Persistent activation of interlinked type 2 airway epithelial gene networks in sputum-derived cells from aeroallergen-sensitized symptomatic asthmatics

Anya C Jones, Niamh M Troy, Elisha White, Elysia M Hollams, Alexander M Gout, Kak-Ming Ling, Anthony Kicic, Stephen M Stick, Peter D Sly, Patrick G Holt, Graham L Hall, Anthony Bosco, Anya C Jones, Niamh M Troy, Elisha White, Elysia M Hollams, Alexander M Gout, Kak-Ming Ling, Anthony Kicic, Stephen M Stick, Peter D Sly, Patrick G Holt, Graham L Hall, Anthony Bosco

Abstract

Atopic asthma is a persistent disease characterized by intermittent wheeze and progressive loss of lung function. The disease is thought to be driven primarily by chronic aeroallergen-induced type 2-associated inflammation. However, the vast majority of atopics do not develop asthma despite ongoing aeroallergen exposure, suggesting additional mechanisms operate in conjunction with type 2 immunity to drive asthma pathogenesis. We employed RNA-Seq profiling of sputum-derived cells to identify gene networks operative at baseline in house dust mite-sensitized (HDMS) subjects with/without wheezing history that are characteristic of the ongoing asthmatic state. The expression of type 2 effectors (IL-5, IL-13) was equivalent in both cohorts of subjects. However, in HDMS-wheezers they were associated with upregulation of two coexpression modules comprising multiple type 2- and epithelial-associated genes. The first module was interlinked by the hubs EGFR, ERBB2, CDH1 and IL-13. The second module was associated with CDHR3 and mucociliary clearance genes. Our findings provide new insight into the molecular mechanisms operative at baseline in the airway mucosa in atopic asthmatics undergoing natural aeroallergen exposure, and suggest that susceptibility to asthma amongst these subjects involves complex interactions between type 2- and epithelial-associated gene networks, which are not operative in equivalently sensitized/exposed atopic non-asthmatics.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Differential gene network comparing HDMS nonwheezers versus nonatopic controls. Differentially expressed genes were identified with an edgeR analysis. Gene expression patterns in sputum were compared between HDMS nonwheezers and nonatopic controls. The wiring diagram of the underlying gene network was reconstructed employing prior knowledge from the literature (Ingenuity Knowledge Base). Genes highlighted in red denote upregulation, whilst green indicates downregulation in HDMS nonwheezers.
Figure 2
Figure 2
Differential gene network comparing HDMS wheezers versus nonatopic controls. Differentially expressed genes were identified with an edgeR analysis. Gene expression patterns in sputum were compared between HDMS wheezers and nonatopic controls. The wiring diagram of the underlying gene network was reconstructed using prior knowledge from the literature (Ingenuity Knowledge Base). Genes highlighted in red denote upregulation, whilst molecules in green indicate downregulation in HDMS wheezers.
Figure 3
Figure 3
Differential gene network comparing HDMS wheezers versus HDMS nonwheezers. Differentially expressed genes were identified with an edgeR analysis. Gene expression patterns in sputum were compared between HDMS wheezers versus HDMS nonwheezers. The wiring diagram of the underlying gene network was reconstructed employing prior knowledge from the literature (Ingenuity Knowledge Base). Genes highlighted in red denote upregulation, whilst molecules in green indicate downregulation in HDMS wheezers.
Figure 4
Figure 4
Common and unique genes networked with the 4 hub genes: EGFR, ERBB2, CDH1 and IL-13. Venn diagram illustrating common and unique genes that are networked with each hub gene in merged modules “P” and “Q”.
Figure 5
Figure 5
Differential gene network comparing HDMS wheezers versus nonatopic controls/HDMS nonwheezers. Gene co-expression network analysis (WGCNA) was employed to construct the gene networks and unbiased correlation patterns were utilized to reconstruct the underlying wiring diagram of the mucociliary clearance module “A”. CDHR3 is identified as a hub and dominant hubs are highlighted in red.
Figure 6
Figure 6
Increased expression of CDHR3 and EGFR in bronchial epithelial cells from HDM sensitized asthmatics versus nonatopic controls. The expression of the hub genes CDHR3, EGFR and ERBB2 was validated at the protein-level in an independent cohort. Bronchial epithelial cells were obtained from HDM sentitized atopics with asthma and nonatopic controls. (a) Bronchial epithelial cells were immunofluorescently stained for CDHR3 expression, EGFR expression, ERBB2 expression and nuclei with DAPI (blue). Staining images were then overlaid over bright field images taken of the same field of view. Note: mag 200×; inset 400×. (b) Quantification of the images demonstrated that the expression of CDHR3 and EGFR was more intense in the atopics with asthma. The expression of ERBB2 was not different between the groups. Mann-Whitney U Test was utilized to test for statistical significance. ***p-value < 0.001, **p-value < 0.01.

References

    1. Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012;18:716–725. doi: 10.1038/nm.2678.
    1. Busse WW, Lemanske RF, Jr., Gern JE. Role of viral respiratory infections in asthma and asthma exacerbations. Lancet (London, England) 2010;376:826–834. doi: 10.1016/S0140-6736(10)61380-3.
    1. Holt PG, Sly PD. Viral infections and atopy in asthma pathogenesis: new rationales for asthma prevention and treatment. Nat Med. 2012;18:726–735. doi: 10.1038/nm.2768.
    1. Bai TR, Vonk JM, Postma DS, Boezen HM. Severe exacerbations predict excess lung function decline in asthma. The European respiratory journal. 2007;30:452–456. doi: 10.1183/09031936.00165106.
    1. O’Byrne PM, Pedersen S, Lamm CJ, Tan WC, Busse WW. Severe exacerbations and decline in lung function in asthma. American journal of respiratory and critical care medicine. 2009;179:19–24. doi: 10.1164/rccm.200807-1126OC.
    1. Bosco A, Ehteshami S, Stern DA, Martinez FD. Decreased activation of inflammatory networks during acute asthma exacerbations is associated with chronic airflow obstruction. Mucosal Immunol. 2010;3:399–409. doi: 10.1038/mi.2010.13.
    1. Saglani S, Lloyd CM. Novel concepts in airway inflammation and remodelling in asthma. The European respiratory journal. 2015;46:1796–1804. doi: 10.1183/13993003.01196-2014.
    1. Busse WW, et al. Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children. N Engl J Med. 2011;364:1005–1015. doi: 10.1056/NEJMoa1009705.
    1. Teach SJ, et al. Preseasonal treatment with either omalizumab or an inhaled corticosteroid boost to prevent fall asthma exacerbations. The Journal of allergy and clinical immunology. 2015;136:1476–1485. doi: 10.1016/j.jaci.2015.09.008.
    1. Wenzel S, Wilbraham D, Fuller R, Getz EB, Longphre M. Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet (London, England) 2007;370:1422–1431. doi: 10.1016/S0140-6736(07)61600-6.
    1. Straker LM, et al. Rationale, design and methods for the 22 year follow-up of the Western Australian Pregnancy Cohort (Raine) Study. BMC Public Health. 2015;15:663. doi: 10.1186/s12889-015-1944-6.
    1. Hollams EM, et al. Elucidation of asthma phenotypes in atopic teenagers through parallel immunophenotypic and clinical profiling. The Journal of allergy and clinical immunology. 2009;124(463–470):470 e461–416.
    1. Colloff MJ, Stewart GA, Thompson PJ. House dust acarofauna and Der p I equivalent in Australia: the relative importance of Dermatophagoides pteronyssinus and Euroglyphus maynei. Clin Exp Allergy. 1991;21:225–230. doi: 10.1111/j.1365-2222.1991.tb00834.x.
    1. Baines KJ, Simpson JL, Wood LG, Scott RJ, Gibson PG. Transcriptional phenotypes of asthma defined by gene expression profiling of induced sputum samples. The Journal of allergy and clinical immunology. 2011;127(153–160):160 e151–159.
    1. Yan X, et al. Noninvasive analysis of the sputum transcriptome discriminates clinical phenotypes of asthma. American journal of respiratory and critical care medicine. 2015;191:1116–1125. doi: 10.1164/rccm.201408-1440OC.
    1. Bosco A, Ehteshami S, Panyala S, Martinez FD. Interferon regulatory factor 7 is a major hub connecting interferon-mediated responses in virus-induced asthma exacerbations in vivo. J Allergy Clin Immunol. 2012;129:88–94. doi: 10.1016/j.jaci.2011.10.038.
    1. Ma’ayan A. Insights into the organization of biochemical regulatory networks using graph theory analyses. J Biol Chem. 2009;284:5451–5455. doi: 10.1074/jbc.R800056200.
    1. Lu X, Jain VV, Finn PW, Perkins DL. Hubs in biological interaction networks exhibit low changes in expression in experimental asthma. Molecular systems biology. 2007;3:98. doi: 10.1038/msb4100138.
    1. Bosco A, McKenna KL, Firth MJ, Sly PD, Holt PG. A network modeling approach to analysis of the Th2 memory responses underlying human atopic disease. J Immunol. 2009;182:6011–6021. doi: 10.4049/jimmunol.0804125.
    1. Bonnelykke K, et al. A genome-wide association study identifies CDHR3 as a susceptibility locus for early childhood asthma with severe exacerbations. Nat Genet. 2014;46:51–55. doi: 10.1038/ng.2830.
    1. Bochkov YA, et al. Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:5485–5490. doi: 10.1073/pnas.1421178112.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Kramer A, Green J, Pollard J, Jr., Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics (Oxford, England) 2014;30:523–530. doi: 10.1093/bioinformatics/btt703.
    1. Wills-Karp M. Interleukin-13 in asthma pathogenesis. Immunol Rev. 2004;202:175–190. doi: 10.1111/j.0105-2896.2004.00215.x.
    1. Hollams EM, et al. Elucidation of asthma phenotypes in atopic teenagers through parallel immunophenotypic and clinical profiling. J Allergy Clin Immunol. 2009;124(463–470):470 e461–416.
    1. Djukanovic R, et al. Mucosal inflammation in asthma. The American review of respiratory disease. 1990;142:434–457. doi: 10.1164/ajrccm/142.2.434.
    1. Holgate S. The inflammation-repair cycle in asthma: the pivotal role of the airway epithelium. Clin Exp Allergy. 1998;28(S5):97–103. doi: 10.1046/j.1365-2222.1998.028s5097.x.
    1. Djukanovic R, et al. Bronchial mucosal manifestations of atopy: a comparison of markers of inflammation between atopic asthmatics, atopic nonasthmatics and healthy controls. The European respiratory journal. 1992;5:538–544.
    1. Vallath S, Hynds RE, Succony L, Janes SM, Giangreco A. Targeting EGFR signalling in chronic lung disease: therapeutic challenges and opportunities. The European respiratory journal. 2014;44:513–522. doi: 10.1183/09031936.00146413.
    1. Puddicombe SM, et al. Involvement of the epidermal growth factor receptor in epithelial repair in asthma. FASEB J. 2000;14:1362–1374. doi: 10.1096/fj.14.10.1362.
    1. Le Cras TD, et al. Epithelial EGF receptor signaling mediates airway hyperreactivity and remodeling in a mouse model of chronic asthma. Am J Physiol Lung Cell Mol Physiol. 2011;300:L414–421. doi: 10.1152/ajplung.00346.2010.
    1. Polosa R, et al. Expression of c-erbB receptors and ligands in the bronchial epithelium of asthmatic subjects. The Journal of allergy and clinical immunology. 2002;109:75–81. doi: 10.1067/mai.2002.120274.
    1. Song GG, Lee YH. Pathway analysis of genome-wide association study on asthma. Hum Immunol. 2013;74:256–260. doi: 10.1016/j.humimm.2012.11.003.
    1. Modena BD, et al. Gene Expression Correlated with Severe Asthma Characteristics Reveals Heterogeneous Mechanisms of Severe Disease. American journal of respiratory and critical care medicine. 2017;195:1449–1463. doi: 10.1164/rccm.201607-1407OC.
    1. Jones AC, Bosco A. Using Network Analysis to Understand Severe Asthma Phenotypes. American journal of respiratory and critical care medicine. 2017;195:1409–1411. doi: 10.1164/rccm.201612-2572ED.
    1. Vermeer PD, Panko L, Karp P, Lee JH, Zabner J. Differentiation of human airway epithelia is dependent on erbB2. Am J Physiol Lung Cell Mol Physiol. 2006;291:L175–180. doi: 10.1152/ajplung.00547.2005.
    1. Kettle R, et al. Regulation of neuregulin 1beta1-induced MUC5AC and MUC5B expression in human airway epithelium. Am J Respir Cell Mol Biol. 2010;42:472–481. doi: 10.1165/rcmb.2009-0018OC.
    1. Mackay A, et al. cDNA microarray analysis of genes associated with ERBB2 (HER2/neu) overexpression in human mammary luminal epithelial cells. Oncogene. 2003;22:2680–2688. doi: 10.1038/sj.onc.1206349.
    1. Angelini PD, et al. Constitutive HER2 signaling promotes breast cancer metastasis through cellular senescence. Cancer Res. 2013;73:450–458. doi: 10.1158/0008-5472.CAN-12-2301.
    1. Erle DJ, Sheppard D. The cell biology of asthma. J Cell Biol. 2014;205:621–631. doi: 10.1083/jcb.201401050.
    1. Schroeder BW, et al. AGR2 is induced in asthma and promotes allergen-induced mucin overproduction. Am J Respir Cell Mol Biol. 2012;47:178–185. doi: 10.1165/rcmb.2011-0421OC.
    1. Lyons JJ, et al. ERBIN deficiency links STAT3 and TGF-beta pathway defects with atopy in humans. J Exp Med. 2017;214:669–680.
    1. Nawijn MC, Hackett TL, Postma DS, van Oosterhout AJ, Heijink IH. E-cadherin: gatekeeper of airway mucosa and allergic sensitization. Trends Immunol. 2011;32:248–255. doi: 10.1016/j.it.2011.03.004.
    1. Post S, et al. The composition of house dust mite is critical for mucosal barrier dysfunction and allergic sensitisation. Thorax. 2012;67:488–495. doi: 10.1136/thoraxjnl-2011-200606.
    1. Ierodiakonou D, et al. E-cadherin gene polymorphisms in asthma patients using inhaled corticosteroids. The European respiratory journal. 2011;38:1044–1052. doi: 10.1183/09031936.00194710.
    1. Kim EY, et al. Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease. Nat Med. 2008;14:633–640. doi: 10.1038/nm1770.
    1. Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2015;16:45–56. doi: 10.1038/ni.3049.
    1. Licona-Limon P, Kim LK, Palm NW, Flavell RA. TH2, allergy and group 2 innate lymphoid cells. Nat Immunol. 2013;14:536–542. doi: 10.1038/ni.2617.
    1. Schmitz J, et al. 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. 2005;23:479–490. doi: 10.1016/j.immuni.2005.09.015.
    1. Llop-Guevara A, et al. A GM-CSF/IL-33 pathway facilitates allergic airway responses to sub-threshold house dust mite exposure. PLoS One. 2014;9:e88714. doi: 10.1371/journal.pone.0088714.
    1. Hajek AR, et al. 12/15-Lipoxygenase deficiency protects mice from allergic airways inflammation and increases secretory IgA levels. The Journal of allergy and clinical immunology. 2008;122:633–639 e633. doi: 10.1016/j.jaci.2008.06.021.
    1. Sevin CM, et al. Deficiency of gp91phox inhibits allergic airway inflammation. Am J Respir Cell Mol Biol. 2013;49:396–402. doi: 10.1165/rcmb.2012-0442OC.
    1. Sehra S, et al. Periostin regulates goblet cell metaplasia in a model of allergic airway inflammation. J Immunol. 2011;186:4959–4966. doi: 10.4049/jimmunol.1002359.
    1. Sivaprasad U, et al. A nonredundant role for mouse Serpinb3a in the induction of mucus production in asthma. The Journal of allergy and clinical immunology. 2011;127(254–261):261 e251–256.
    1. Rajavelu P, et al. Airway epithelial SPDEF integrates goblet cell differentiation and pulmonary Th2 inflammation. J Clin Invest. 2015;125:2021–2031. doi: 10.1172/JCI79422.
    1. Zhen G, et al. IL-13 and epidermal growth factor receptor have critical but distinct roles in epithelial cell mucin production. Am J Respir Cell Mol Biol. 2007;36:244–253. doi: 10.1165/rcmb.2006-0180OC.
    1. Tyner JW, et al. Blocking airway mucous cell metaplasia by inhibiting EGFR antiapoptosis and IL-13 transdifferentiation signals. J Clin Invest. 2006;116:309–321. doi: 10.1172/JCI25167.
    1. Thomas B, et al. Ciliary dysfunction and ultrastructural abnormalities are features of severe asthma. The Journal of allergy and clinical immunology. 2010;126:722–729 e722. doi: 10.1016/j.jaci.2010.05.046.
    1. Griggs TF, et al. Rhinovirus C targets ciliated airway epithelial cells. Respir Res. 2017;18:84. doi: 10.1186/s12931-017-0567-0.
    1. Ross AJ, Dailey LA, Brighton LE, Devlin RB. Transcriptional profiling of mucociliary differentiation in human airway epithelial cells. Am J Respir Cell Mol Biol. 2007;37:169–185. doi: 10.1165/rcmb.2006-0466OC.
    1. Woodruff PG, et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. American journal of respiratory and critical care medicine. 2009;180:388–395. doi: 10.1164/rccm.200903-0392OC.
    1. Dougherty RH, Fahy JV. Acute exacerbations of asthma: epidemiology, biology and the exacerbation-prone phenotype. Clin Exp Allergy. 2009;39:193–202. doi: 10.1111/j.1365-2222.2008.03157.x.
    1. Gerstner JR, et al. Removal of unwanted variation reveals novel patterns of gene expression linked to sleep homeostasis in murine cortex. BMC Genomics. 2016;17:727. doi: 10.1186/s12864-016-3065-8.
    1. Wood LG, Powell H, Gibson PG. Mannitol challenge for assessment of airway responsiveness, airway inflammation and inflammatory phenotype in asthma. Clin Exp Allergy. 2010;40:232–241. doi: 10.1111/j.1365-2222.2009.03371.x.
    1. Pizzichini E, Pizzichini MM, Efthimiadis A, Hargreave FE, Dolovich J. Measurement of inflammatory indices in induced sputum: effects of selection of sputum to minimize salivary contamination. The European respiratory journal. 1996;9:1174–1180. doi: 10.1183/09031936.96.09061174.
    1. Liao Y, Smyth GK, Shi W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic acids research. 2013;41:e108. doi: 10.1093/nar/gkt214.
    1. Anders S, et al. Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat Protoc. 2013;8:1765–1786. doi: 10.1038/nprot.2013.099.
    1. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat Biotechnol. 2014;32:896–902. doi: 10.1038/nbt.2931.
    1. Chen EY, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC bioinformatics. 2013;14:128. doi: 10.1186/1471-2105-14-128.
    1. Jones, A. C. et al. Persistent activation of interlinked Th2-airway epithelial gene networks in sputum-derived cells from aeroallergen-sensitized symptomatic atopic asthmatics. bioRxiv 063602, 10.1101/063602.
    1. Boulet LP, FitzGerald JM, Reddel HK. The revised 2014 GINA strategy report: opportunities for change. Curr Opin Pulm Med. 2015;21:1–7. doi: 10.1097/MCP.0000000000000125.
    1. Quanjer PH, et al. Multi-ethnic reference values for spirometry for the 3-95-yr age range: the global lung function 2012 equations. The European respiratory journal. 2012;40:1324–1343. doi: 10.1183/09031936.00080312.

Source: PubMed

Подписаться