Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 204(6): 1592-1597, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31988179

ABSTRACT

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.


Subject(s)
Asthma/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Respiratory Mucosa/immunology , Signal Transduction/immunology , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/blood , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Eosinophils/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/administration & dosage , Interleukin-33/genetics , Mice , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Signal Transduction/genetics
2.
J Immunol ; 200(12): 4036-4043, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29728508

ABSTRACT

IL-33 and its receptor ST2 play important roles in airway inflammation and contribute to asthma onset and exacerbation. The IL-33/ST2 signaling pathway recruits adapter protein myeloid differentiation primary response 88 (MyD88) to transduce intracellular signaling. MyD88 forms a complex with IL-R-associated kinases (IRAKs), IRAK4 and IRAK2, called the Myddosome (MyD88-IRAK4-IRAK2). The myddosome subsequently activates downstream NF-κB and MAPKs p38 and JNK. We established an asthma-like mouse model by intratracheal administration of IL-33. The IL-33 model has a very similar phenotype compared with the OVA-induced mouse asthma model. The importance of MyD88 in the IL-33/ST2 signaling transduction was demonstrated by the MyD88 knockout mice, which were protected from the IL-33-induced asthma. We synthesized small molecule mimetics of the α-helical domain of IRAK2 with drug-like characteristics based on the recent advances in the designing of α-helix compounds. The mimetics can competitively interfere in the protein-protein interaction between IRAK2 and IRAK4, leading to disruption of Myddosome formation. A series of small molecules were screened using an NF-κB promoter assay in vitro. The lead compound, 7004, was further studied in the IL-33-induced and OVA-induced asthma mouse models in vivo. Compound 7004 can inhibit the IL-33-induced NF-κB activity, disrupt Myddosome formation, and attenuate the proinflammatory effects in asthma-like models. Our data indicate that the Myddosome may represent a novel intracellular therapeutic target for diseases in which IL-33/ST2 plays important roles, such as asthma and other inflammatory diseases.


Subject(s)
Asthma/drug therapy , Inflammation/drug therapy , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-33/metabolism , Protein Conformation, alpha-Helical/drug effects , Small Molecule Libraries/pharmacology , Animals , Asthma/metabolism , Cells, Cultured , Disease Models, Animal , Inflammation/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phenotype , Protein Binding/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...