout, ST2 knockout mice were either intratracheally administrated with 500 ng of rIL-33 per day for four consecutive days or were sensitized and challenged with OVA over 21 d. In wild-type mice, IL-33 or OVA induced similar airway hyperresponsiveness and eosinophilic airway inflammation. IL-33 induced its own mRNA and ST2L mRNA expression in the lung. IL-33 autoamplified itself and ST2 protein expression in airway epithelial cells. OVA also induced IL-33 and ST2 protein expression. In IL-33 knockout mice, the IL-33– and OVA-induced airway hyperresponsiveness and eosinophilic airway inflammation were both significantly attenuated, whereas IL-33–induced ST2L mRNA expression was preserved, although no autoamplification of IL-33/ST2 pathway was observed. In ST2 knockout mice, IL-33 and OVA induced airway hyperresponsiveness and eosinophilic airway inflammation were both completely diminished, and no IL-33/ST2 autoamplification was observed. These results suggest that endogenous IL-33 and its autoamplification of IL-33/ST2 pathway play an important role in the induction of asthma-like phenotype. Thus an intact IL-33/ST2 pathway is necessary for both Ag-dependent and Ag-independent asthma-like mouse models.

Interleukin-33 was identified as the functional ligand for the IL-1R family member ST2 (1). ST2 has two forms. One is the transmembrane ST2 (ST2L), which transduces the IL-33 signaling. The other one is a soluble ST2 molecule (sST2), caused by alternative splicing (2). sST2 acts as a decoy receptor for IL-33 (3). Polymorphisms of human IL-33 and ST2 genes are associated with asthma and increased numbers of eosinophils (4, 5). IL-33 is a contributing factor to airway inflammation and asthma exacerbation (6, 7). The serum concentrations of sST2 are higher in asthma patients during acute exacerbation, suggesting that the host tries to attenuate IL-33 signaling (8, 9). An IL-33 loss-of-function mutation is associated with reduced blood eosinophil counts, and people with this mutation are protected from asthma (10).

IL-33 was originally characterized as a nuclear protein (11). It has a dual function of a potent cytokine and a nuclear transcription factor (12, 13). IL-33 binds to its receptor ST2 to augment the production of Th2 cytokines IL-4, IL-5, and IL-13 in vitro and in vivo (1, 14). It has been shown that epithelial derived IL-33 is a potent stimulator to induce IL-13–producing type 2 innate lymphoid cells (ILC2) (15, 16). ILC2 contribute to Ag-dependent and Ag-independent airway hyperactivity (15, 16). The role of IL-33 nuclear localization and chromatin association was not well established (17). IL-33 was found to sequester nuclear NF-κB and dampen proinflammatory signaling (13). We have previously shown that the intratracheal administration of IL-33 for 4 d induced an asthma-like phenotype mouse model with increased production of Th2 cytokines (18). The IL-33/ST2 signaling pathway may work in an autocrine and paracrine manner to exacerbate airway inflammation. In addition, recent data showed multiple feedback circuits involving IL-33 are required for asthma persistence (19).

In this study, we address the role of autoamplification of IL-33/ST2 pathway in Ag-dependent and Ag-independent asthma-like mouse models. In IL-33 knockout mice, we found that exogenous IL-33 could activate the IL-33/ST2 pathway but no IL-33 autoamplification happened. As a result, no robust airway hyperresponsiveness or eosinophilic inflammation were elicited. In ST2 knockout mice, no IL-33/ST2 autoamplification was observed and no airway hyperresponsiveness and eosinophilic inflammation were found. Endogenous IL-33 and its autoamplification of IL-33/ST2 pathway play an important role in the induction of asthma-like phenotype.

All studies were performed according to National Institutes of Health Guidelines for the Care and Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of the San Diego VA Healthcare System. Mouse strains studied included C57BL/6 from Jackson Laboratories (Bar Harbor, ME), IL-33 knockout from Dr. R. Lee (Brigham and Women’s Hospital, Harvard Medical School); and ST2 knockout from Dr. S. Akira (Research Institute for Microbial Disease, Osaka University, Japan). Asthma-like phenotype mouse models were previously described (18). In brief, for the IL-33 model, 6–8-wk-old mice were intratracheally administrated with 500 ng of rIL-33 per day for four consecutive days. For the OVA model, 6–8-wk-old mice were immunized and sensitized to OVA using a 21-d protocol. Mice were immunized by i.p. injection of 50 μg OVA and 1 mg alum in PBS on day 0 and 7, followed by four intratracheal injections of 20 μg OVA in 50 μl of PBS on days 17, 18, 19, and 20. On day 5 for the IL-33 model or on day 21 for the OVA model, the mice were intubated with a 20 gauge i.v. catheter and placed on a computer-controlled small animal ventilator (Flexivent; SCIREQ, Montreal, Canada) delivering 2% isoflurane continuously (20). Baseline airway resistance was measured. The animals were then challenged with an ultrasonic aerosol of 1.5, 3, 6, 12, and 24 mg/ml methacholine. The peak airway resistance with each dose was obtained.

Following the methacholine challenges, the mice were euthanized and bronchoalveolar lavage (BAL) fluid were obtained with 0.5 ml of PBS repeated for three times. The total cell counts and differentiation of the cells in BAL were studied. One lung was removed for RNA extraction and protein extraction. The other lung was fixed for histological analysis and immunohistochemistry (IHC). Total RNA was extracted from lung tissue by using TRIzol (Invitrogen, Carlsbad, CA). cDNA was synthesized and real-time quantitative PCR (qPCR) were performed with SYBR Green (Invitrogen). Expression is shown relative to the reference gene GAPDH. Primers are mouse IL-33 forward (F): 5′-ACTCCAAGATTTCGCCG-3′; mouse IL-33 reverse (R): 5′-CATGCAGTAGACATGGCAGAA-3′; mouse ST2L F: 5′-GTGATAGTCTTAAAAGTGTTCTGG-3′; mouse ST2L R: 5′-TCAAAAGTGTTTCAGGTCTAAGCA-3′; mouse sST2 F: 5′-ACGCTCGACTTATCCTGTGG-3′; mouse sST2 R: 5′-CAGCTCAATTGTTGGACACG-3′. ELISA for IL-33 and sST2 were performed on BAL samples with mouse IL-33 and ST2 ELISA kits (R&D Systems, Minneapolis, MN) according to the manufacturer’s instructions.

Formalin-fixed paraffin-embedded specimens were serially sectioned at 5-μm intervals and mounted on glass slides. IHC for IL-33 and ST2 were performed using the Avidin-Biotin Complex Kits (Vector Laboratories, Burlingame, CA). Rabbit anti-mouse ST2 Ab (Abcam, Cambridge, MA), mouse anti-mouse IL-33 Ab (Abcam), and rabbit and mouse IgG isotype controls (Vector Laboratories) were used. Lung inflammation score was performed based on the degree of hypertrophy of the airway epithelium and peribronchial/perivascular cellular inflammation on a scale of 0–5 by a lung pathologist blinded to the experimental groups. Peribronchial/perivascular infiltrates were as follows: 0 (not present); 1 (<20% of the airways affected); 2 (20–40%); 3 (40–60%); 4 (60–80%); and 5 (>80%).

Total lung proteins were prepared with RIPA buffer. Proteins were separated in 10% SDS-PAGE and transferred to nitrocellulose membrane. The membrane was blocked for 1 h in blocking buffer for fluorescent Western blot (Rockland, Gilbertsville, PA). Proteins were detected by Western blot by using anti-IL-33 Ab (Abcam). Fluorescent-labeled secondary Ab (Li-Cor Biosciences, Lincoln, NE) was used to detect the protein bands. An Odyssey imaging system imager was used to capture the images (Li-Cor Biosciences).

ANOVA was performed using GraphPad Prism software (La Jolla, CA). Statistical analyses of data were performed using paired and unpaired Student t tests or using one-way ANOVA. For qPCR results, log fold changes were used for t tests. Statistical significance is defined by p < 0.05.

We have previously shown that intratracheal administration of rIL-33 can induce airway hyperresponsiveness and eosinophilic airway inflammation in wild-type mice (18). In the IL-33 models, we found IL-33 induced its own mRNA and ST2L mRNA levels in the lung tissue (Fig. 1A). We also examined the BAL IL-33 and sST2 protein levels by ELISA. We found that the endogenous IL-33 levels were increased and the sST2 levels were nearly abolished in BAL (Fig. 1B, 1C). In the OVA models, although the IL-33 and ST2 mRNA levels were not significantly changed in the lung tissue (Fig. 1A), the BAL IL-33 levels were increased and sST2 levels were decreased with similar trends of lesser magnitude compared with the IL-33 models (Fig. 1B, 1C).

FIGURE 1.

IL-33 autoamplifies the IL-33/ST2 pathway by inducing its own and ST2 expression. (A) rIL-33 induces endogenous IL-33 and ST2L mRNA in the lung tissue. Wild-type mice were treated with saline, IL-33, or OVA. Real-time qPCR was performed by using IL-33, ST2L, and sST2 specific primers. In the IL-33 model, a 4-fold increase of IL-33 was observed compared with saline controls (n = 6). A 5.8-fold increase of ST2L was observed compared with saline controls (n = 6). Student t test was used to calculate the significance of differences in log fold-changes of the qPCR results, which are normally distributed. (B) IL-33 induces IL-33 in BAL, compared with saline controls (n = 6). OVA has moderate increase of IL-33, compared with saline controls (n = 6). (C) IL-33 and OVA decreases sST2 in the BAL, compared with saline controls (n = 6). *p < 0.01 compared with saline controls.

FIGURE 1.

IL-33 autoamplifies the IL-33/ST2 pathway by inducing its own and ST2 expression. (A) rIL-33 induces endogenous IL-33 and ST2L mRNA in the lung tissue. Wild-type mice were treated with saline, IL-33, or OVA. Real-time qPCR was performed by using IL-33, ST2L, and sST2 specific primers. In the IL-33 model, a 4-fold increase of IL-33 was observed compared with saline controls (n = 6). A 5.8-fold increase of ST2L was observed compared with saline controls (n = 6). Student t test was used to calculate the significance of differences in log fold-changes of the qPCR results, which are normally distributed. (B) IL-33 induces IL-33 in BAL, compared with saline controls (n = 6). OVA has moderate increase of IL-33, compared with saline controls (n = 6). (C) IL-33 and OVA decreases sST2 in the BAL, compared with saline controls (n = 6). *p < 0.01 compared with saline controls.

Close modal

To investigate the changes in the IL-33/ST2 pathway at the cellular level, we performed IHC studies (Fig. 2A). In the saline controls (Fig. 2A, left panel), IL-33 was found in the airway epithelial cells, whereas ST2 was minimally detected in the airway epithelium. In the IL-33 models (Fig. 2A, middle panel), IL-33 was found in the airway and alveolar epithelial cells; interstitial cells, including lymphocytes; macrophages; and the nucleus of type II epithelial cells. ST2 was found in epithelial cells and interstitial cells as well. In the OVA models (Fig. 2A, right panel), in addition to the airway and alveolar epithelial cells and interstitial cells, IL-33 was also found in smooth muscle cells and the cytoplasm of alveolar macrophages. ST2 was found in the epithelium and interstitial cells. Lung inflammation scores were performed by a lung pathologist blinded to the experimental groups (Fig. 2B). Both IL-33 and OVA groups showed significant increased lung inflammation scores compare with saline control group. Examples of lung tissue IL-33 protein levels are shown by Western blots (Fig. 2C). rIL-33 (exogenous) and endogenous IL-33 were distinguishable in sizes. Autoamplified endogenous IL-33 protein levels in IL-33 models were confirmed.

FIGURE 2.

Airway epithelial cells are the major source of endogenous IL-33 production in autoamplification of IL-33/ST2 pathway. (A) Representative images of IHC lung specimens were shown (n = 6). Scale bar, 100 μm at lower or 50 μm at higher magnifications. Higher magnification images are shown for regions indicated by dashed boxes. Left panel, Wild-type mice with saline control. IL-33 was mostly found in the airway epithelial cells (top two rows), much less was found in the alveolar epithelial cells. ST2 was minimally detected in the airway epithelium (bottom two rows). Middle panel, Wild-type mice with IL-33 models, IL-33 was found in the airway and alveolar epithelial cells, interstitial cells, including lymphocytes, macrophages, and the nucleus of Type II epithelial cells. ST2 was found in epithelial cells and interstitial cells as well. Right panel, Wild-type mice with OVA models, in addition to the airway and alveolar epithelial cells and interstitial cells, IL-33 was also found in smooth muscle cells and the cytoplasm of alveolar macrophages. ST2 was found in the epithelium and interstitial cells. (B) Lung inflammation score was performed based on the degree of hypertrophy of the airway epithelium and peribronchial/perivascular cellular inflammation. Both IL-33 and OVA models showed significant increased lung inflammation scores compare with saline control group. *p < 0.01 compared with saline controls. (C) Examples of lung tissue IL-33 protein levels are shown by Western blots. Lung tissue proteins from the indicated mice were subject to SDS-PAGE, the resulting membrane was blotted with anti-IL-33 (green) and anti-β-actin (red), and were analyzed with an Odyssey imaging system imager. Multiple Western blots were done and representative mages are shown. From left: lane 1, rIL-33 protein (exogenous, molecular mass 18.1 kDa). Lane 2, wild-type mice with saline control. Lane 3–6, wild-type mice with IL-33 models. The endogenous IL-33 (lane 2–6) has a molecular mass of 35 kDa. In IL-33 group, the endogenous IL-33 is upregulated compared with the saline control group. In all the mice groups, no detectable rIL-33 was found.

FIGURE 2.

Airway epithelial cells are the major source of endogenous IL-33 production in autoamplification of IL-33/ST2 pathway. (A) Representative images of IHC lung specimens were shown (n = 6). Scale bar, 100 μm at lower or 50 μm at higher magnifications. Higher magnification images are shown for regions indicated by dashed boxes. Left panel, Wild-type mice with saline control. IL-33 was mostly found in the airway epithelial cells (top two rows), much less was found in the alveolar epithelial cells. ST2 was minimally detected in the airway epithelium (bottom two rows). Middle panel, Wild-type mice with IL-33 models, IL-33 was found in the airway and alveolar epithelial cells, interstitial cells, including lymphocytes, macrophages, and the nucleus of Type II epithelial cells. ST2 was found in epithelial cells and interstitial cells as well. Right panel, Wild-type mice with OVA models, in addition to the airway and alveolar epithelial cells and interstitial cells, IL-33 was also found in smooth muscle cells and the cytoplasm of alveolar macrophages. ST2 was found in the epithelium and interstitial cells. (B) Lung inflammation score was performed based on the degree of hypertrophy of the airway epithelium and peribronchial/perivascular cellular inflammation. Both IL-33 and OVA models showed significant increased lung inflammation scores compare with saline control group. *p < 0.01 compared with saline controls. (C) Examples of lung tissue IL-33 protein levels are shown by Western blots. Lung tissue proteins from the indicated mice were subject to SDS-PAGE, the resulting membrane was blotted with anti-IL-33 (green) and anti-β-actin (red), and were analyzed with an Odyssey imaging system imager. Multiple Western blots were done and representative mages are shown. From left: lane 1, rIL-33 protein (exogenous, molecular mass 18.1 kDa). Lane 2, wild-type mice with saline control. Lane 3–6, wild-type mice with IL-33 models. The endogenous IL-33 (lane 2–6) has a molecular mass of 35 kDa. In IL-33 group, the endogenous IL-33 is upregulated compared with the saline control group. In all the mice groups, no detectable rIL-33 was found.

Close modal

To understand if endogenous IL-33 is necessary for Ag-dependent and Ag-independent asthma-like phenotype, we used the IL-33 knockout mice. In IL-33 knockout mice, rIL-33 induced airway hyperresponsiveness and eosinophilic airway inflammation were significantly attenuated compared with wild-type mice (Fig. 3A, 3B). Exogenous rIL-33 was not sufficient to restore the asthma-like phenotype in IL-33 knockout mice, despite the intact ST2 and other components of downstream IL-33/ST2 pathway. In IL-33 knockout mice, OVA induced airway hyperresponsiveness and eosinophilic airway inflammation were significantly attenuated as well (Fig. 3A, 3B).

FIGURE 3.

Endogenous IL-33 is necessary for Ag-dependent and Ag-independent asthma-like phenotype in vivo. (A) IL-33 knockout mice were treated with saline, IL-33 or OVA. IL-33 induced airway hyperresponsiveness was significantly attenuated (n = 6). OVA induced airway hyperresponsiveness was significantly attenuated (n = 6). *p < 0.01 compared with wild-type mice IL-33 model. (B) In IL-33 knockout mice, IL-33 induced eosinophilic infiltration was significantly attenuated (n = 6). OVA induced eosinophilic infiltration was significantly attenuated (n = 6). *p < 0.01 compared with wild-type mice IL-33 model.

FIGURE 3.

Endogenous IL-33 is necessary for Ag-dependent and Ag-independent asthma-like phenotype in vivo. (A) IL-33 knockout mice were treated with saline, IL-33 or OVA. IL-33 induced airway hyperresponsiveness was significantly attenuated (n = 6). OVA induced airway hyperresponsiveness was significantly attenuated (n = 6). *p < 0.01 compared with wild-type mice IL-33 model. (B) In IL-33 knockout mice, IL-33 induced eosinophilic infiltration was significantly attenuated (n = 6). OVA induced eosinophilic infiltration was significantly attenuated (n = 6). *p < 0.01 compared with wild-type mice IL-33 model.

Close modal

To further understand why exogenous IL-33 was not sufficient to restore the asthma-like phenotype in IL-33 knockout mice, we examined the ST2L and sST2 mRNA in lung tissue and IL-33 and sST2 protein levels in BAL. In IL-33 models, we found the exogenous IL-33–induced ST2L mRNA expression was preserved in lung tissue (Fig. 4A), whereas the effects of IL-33 on increasing IL-33 protein levels and decreasing sST2 in the BAL were diminished (Fig. 4B, 4C). In OVA models, OVA-induced ST2L mRNA expression was attenuated (Fig. 4A). The effects of OVA on increasing IL-33 protein levels and decreasing sST2 protein levels in the BAL were NS (Fig. 4B, 4C).

FIGURE 4.

Exogenous IL-33 can induce ST2L mRNA expression but is not sufficient to induce the endogenous autoamplification of IL-33/ST2 pathway. (A) In IL-33 knockout mice, IL-33–induced ST2L mRNA expression was preserved in the lung (n = 6), comparable with wild-type mice saline control. *p < 0.01 compared with wild-type mice saline controls. OVA-induced ST2L mRNA expression was attenuated (n = 6), NS compared with wild-type mice saline controls. IL-33 decreased sST2 mRNA expression in the lung (n = 6), comparable with wild-type mice saline control. **p < 0.05 compared with wild-type mice saline controls. (B) In IL-33 knockout mice, the effects of IL-33 on increasing IL-33 protein levels in the BAL were diminished (n = 6), compared with wild-type mice IL-33 model. **p < 0.01 compared with wild-type mice IL-33 model. The effects of OVA on increasing IL-33 protein levels in the BAL were NS compared with saline controls (n = 6). (C) The effects of IL-33 on decreasing sST2 protein levels in the BAL were diminished (n = 6), compared with wild-type mice IL-33 model. **p < 0.01 compared with wild-type mice IL-33 model. The effects of OVA on decreasing sST2 protein levels in the BAL were NS, compared with saline controls (n = 6).

FIGURE 4.

Exogenous IL-33 can induce ST2L mRNA expression but is not sufficient to induce the endogenous autoamplification of IL-33/ST2 pathway. (A) In IL-33 knockout mice, IL-33–induced ST2L mRNA expression was preserved in the lung (n = 6), comparable with wild-type mice saline control. *p < 0.01 compared with wild-type mice saline controls. OVA-induced ST2L mRNA expression was attenuated (n = 6), NS compared with wild-type mice saline controls. IL-33 decreased sST2 mRNA expression in the lung (n = 6), comparable with wild-type mice saline control. **p < 0.05 compared with wild-type mice saline controls. (B) In IL-33 knockout mice, the effects of IL-33 on increasing IL-33 protein levels in the BAL were diminished (n = 6), compared with wild-type mice IL-33 model. **p < 0.01 compared with wild-type mice IL-33 model. The effects of OVA on increasing IL-33 protein levels in the BAL were NS compared with saline controls (n = 6). (C) The effects of IL-33 on decreasing sST2 protein levels in the BAL were diminished (n = 6), compared with wild-type mice IL-33 model. **p < 0.01 compared with wild-type mice IL-33 model. The effects of OVA on decreasing sST2 protein levels in the BAL were NS, compared with saline controls (n = 6).

Close modal

ST2 is currently the only known receptor for IL-33 signaling. In ST2 knockout mice, both IL-33– and OVA-induced airway hyperresponsiveness or eosinophilic airway inflammation were completely diminished. There were no significant differences in airway hyperresponsiveness and total cell count and cell differentials compared with saline controls (Fig. 5A, 5B). In ST2 knockout mice, IL-33–induced autoamplification of IL-33 was completely diminished (Fig. 5C). OVA–induced IL-33 was also completely diminished (Fig. 5C).

FIGURE 5.

ST2 is necessary for Ag-dependent and Ag-independent asthma-like phenotype in vivo. (A) ST2 knockout mice were treated with saline, IL-33, or OVA. IL-33–induced airway hyperresponsiveness was completely diminished (n = 6). OVA induced airway hyperresponsiveness was completely diminished (n = 6). *p < 0.01 compared with wild-type mice IL-33 model. (B) In ST2 knockout mice, IL-33 induced eosinophilic infiltration was diminished (n = 6). OVA induced eosinophilic infiltration was diminished (n = 6). *p < 0.01 compared with wild-type mice IL-33 model. (C) In ST2 knockout mice, rIL-33 induces endogenous IL-33 mRNA in the lung tissue was completely diminished. OVA induced IL-33 mRNA in the lung tissue was also completely diminished. *p < 0.01 compared with wild-type mice saline controls.

FIGURE 5.

ST2 is necessary for Ag-dependent and Ag-independent asthma-like phenotype in vivo. (A) ST2 knockout mice were treated with saline, IL-33, or OVA. IL-33–induced airway hyperresponsiveness was completely diminished (n = 6). OVA induced airway hyperresponsiveness was completely diminished (n = 6). *p < 0.01 compared with wild-type mice IL-33 model. (B) In ST2 knockout mice, IL-33 induced eosinophilic infiltration was diminished (n = 6). OVA induced eosinophilic infiltration was diminished (n = 6). *p < 0.01 compared with wild-type mice IL-33 model. (C) In ST2 knockout mice, rIL-33 induces endogenous IL-33 mRNA in the lung tissue was completely diminished. OVA induced IL-33 mRNA in the lung tissue was also completely diminished. *p < 0.01 compared with wild-type mice saline controls.

Close modal

IL-33 is expressed in higher levels in endobronchial biopsies from human asthma patients (7). The elevated IL-33 expression was particularly evident in severe asthmatic subjects (6, 7). IL-33 functions both extracellularly as a cytokine through activation of the ST2L receptor and intracellular as a nuclear transcription factor (12, 13). IL-33 can increase ST2 expression in eosinophils (21). In animal models, multiple feedback circuits involving IL-33 are required for asthma persistence (19). In our IL-33 models, we found parallel increases of IL-33 and ST2L mRNA levels in the lung. In both IL-33 and OVA models, we found increased IL-33 and decreased sST2 in BAL. We found that epithelial cells are the major source of endogenous IL-33 and ST2 production. These data suggest the autoamplification of IL-33/ST2 pathway. OVA-induced IL-33 and ST2 expression may have a different mechanism. We found enlarged mediastinal lymph nodes in the OVA models, and ST2 was significantly increased in the lymph nodes (data not shown). Our data are consistent with the findings that dust mites and other allergens can induce IL-33 (22, 23) and elevate serum sST2 levels (24). Autoamplification of IL-33/ST2 pathway is necessary for the induction of asthma-like phenotype in both IL-33 models and OVA models.

We used IHC to examine the major source of IL-33 in lung tissue. IL-33 was localized to the nuclei of airway and alveolar epithelial cells. IL-33 induced IL-33 and ST2 expression localizing at the same epithelial cells and interstitial cells. It was reported that IL-33 accumulated in the nucleus of producing cells and binds to histones and chromatin (25, 26). The function of IL-33 as an NF was not well established. A recent study showed the only identified protein modulated by IL-33 as an NF was IL-33 itself (27). Our data provided the evidence that IL-33 autoamplified itself and ST2 protein expression in airway epithelial cells. It was reported that multiple feedback and feed-forward circuits between epithelial cells and ILC2 were identified (19). IL-33 may regulate IL-33/ST2 pathway in several manners, such as autocrine and paracrine regulation as a cytokine, and transcriptional regulation or epigenetic regulation at the chromatin level as an NF.

In IL-33 knockout mice, we showed that exogenous rIL-33 induced a modest airway hyperresponsiveness and mild eosinophilic airway inflammation. Exogenous IL-33–induced ST2L mRNA expression was preserved in lung tissue. However, because of the lack of endogenous IL-33 and its autoamplification of IL-33/ST2 pathway, BAL IL-33 levels and sST2 levels were not changed, leading to only moderate asthma phenotype. These data indicate that exogenous IL-33 was able to induce ST2L mRNA expression based on the intact ST2 and downstream pathway. The IL-33/ST2 signaling was successfully initiated but failed to maintain because of lack of endogenous IL-33 and its autoamplification. We conclude that multiple feedback loops of continuous autoamplification of IL-33/ST2 pathway are necessary for maintaining the asthma-like phenotype in vivo. It was previously reported that IL-33 knockout mice showed attenuated eosinophil and lymphocyte recruitment to the lung in OVA model (28, 29). We observed similar results. OVA can elicit a moderate response because of the intact adaptive immune response in IL-33 knockout mice. In both IL-33 and OVA models, the responses are reduced because of lack of endogenous IL-33. These data indicate that the endogenous IL-33 and its autoamplification of IL-33/ST2 pathway are necessary for Ag-dependent and Ag-independent asthma-like phenotype in vivo.

We showed that ST2 expression in the airway epithelium and inflammatory cell infiltrate were increased in both IL-33– and OVA-induced asthma models. In ST2 knockout mice, both IL-33– and OVA-induced airway hyperresponsiveness or eosinophilic airway inflammation were completely diminished. ST2 knockout mice are partially protected from house dust mite induced asthma, especially in the peripheral lung (23). An intact IL-33/ST2 pathway is necessary for both Ag-dependent and Ag-independent asthma-like mouse models.

In summary, endogenous IL-33 and its autoamplification of IL-33/ST2 pathway play an important role in both Ag-dependent and Ag-independent asthma-like mouse models. Airway epithelial cells are the major source of endogenous IL-33 production in autoamplification of IL-33/ST2 pathway. IL-33 and sST2 are possible biomarkers for asthma exacerbation (24, 30) and also could be therapeutic targets to inhibit the IL-33/ST2 autoamplification. Novel treatment including IL-33 Abs, sST2, and vaccination against IL-33 are under investigation in asthma (3, 3133). Our recent study of small molecule inhibitors of the IL-33/ST2 pathway showed beneficial results (18). Clinical trials with Abs targeting IL-33 have already been initiated (34).

This work was supported by National Institutes of Health Grant T32-HL098062 (to J.L., J.P.D.) and an American Thoracic Society Foundation grant (to J.L.).

Abbreviations used in this article:

BAL

bronchoalveolar lavage

F

forward

IHC

immunohistochemistry

ILC2

type 2 innate lymphoid cell

qPCR

quantitative PCR

R

reverse

sST2

soluble ST2 molecule.

1
Schmitz
,
J.
,
A.
Owyang
,
E.
Oldham
,
Y.
Song
,
E.
Murphy
,
T. K.
McClanahan
,
G.
Zurawski
,
M.
Moshrefi
,
J.
Qin
,
X.
Li
, et al
.
2005
.
IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines.
Immunity
23
:
479
490
.
2
Tominaga
,
S.
,
K.
Kuroiwa
,
K.
Tago
,
H.
Iwahana
,
K.
Yanagisawa
,
N.
Komatsu
.
1999
.
Presence and expression of a novel variant form of ST2 gene product in human leukemic cell line UT-7/GM.
Biochem. Biophys. Res. Commun.
264
:
14
18
.
3
Hayakawa
,
H.
,
M.
Hayakawa
,
A.
Kume
,
S.
Tominaga
.
2007
.
Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation.
J. Biol. Chem.
282
:
26369
26380
.
4
GABRIEL Consortium
.
2010
.
A large-scale, consortium-based genomewide association study of asthma.
N. Engl. J. Med.
363
:
1211
1221
.
5
Gudbjartsson
,
D. F.
,
U. S.
Bjornsdottir
,
E.
Halapi
,
A.
Helgadottir
,
P.
Sulem
,
G. M.
Jonsdottir
,
G.
Thorleifsson
,
H.
Helgadottir
,
V.
Steinthorsdottir
,
H.
Stefansson
, et al
.
2009
.
Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction.
Nat. Genet.
41
:
342
347
.
6
Kurowska-Stolarska
,
M.
,
B.
Stolarski
,
P.
Kewin
,
G.
Murphy
,
C. J.
Corrigan
,
S.
Ying
,
N.
Pitman
,
A.
Mirchandani
,
B.
Rana
,
N.
van Rooijen
, et al
.
2009
.
IL-33 amplifies the polarization of alternatively activated macrophages that contribute to airway inflammation.
J. Immunol.
183
:
6469
6477
.
7
Préfontaine
,
D.
,
S.
Lajoie-Kadoch
,
S.
Foley
,
S.
Audusseau
,
R.
Olivenstein
,
A. J.
Halayko
,
C.
Lemière
,
J. G.
Martin
,
Q.
Hamid
.
2009
.
Increased expression of IL-33 in severe asthma: evidence of expression by airway smooth muscle cells.
J. Immunol.
183
:
5094
5103
.
8
Oshikawa
,
K.
,
K.
Kuroiwa
,
K.
Tago
,
H.
Iwahana
,
K.
Yanagisawa
,
S.
Ohno
,
S. I.
Tominaga
,
Y.
Sugiyama
.
2001
.
Elevated soluble ST2 protein levels in sera of patients with asthma with an acute exacerbation.
Am. J. Respir. Crit. Care Med.
164
:
277
281
.
9
Watanabe
,
M.
,
K.
Nakamoto
,
T.
Inui
,
M.
Sada
,
K.
Honda
,
M.
Tamura
,
Y.
Ogawa
,
T.
Yokoyama
,
T.
Saraya
,
D.
Kurai
, et al
.
2018
.
Serum sST2 levels predict severe exacerbation of asthma.
Respir. Res.
19
:
169
.
10
Smith
,
D.
,
H.
Helgason
,
P.
Sulem
,
U. S.
Bjornsdottir
,
A. C.
Lim
,
G.
Sveinbjornsson
,
H.
Hasegawa
,
M.
Brown
,
R. R.
Ketchem
,
M.
Gavala
, et al
.
2017
.
A rare IL33 loss-of-function mutation reduces blood eosinophil counts and protects from asthma.
PLoS Genet.
13
: e1006659.
11
Baekkevold
,
E. S.
,
M.
Roussigné
,
T.
Yamanaka
,
F. E.
Johansen
,
F. L.
Jahnsen
,
F.
Amalric
,
P.
Brandtzaeg
,
M.
Erard
,
G.
Haraldsen
,
J. P.
Girard
.
2003
.
Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules.
Am. J. Pathol.
163
:
69
79
.
12
Haraldsen
,
G.
,
J.
Balogh
,
J.
Pollheimer
,
J.
Sponheim
,
A. M.
Küchler
.
2009
.
Interleukin-33 - cytokine of dual function or novel alarmin?
Trends Immunol.
30
:
227
233
.
13
Ali
,
S.
,
A.
Mohs
,
M.
Thomas
,
J.
Klare
,
R.
Ross
,
M. L.
Schmitz
,
M. U.
Martin
.
2011
.
The dual function cytokine IL-33 interacts with the transcription factor NF-κB to dampen NF-κB-stimulated gene transcription.
J. Immunol.
187
:
1609
1616
.
14
Kurowska-Stolarska
,
M.
,
P.
Kewin
,
G.
Murphy
,
R. C.
Russo
,
B.
Stolarski
,
C. C.
Garcia
,
M.
Komai-Koma
,
N.
Pitman
,
Y.
Li
,
W.
Niedbala
, et al
.
2008
.
IL-33 induces antigen-specific IL-5+ T cells and promotes allergic-induced airway inflammation independent of IL-4. [Published erratum appears in 2008 J. Immunol. 181: 8170.]
J. Immunol.
181
:
4780
4790
.
15
Barlow
,
J. L.
,
S.
Peel
,
J.
Fox
,
V.
Panova
,
C. S.
Hardman
,
A.
Camelo
,
C.
Bucks
,
X.
Wu
,
C. M.
Kane
,
D. R.
Neill
, et al
.
2013
.
IL-33 is more potent than IL-25 in provoking IL-13-producing nuocytes (type 2 innate lymphoid cells) and airway contraction.
J. Allergy Clin. Immunol.
132
:
933
941
.
16
Kim
,
H. Y.
,
Y. J.
Chang
,
S.
Subramanian
,
H. H.
Lee
,
L. A.
Albacker
,
P.
Matangkasombut
,
P. B.
Savage
,
A. N.
McKenzie
,
D. E.
Smith
,
J. B.
Rottman
, et al
.
2012
.
Innate lymphoid cells responding to IL-33 mediate airway hyperreactivity independently of adaptive immunity.
J. Allergy Clin. Immunol.
129
:
216
227.e1–6
.
17
Bessa
,
J.
,
C. A.
Meyer
,
M. C.
de Vera Mudry
,
S.
Schlicht
,
S. H.
Smith
,
A.
Iglesias
,
J.
Cote-Sierra
.
2014
.
Altered subcellular localization of IL-33 leads to non-resolving lethal inflammation.
J. Autoimmun.
55
:
33
41
.
18
Li
,
J.
,
K.
Saruta
,
J. P.
Dumouchel
,
J. M.
Magat
,
J. L.
Thomas
,
D.
Ajami
,
M.
Rebek
,
J.
Rebek
Jr.
,
T. D.
Bigby
.
2018
.
Small molecule mimetics of α-helical domain of IRAK2 attenuate the proinflammatory effects of IL-33 in asthma-like mouse models.
J. Immunol.
200
:
4036
4043
.
19
Christianson
,
C. A.
,
N. P.
Goplen
,
I.
Zafar
,
C.
Irvin
,
J. T.
Good
Jr.
,
D. R.
Rollins
,
B.
Gorentla
,
W.
Liu
,
M. M.
Gorska
,
H.
Chu
, et al
.
2015
.
Persistence of asthma requires multiple feedback circuits involving type 2 innate lymphoid cells and IL-33.
J. Allergy Clin. Immunol.
136
:
59
68.e14
.
20
Thomas
,
J. L.
,
J.
Dumouchel
,
J.
Li
,
J.
Magat
,
D.
Balitzer
,
T. D.
Bigby
.
2014
.
Endotracheal intubation in mice via direct laryngoscopy using an otoscope.
J. Vis. Exp.
86
:
e50269
21
Stolarski
,
B.
,
M.
Kurowska-Stolarska
,
P.
Kewin
,
D.
Xu
,
F. Y.
Liew
.
2010
.
IL-33 exacerbates eosinophil-mediated airway inflammation.
J. Immunol.
185
:
3472
3480
.
22
Castanhinha
,
S.
,
R.
Sherburn
,
S.
Walker
,
A.
Gupta
,
C. J.
Bossley
,
J.
Buckley
,
N.
Ullmann
,
R.
Grychtol
,
G.
Campbell
,
M.
Maglione
, et al
.
2015
.
Pediatric severe asthma with fungal sensitization is mediated by steroid-resistant IL-33.
J. Allergy Clin. Immunol.
136
:
312
322.e7
.
23
Zoltowska
,
A. M.
,
Y.
Lei
,
B.
Fuchs
,
C.
Rask
,
M.
Adner
,
G. P.
Nilsson
.
2016
.
The interleukin-33 receptor ST2 is important for the development of peripheral airway hyperresponsiveness and inflammation in a house dust mite mouse model of asthma.
Clin. Exp. Allergy
46
:
479
490
.
24
Mitchell
,
P. D.
,
B. M.
Salter
,
J. P.
Oliveria
,
A.
El-Gammal
,
D.
Tworek
,
S. G.
Smith
,
R.
Sehmi
,
G. M.
Gauvreau
,
P. M.
O Apos Byrne
.
2018
.
IL-33 and its receptor ST2 after inhaled allergen challenge in allergic asthmatics.
Int. Arch. Allergy Immunol.
176
:
133
142
.
25
Carriere
,
V.
,
L.
Roussel
,
N.
Ortega
,
D. A.
Lacorre
,
L.
Americh
,
L.
Aguilar
,
G.
Bouche
,
J. P.
Girard
.
2007
.
IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo.
Proc. Natl. Acad. Sci. USA
104
:
282
287
.
26
Roussel
,
L.
,
M.
Erard
,
C.
Cayrol
,
J. P.
Girard
.
2008
.
Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket.
EMBO Rep.
9
:
1006
1012
.
27
Gautier
,
V.
,
C.
Cayrol
,
D.
Farache
,
S.
Roga
,
B.
Monsarrat
,
O.
Burlet-Schiltz
,
A.
Gonzalez de Peredo
,
J. P.
Girard
.
2016
.
Extracellular IL-33 cytokine, but not endogenous nuclear IL-33, regulates protein expression in endothelial cells.
Sci. Rep.
6
:
34255
.
28
Oboki
,
K.
,
T.
Ohno
,
N.
Kajiwara
,
K.
Arae
,
H.
Morita
,
A.
Ishii
,
A.
Nambu
,
T.
Abe
,
H.
Kiyonari
,
K.
Matsumoto
, et al
.
2010
.
IL-33 is a crucial amplifier of innate rather than acquired immunity.
Proc. Natl. Acad. Sci. USA
107
:
18581
18586
.
29
Louten
,
J.
,
A. L.
Rankin
,
Y.
Li
,
E. E.
Murphy
,
M.
Beaumont
,
C.
Moon
,
P.
Bourne
,
T. K.
McClanahan
,
S.
Pflanz
,
R.
de Waal Malefyt
.
2011
.
Endogenous IL-33 enhances Th2 cytokine production and T-cell responses during allergic airway inflammation.
Int. Immunol.
23
:
307
315
.
30
Werder
,
R. B.
,
V.
Zhang
,
J. P.
Lynch
,
N.
Snape
,
J. W.
Upham
,
K.
Spann
,
S.
Phipps
.
2018
.
Chronic IL-33 expression predisposes to virus-induced asthma exacerbations by increasing type 2 inflammation and dampening antiviral immunity.
J. Allergy Clin. Immunol.
141
:
1607
1619.e9
.
31
Lei
,
Y.
,
V.
Boinapally
,
A.
Zoltowska
,
M.
Adner
,
L.
Hellman
,
G.
Nilsson
.
2015
.
Vaccination against IL-33 inhibits airway hyperresponsiveness and inflammation in a house dust mite model of asthma.
PLoS One
10
: e0133774.
32
Lee
,
H. Y.
,
C. K.
Rhee
,
J. Y.
Kang
,
J. H.
Byun
,
J. Y.
Choi
,
S. J.
Kim
,
Y. K.
Kim
,
S. S.
Kwon
,
S. Y.
Lee
.
2014
.
Blockade of IL-33/ST2 ameliorates airway inflammation in a murine model of allergic asthma.
Exp. Lung Res.
40
:
66
76
.
33
Zoltowska Nilsson
,
A. M.
,
Y.
Lei
,
M.
Adner
,
G. P.
Nilsson
.
2018
.
Mast cell-dependent IL-33/ST2 signaling is protective against the development of airway hyperresponsiveness in a house dust mite mouse model of asthma.
Am. J. Physiol. Lung Cell. Mol. Physiol.
314
:
L484
L492
.
34
Mitchell
,
P. D.
,
A. I.
El-Gammal
,
P. M.
O’Byrne
.
2016
.
Emerging monoclonal antibodies as targeted innovative therapeutic approaches to asthma.
Clin. Pharmacol. Ther.
99
:
38
48
.

The authors have no financial conflicts of interest.