Oxidised IL-33 drives COPD epithelial pathogenesis via ST2-independent RAGE/EGFR signalling complex

Extract The airway epithelium plays a central role in the initiation and development of chronic airway diseases [1]. It is the first line of defence against the external environment; therefore, numerous inflammatory and tissue remodelling pathways exist to protect against damage caused by constant exposure to inhaled stimuli (e.g. allergens, cigarette smoke or pathogens) [1, 2]. Dysregulation of these pathways alters the structure of the airway epithelium, making it an inadequate defence against further insult from inhaled stimuli. This results in a cycle of abnormal epithelial responses and further damage, which may contribute to irreversible airway changes and severe chronic airway disease, such as chronic obstructive pulmonary disease (COPD) [1, 2]. Graphical abstract IL-33ox binds to receptor for advanced glycation end products (RAGE) to signal via epidermal growth factor receptor (EGFR). Activation of the IL-33ox–RAGE/EGFR pathway redirects epithelial cell fate, promoting a mucin hypersecretion phenotype at the expense of epithelial defence functions. Background Epithelial damage, repair and remodelling are critical features of chronic airway diseases including chronic obstructive pulmonary disease (COPD). Interleukin (IL)-33 released from damaged airway epithelia causes inflammation via its receptor, serum stimulation-2 (ST2). Oxidation of IL-33 to a non-ST2-binding form (IL-33ox) is thought to limit its activity. We investigated whether IL-33ox has functional activities that are independent of ST2 in the airway epithelium. Methods In vitro epithelial damage assays and three-dimensional, air–liquid interface (ALI) cell culture models of healthy and COPD epithelia were used to elucidate the functional role of IL-33ox. Transcriptomic changes occurring in healthy ALI cultures treated with IL-33ox and COPD ALI cultures treated with an IL-33-neutralising antibody were assessed with bulk and single-cell RNA sequencing analysis. Results We demonstrate that IL-33ox forms a complex with receptor for advanced glycation end products (RAGE) and epidermal growth factor receptor (EGFR) expressed on airway epithelium. Activation of this alternative, ST2-independent pathway impaired epithelial wound closure and induced airway epithelial remodelling in vitro. IL-33ox increased the proportion of mucus-producing cells and reduced epithelial defence functions, mimicking pathogenic traits of COPD. Neutralisation of the IL-33ox pathway reversed these deleterious traits in COPD epithelia. Gene signatures defining the pathogenic effects of IL-33ox were enriched in airway epithelia from patients with severe COPD. Conclusions Our study reveals for the first time that IL-33, RAGE and EGFR act together in an ST2-independent pathway in the airway epithelium and govern abnormal epithelial remodelling and muco-obstructive features in COPD. Tweetable abstract An ST2-independent signalling complex of oxidised IL-33, receptor for advanced glycation end products (RAGE) and epidermal growth factor receptor (EGFR) governs epithelial remodelling and mucin hypersecretion in COPD https://bit.ly/3pxgaqx

[1]  P. Cicuta,et al.  Single-Cell and Population Transcriptomics Reveal Pan-epithelial Remodeling in Type 2-High Asthma , 2020, Cell reports.

[2]  A. Korobeynikov,et al.  Sequence Analysis , 2020, Encyclopedia of Bioinformatics and Computational Biology.

[3]  D. Pe’er,et al.  A single-cell atlas of the human healthy airways , 2019, bioRxiv.

[4]  Irving L. Weissman,et al.  A molecular cell atlas of the human lung from single cell RNA sequencing , 2019, Nature.

[5]  Fabian J Theis,et al.  A cellular census of human lungs identifies novel cell states in health and in asthma , 2019, Nature Medicine.

[6]  C. Marquette,et al.  Novel dynamics of human mucociliary differentiation revealed by single-cell RNA sequencing of nasal epithelial cultures , 2019, Development.

[7]  John M. Gaspar,et al.  NGmerge: merging paired-end reads via novel empirically-derived models of sequencing errors , 2018, BMC Bioinformatics.

[8]  Paul J. Hoffman,et al.  Comprehensive Integration of Single-Cell Data , 2018, Cell.

[9]  Brent S. Pedersen,et al.  Bioconda: sustainable and comprehensive software distribution for the life sciences , 2018, Nature Methods.

[10]  Joseph G Ibrahim,et al.  Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences , 2018, bioRxiv.

[11]  Paolo Di Tommaso,et al.  Nextflow enables reproducible computational workflows , 2017, Nature Biotechnology.

[12]  J. Tedrow,et al.  Expression of RXFP1 Is Decreased in Idiopathic Pulmonary Fibrosis. Implications for Relaxin-based Therapies. , 2016, American journal of respiratory and critical care medicine.

[13]  Måns Magnusson,et al.  MultiQC: summarize analysis results for multiple tools and samples in a single report , 2016, Bioinform..

[14]  Jeffrey T. Chang,et al.  A dynamic bronchial airway gene expression signature of chronic obstructive pulmonary disease and lung function impairment. , 2013, American journal of respiratory and critical care medicine.

[15]  Justin Guinney,et al.  GSVA: gene set variation analysis for microarray and RNA-Seq data , 2013, BMC Bioinformatics.

[16]  A. Clark,et al.  Biologic Phenotyping of the Human Small Airway Epithelial Response to Cigarette Smoking , 2011, PloS one.

[17]  John D. Storey,et al.  Statistical significance for genomewide studies , 2003, Proceedings of the National Academy of Sciences of the United States of America.