@prefix vivo: . @prefix edm: . @prefix ns0: . @prefix dcterms: . @prefix skos: . vivo:departmentOrSchool "Medicine, Faculty of"@en, "Non UBC"@en, "Anesthesiology, Pharmacology and Therapeutics, Department of"@en ; edm:dataProvider "DSpace"@en ; ns0:identifierCitation "Respiratory Research. 2016 Sep 22;17(1):119"@en ; ns0:rightsCopyright "The Author(s)."@en ; dcterms:creator "Moheimani, Fatemeh"@en, "Hsu, Alan C"@en, "Reid, Andrew T"@en, "Williams, Teresa"@en, "Kicic, Anthony"@en, "Stick, Stephen M"@en, "Hansbro, Philip M"@en, "Wark, Peter A"@en, "Knight, Darryl A"@en ; dcterms:issued "2018-05-14T16:30:20Z"@en, "2016-09-22"@en ; dcterms:description "Asthma is a global health problem with increasing prevalence. The airway epithelium is the initial barrier against inhaled noxious agents or aeroallergens. In asthma, the airway epithelium suffers from structural and functional abnormalities and as such, is more susceptible to normally innocuous environmental stimuli. The epithelial structural and functional impairments are now recognised as a significant contributing factor to asthma pathogenesis. Both genetic and environmental risk factors play important roles in the development of asthma with an increasing number of genes associated with asthma susceptibility being expressed in airway epithelium. Epigenetic factors that regulate airway epithelial structure and function are also an attractive area for assessment of susceptibility to asthma. In this review we provide a comprehensive discussion on genetic factors; from using linkage designs and candidate gene association studies to genome-wide association studies and whole genome sequencing, and epigenetic factors; DNA methylation, histone modifications, and non-coding RNAs (especially microRNAs), in airway epithelial cells that are functionally associated with asthma pathogenesis. Our aims were to introduce potential predictors or therapeutic targets for asthma in airway epithelium. Overall, we found very small overlap in asthma susceptibility genes identified with different technologies. Some potential biomarkers are IRAKM, PCDH1, ORMDL3/GSDMB, IL-33, CDHR3 and CST1 in airway epithelial cells. Recent studies on epigenetic regulatory factors have further provided novel insights to the field, particularly their effect on regulation of some of the asthma susceptibility genes (e.g. methylation of ADAM33). Among the epigenetic regulatory mechanisms, microRNA networks have been shown to regulate a major portion of post-transcriptional gene regulation. Particularly, miR-19a may have some therapeutic potential."@en ; edm:aggregatedCHO "https://circle.library.ubc.ca/rest/handle/2429/65851?expand=metadata"@en ; skos:note "REVIEW Open AccessThe genetic and epigenetic landscapes ofthe epithelium in asthmaFatemeh Moheimani1,2* , Alan C-Y Hsu1,2, Andrew T Reid1,2, Teresa Williams1,2,3, Anthony Kicic4,5,6,7,Stephen M. Stick4,5,6,7, Philip M. Hansbro1,2, Peter A.B. Wark2,8 and Darryl A. Knight1,2,9AbstractAsthma is a global health problem with increasing prevalence. The airway epithelium is the initial barrier againstinhaled noxious agents or aeroallergens. In asthma, the airway epithelium suffers from structural and functionalabnormalities and as such, is more susceptible to normally innocuous environmental stimuli. The epithelial structuraland functional impairments are now recognised as a significant contributing factor to asthma pathogenesis. Bothgenetic and environmental risk factors play important roles in the development of asthma with an increasing numberof genes associated with asthma susceptibility being expressed in airway epithelium. Epigenetic factors that regulateairway epithelial structure and function are also an attractive area for assessment of susceptibility to asthma. In thisreview we provide a comprehensive discussion on genetic factors; from using linkage designs and candidate geneassociation studies to genome-wide association studies and whole genome sequencing, and epigenetic factors; DNAmethylation, histone modifications, and non-coding RNAs (especially microRNAs), in airway epithelial cells that arefunctionally associated with asthma pathogenesis. Our aims were to introduce potential predictors or therapeutictargets for asthma in airway epithelium. Overall, we found very small overlap in asthma susceptibility genes identifiedwith different technologies. Some potential biomarkers are IRAKM, PCDH1, ORMDL3/GSDMB, IL-33, CDHR3 and CST1 inairway epithelial cells. Recent studies on epigenetic regulatory factors have further provided novel insights to the field,particularly their effect on regulation of some of the asthma susceptibility genes (e.g. methylation of ADAM33).Among the epigenetic regulatory mechanisms, microRNA networks have been shown to regulate a majorportion of post-transcriptional gene regulation. Particularly, miR-19a may have some therapeutic potential.Keywords: Epithelial cells, Asthma, Genes, DNA methylation, Histone acetylation, microRNABackgroundAsthma affects people of all ethnicities and ages andthere has been a substantial increase in the prevalence ofasthma over the past few decades, with current estimatesof approximately 300 million people suffering from thedisease worldwide [1]. Asthma is characterised by cough-ing, shortness of breath, chest tightness and wheezing,often triggered by exposure to allergens and foreign patho-gens [1]. The initial response consists primarily of airwaysmooth muscle constriction and airway inflammation(oedema, inflammatory cell infiltration, increased airwaysecretions). Whereas more chronic responses such asstructural remodelling of the airway including smoothmuscle and sub-mucosal gland hyperplasia and hyper-trophy, extracellular matrix (ECM) deposition and angio-genesis are generally thought to occur in parallel withinflammatory responses [1].The airway epithelium is the interface between the res-pirable environment and the sub-mucosa and acts as thefirst defence line against inhaled noxious agents andaeroallergens [2]. The epithelium of conducting airwaysis pseudo-stratified and consists of ciliated columnar epi-thelial cells, goblet cells, intermediate columnar epithe-lial cells, side population cells, serous cells, and basalcells [3]. The epithelium of asthmatics presents severalstructural and functional abnormalities, including agreater proportion of resident stem cells and basal cells,goblet-cell hyperplasia and excessive mucus production* Correspondence: fatemeh.moheimani@newcastle.edu.au1School of Biomedical Sciences and Pharmacy, Faculty of Health andMedicine, HMRI building, The University of Newcastle, Callaghan, NSW 2308,Australia2Priority Research Centre for Healthy Lungs, Hunter Medical ResearchInstitute, The University of Newcastle, New South Wales, AustraliaFull list of author information is available at the end of the article© 2016 The Author(s). Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, andreproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link tothe Creative Commons license, and indicate if changes were made. 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.Moheimani et al. Respiratory Research (2016) 17:119 DOI 10.1186/s12931-016-0434-4as well as fewer ciliated cells compared to healthy individ-uals, suggesting dysregulated differentiation [4, 5]. Theepithelial abnormalities are associated with increasedsusceptibility to oxidant-induced stress, aberrant cyto-kine and ECM release [6], mitotic dyssynchrony [7]and a deficient innate immune response [8–10]. Theseabnormalities also affect epithelial repair and regener-ation processes after injury, leading to defective main-tenance of the epithelial barrier and its normalfunction [2, 11, 12].Both genetic susceptibility and environmental risk factorsinfluence asthma [13]. Genetic studies have progressedfrom using linkage designs and candidate gene associationstudies to genome-wide association studies (GWAS) [14]and whole genome sequencing (WGS) [15], and detecteddifferent genes associated with asthma susceptibility. Sev-eral of the recent GWAS have shown to have a significantepithelial contribution, which also segregates away from al-lergy/atopy. Furthermore, different environmental chal-lenges (such as smoking, air pollution, and microbialexposures) can affect gene expression through epigeneticregulation. Epigenetic factors are an important regulator ofgene transcription, that do not influence gene sequence[16]. Epigenetic mechanisms include DNA methylation,histone modifications, and regulation by non-coding RNAs,especially microRNAs (miRNAs). This review will focus onidentifying genetic and epigenetic candidates in airwayepithelium, which are functionally associated with asthmaand may act as predictors or therapeutic targets.Asthma susceptibility genes in airway epitheliumEarly genetic studies relied on positional cloning in com-bination with linkage analysis leading to detection ofgenes associated with asthma expressed in airway epithe-lium including A disintegrin and metalloprotease 33(ADAM33), GPRA, protocadherin-1 (PCDH1), Serineprotease inhibitor Kazal type-5 (SPINK5), IL-1 receptorassociated kinase-M (IRAKM), Dipeptidyl-peptidase 10(DPP10) and HLA-G genes (Table 1) [17–31]. ADAM33on chromosome 20p13, was the first asthma susceptibil-ity gene discovered [17]. ADAM33 protein is expressedin many cells including the airway epithelium [18], fibro-blasts and smooth muscle cells [17, 18, 32] and is knownas a membrane-anchored metalloprotease with diversefunctions, including shedding of cell-surface proteinssuch as cytokines and cytokine receptors [17]. ADAM33has be associated with airway remodelling and bronchialhyperresponsiveness (BHR) through epithelial–mesen-chymal trophic unit (EMTU), leading to proliferation ofbiosynthetically active fibroblasts, myofibroblasts andsmooth muscle [17]. PCDH1 is located on chromosome5q31-q33 and encodes the protocadherin-1 protein[22, 23]. The expression of PCDH1 is aligned with theapical adhesion complex expression in airway epithelialTable 1 Asthma susceptibility genes identified by positional cloning and genome-wide association (GWAS) in airway epitheliumPositional cloningChromosome Gene Function Reference20p13 ADAM33 Airway remodelling and BHR [17]5q31-q33 PCDH1a Airway remodelling and BHR [22–24]2q14-32 DPP10 BHR [27–29]6p21 HLA-G BHR [30, 31]7p15-p14 GPRA/NPSR1/GPR154 Cell homeostasis [19–21]5q31-35 SPINK5/LEKTI Protective against allergen/inflammation [25]12q13-24 IRAKMb Inflammation [26]Genome-wide association (GWAS) and associated SNPChromosome SNP Gene Function References2 rs3771166 IL1RL1 andIL18R1Alarmin to alert the immune system after epithelial cell damageduring trauma or infection[45, 50]6 re9273349 HLA-DQ Recognition of non-self antigens [13]9 rs1342326 IL33+ Alarmin [13, 50]15 rs744910 SMAD3 TGF-β1 signaling and response to respiratory viral infection [50, 57–59]17 rs2305480 ORMDL3b+GSDMBb+BHREpithelial cell homeostasis[13, 64, 66–68]5 rs1837253 TSLP+ Epithelial cell homeostasis and improving wound healing-Protectiverole against asthma[42, 43, 71–74]aAdult and childrenbEarly onset+common between different ethnic groupMoheimani et al. Respiratory Research (2016) 17:119 Page 2 of 15cells hence association of PCDH1 with asthma is proposedto be through epithelial structural defects leading to BHR[22, 23] and is IgE independent [24]. Dysregulation ofPCDH1 expression in asthma also leads to impaired differ-entiation of epithelial cells [23]. Another gene is DPP10which shown to preferentially expressed in the epitheliumof asthmatics [27]. DPP10, is located on 2q14-32 and en-codes the di-peptidyl peptidase like 10 protein, which un-like other members of DPP family is unable to cleave theterminal of dipeptides from cytokines and chemokines[27, 28]. These suggest other potential mechanisms forDPP10 association with asthma. In the nervous system,DPP10 has been shown to modulate the electrophysio-logical properties, cell-surface expression and subcellularlocalisation of voltage-gated potassium channels [33].Considering the important role of potassium ion channelsin asthma [34], DPP10 may also be involved in thisprocess although this requires further investigation. Fur-thermore, Zhou et al. reported the association of DPP10with BHR in Chinese population [29]. HLA-G on chromo-some 6p21 is also expressed highly in bronchial epithelialcells of asthmatics and is associated with BHR [30].HLA-G inhibits the effecter function of T cells and nat-ural killer (NK) cells [35]. Three miRNAs; miR-148a,miR-148b, and miR-152 have been reported to affectHLA-G expression, suggesting that miRNA mediatedmechanisms may contribute to the impact of HLA-Gon asthma risk [31].Other studies detected GPRA (also known as Neuro-peptide S Receptor 1; NPSR1, and GPR154) on chromo-some 7p15-p14 [19–21]. Both GPRA, which belongs tothe G protein-coupled receptor family, and its agonist,Neuropeptide S (NPS) are co-expressed in bronchial epi-thelium and specific activation of the GPRA-A isoformwith NPS inhibits cell growth [19, 20]. Since the balancebetween epithelial cell proliferation and regeneration isdysregulated in asthmatics [4, 36], GPRA likely plays animportant role in the pathogenesis of disease [19, 20].Further studies identified the SPINK5 gene on chromosome5q31-35 which encodes a multidomain serine proteaseinhibitor known as lympho-epithelial Kazal-type-relatedinhibitor (LEKTI). LEKTI has been shown to be a majorphysiological inhibitor of multiple serine proteinases, in-cluding the exogenous serine proteases trypsin, plasmin,subtilisin A, cathepsin G and neutrophil elastase [37].SPINK5 is essential in the epidermal barrier functionthrough regulating protease activity [38] and LEKTI playsa crucial role in skin homeostasis by selectively inhibitinghuman kallikrein-related peptidase genes including, KLK5,KLK7 and KLK14 [39]. LEKTI may therefore protect theepithelium against allergens or inflammatory related pro-teases. However, the exact function of SPINK5 in airwayepithelium remains to be elucidated. Another asthma sus-ceptibly gene is IRAKM, which is located on chromosome12q13-24. IRAK-M regulates NF-kB and inflammation viasuppressing Toll-like receptor/IL-1R pathways. WhenIRAK-M function is hampered, overproduction of inflam-matory cytokines in the lung in response to infection/aller-gens may result in a Th2-mediated allergic response and/or Th1-dependent exacerbation of asthma symptoms [26].Further technological advances led to GWAS [40], andassociated single nucleotide polymorphisms (SNPs) [13],which detected a completely different set of genes; inter-leukin (IL) 1 receptor-like 1 (IL1RL1) and IL18 receptor1 (IL18R1), IL33, HLA-DQ, SMAD3, thymic stromallymphopoietin (TSLP), ORM1-like 3 (ORMDL3) andgasdermin B (GSDMB) as asthma susceptibility genesexpressed in airway epithelium (Table 1) [13, 14, 41–43].IL1RL1 and IL18R1 contain SNP rs3771166 on chromo-some 2 [13, 44]. IL1RL1 (also known as T1, ST2, DER4,or FIT-1) belongs to the IL-1 superfamily and is the re-ceptor for IL-33 [45]. IL33 with SNP rs1342326 locatedon chromosome 9 is also associated with atopic asthma[13, 46, 47]. IL-33 possesses potent transcriptional-repressive properties and is constitutively expressed inepithelial cells [48]. It has been shown that IL-33 activatesNF-kB and mitogen-activated protein (MAP) kinases, andinduces production of T-helper (Th) 2-associated cyto-kines, including IL-4, IL-5, and IL-13 [49]. In this context,IL-33 functions as a prototypical ‘alarmin’ and an en-dogenous ‘danger’ signal to alert the immune system afterepithelial cell damage during trauma or infection [50] andplays an essential role in pro-inflammatory pathway inasthma [13]. IL18R1 encodes the receptor for IL-18 [45].IL-18 modulates innate and adaptive immune responsesby increasing interferon (IFN)-γ production by Th1 andnatural killer (NK) cells or by activating IgE productionand Th2 cell differentiation [45].Another candidate gene detected by GWAS is HLA-DQregion of the major histocompatibility (MHC) gene lo-cated on chromosome 6, which contains SNP rs9273349[51]. The airway epithelium expresses MHC class II; a het-erodimer molecule that consists of an α- and a β-chain inone of three HLA loci: DR, DP and DQ [52], on theirsurface [53]. Immune response to allergens is also re-lated to specific HLA-DR and DQ haplotypes [13], andis associated with asthma induced by house dust mite,aspirin, soybean, and occupational triggers [54]. How-ever, the exact role of HLA-DQ in airway epitheliumstill remains unclear.SMAD3, with SNP rs744910 located on chromosome15, is another asthma susceptibility gene [13]. SMAD3 isan essential signal transducer in transforming growthfactor (TGF)-β signalling, which is elevated in airwayepithelial cells of some asthmatics [55]. TGF-β1 inducesepithelial–mesenchymal transition (EMT) in airwayepithelial cells via a SMAD3-dependent transcriptionfactor snail1 (SNAI1) which transcriptionally supressesMoheimani et al. Respiratory Research (2016) 17:119 Page 3 of 15E-cadherin [36, 56]. Furthermore, the TGF-β/SMAD3pathway play essential roles in the airway epithelial re-sponse to respiratory viral infection [57–59], includingincreasing replication of both respiratory syncytial virus[57, 58] and rhinovirus [59].Among asthma susceptibility genes, ORMDL3 andGSDMB, with SNP rs2305480 at chromosome 17q21,are associated with childhood asthma [60]. ORMDL3 isa member of a gene family that encodes transmembraneproteins anchored in the endoplasmic reticulum of airwayepithelial cells, predominantly [60, 61]. Allergens induceORMDL3 expression in airway epithelium leading to in-creased expression of asthma-associated chemokines,metalloproteases and the unfolded protein response(UPR), which may implicate the potential link betweenORMDL3 and asthma [61]. ORMDL regulates ORMprotein expression in airway epithelial cells, which isinduced in response to allergen challenge [62]. ORMproteins are important homeostatic regulators of sphingo-lipid metabolism [63], which is associated with the patho-genesis of asthma [64]. Sphingolipids are pivotal inmaintenance of cell structure and signaling pathways inphysiological and pathological processes; e.g. proliferation,apoptosis and migration [63, 65] and have been shown tocontribute to BHR in experimental models of asthma [66].Further meta-analysis has showed that SNP rs7216389 inthe ORMDL3 may play essential and independent predis-posing roles in ethnically diverse populations for bothchildhood and adult-onset asthma [41]. GSDMB is adja-cent to ORMDL3 and is a member of gasdermin familythat encodes gasdermin B protein which has roles insecretory pathways, epithelial cell differentiation, cell cyclecontrol and apoptosis [67, 68]. Furthermore, there are sev-eral response elements for interferon regulatory factorspresent in the GSDMB promoter region and epithelialinterferon-α induces GSDMB gene and protein in humannasal epithelial cells, in vitro [69]. GSDMB has beenproposed to be the causative gene associated withasthma [70].Among the candidate genes identified by GWAS, TSLPon chromosome 5 plays protective roles against the riskof asthma, atopic asthma and BHR across various ethnicgroups [42, 43, 71–73]. The rs1837253 SNP may be dir-ectly involved in the regulation of TSLP secretion inprimary nasal epithelial cells [42]. TSLP is an IL-7 likecytokine that induces myeloid dendritic cells to stimu-late the differentiation of naive CD4+ T cells to Th2cells. TSLP mRNA and protein are highly expressed inthe asthmatic airway epithelium [72–74]. TSLP hasbeen shown to induce bronchial epithelial cell prolifer-ation and increases repair responses to injury throughIL-13 production [74].Collectively these genes are important in epithelialcell damage, innate and adaptive immunity, and airwayinflammation, which are pivotal in the pathology ofasthma. Furthermore, some of the products associatedwith these genes can determine the phenotype of asthma.For instance, the level of IL-33 is highly elevated andwidely distributed in bronchial epithelial cells of moderateand severe asthmatics [48]. IL-18 may also contribute toasthma exacerbations in mild and moderate asthmaticsthrough activation of immunologic responses [51]. Giventhe relationship to asthma endotypes, these genes may in-dicate pathways for therapeutic intervention. In fact, PhaseII trials are currently proceeding using an anti-TSLP anti-body; AMG 157 from Amgen Corp., to neutralise theTSLP cytokine for the treatment of allergic diseases asasthma [75].Notably, only a few genes, such as IL33 and TSLP, areshared among all asthmatics [42, 76, 77] and may playroles as potential biomarkers. Furthermore, while theassociation of the 17q21 locus (ORMD/GSDMB) withasthma is the most consistent finding from differentstudies, there is limited evidence to validate certainSNPs [14]. Integrative genomics defined as identificationof causal genes and variants, with improved statisticalpower, is a promising new approach. By using gene ex-pression as a phenotype and examining how DNApolymorphisms contribute to both gene expression(expression quantitative trait loci; eQTLs) and diseasephenotypes, true causal relationships can be discovered[78–80]. Although GWAS have identified loci that arestrongly associated with asthma, the molecular mecha-nisms underlying these associations rely on other technol-ogy such as eQTLs [78].One eQTL study showed that chromosome 17q21,which contained strong GWAS hits, also regulates ex-pression levels of cyclin-dependent kinase 12 (CDK12),protein phosphatase 1 regulatory subunit 1B (PPP1R1B),titin-cap (TCAP) and StAR-related lipid transfer(START) domain containing 3 (STARD3) genes in theairway epithelium [78]. CDK12 is a member of thecyclin-dependent kinase (CDK) family, which areserine/threonine kinases regulating cell cycle progression[78, 81]. Airway epithelial cells from asthmatics overexpressthe CDK inhibitor; p21waf [82], which may explain the ab-normal repair responses of the airway epithelium of asth-matics after wounding [82]. However, the role of TCAP,PPP1R1B, STARD3 in asthma are still unknown [78]. Fur-thermore, epithelial eQTL detected Cystatin SN (CST1) onchromosome 20p11.21, which contains SNP rs16856186[78]. CST1 may neutralise cystatin C; a potent cathepsinB inhibitor, and increase cell proliferation [83]. CST1 isexpressed differentially in airway cells of asthmaticswith exercise-induced bronchoconstriction (EIB) comparedto asthmatics without EIB [84]. eQTL also confirmedcadherin-related family member 3 (CDHR3) gene, as anepithelial susceptibly gene for severe exacerbations inMoheimani et al. Respiratory Research (2016) 17:119 Page 4 of 15childhood asthma [78, 85]. CDHR3 encodes a hemophiliccell adhesion molecule, which may be involved in maintain-ing cell integrity by forming cell-cell junctions. Further-more, functional disruption of CDHR3 has been reportedin human rhinovirus-induced asthma exacerbation [86].Also, epithelial eQTL supported SPINK5 as an asthma sus-ceptibility gene [86], as described earlier.It is also essential to note that in a disease as complexas asthma, it is unlikely that one or a few functional genevariants will be responsible for all pathophysiologicalevents. While GWAS have been useful and continue toidentify novel genes for allergic diseases through in-creased sample sizes and phenotype refinement, furtherapproaches to integrate analyses of rare variants, eQTLapproaches, and epigenetic mechanisms will likely leadto greater insight into the genetic basis of the disease.The advent of whole genome sequencing (WGS), whichincludes copy number variants (CNVs) and low-frequencyvariants, has been proposed to overcome the drawbacks ofthe earlier technologies [15, 76]. CNVs, which are geneticvariants including the deletion or duplication of more than50 bp of gene sequence [15], are one the most recent ad-vances to detect asthma susceptibility genes. Recently, anassociation between a 6 kbp deletion in an intron ofNEDD4L with increased risk of asthma was reported butonly in Hutterites [15]. NEDD4L is expressed in bronchialepithelial cells, and NEDD4L knockout mice showed se-vere airway inflammation and mucus accumulation [15].To adequately assess the entire genome, a large numberof genetic polymorphisms (250,000 to 1 million) is requiredand the number of polymorphisms will vary between stud-ies due to different levels of linkage disequilibrium [14].Currently, WGS is neither affordable nor feasible on thelarge number of individuals to acquire sufficient power fordetecting associations with asthma [15].Effect of environmental exposure on asthmaEnvironmental factors play essential roles in asthma aeti-ology. The increase in the prevalence of asthma world-wide during recent decades, the substantial variations inpopulations with a similar racial and ethnic backgroundbut exposed to different environmental stimuli, and thesignificant increase in the frequency of occupationalasthma are all pointing out toward the important role ofenvironmental factors [87].Environmental stimuli affecting asthma are categorisedto outdoor and indoor factors. Outdoor stimuli thattrigger or exacerbate asthma include microbial and viralpathogens, airborne particulates, ozone, diesel exhaustparticles, pollens, outdoor moulds, environmental tobaccosmoke, cold air, and humidity [87, 88]. Indoor environ-mental factors include allergens derived from dust mites,cockroaches, mice and pets which has been shown to in-duce airway inflammation; particles generated from indoorburning of tobacco, wood, and biomass; and biologicalagents such as indoor endotoxin, products from gram-positive bacteria, and 1,3-β-glucans from moulds [87, 88].In particular relation to asthma susceptibility, the ex-posure to specific environmental factors can play keyfactor in the induction or suppression of asthma-relatedgenes. The main areas of studies in regards to the impactof gene-environment interactions on asthma developmentand pathogenesis have been so far related to smoking, airpollution, and microbial exposures. Maternal smoking isone of the major risk factor for asthma in offspring. Ma-ternal smoking substantially enhances the strength of thelinkage signal on chromosome 5q31–34 to asthma in thechildren [89, 90]. Furthermore, polymorphic variationin candidate genes known to be involved in asthma, forexample TNF-308 and glutathione-S-transferase M1(GSTM1; involved in detoxification of oxidative stressand lung function growth in children), are predictors ofBHR to passive smoking [89, 91]. The most well-knowninteraction between environmental factors and gene isbetween endotoxin with Toll like receptor (TLR)-4 withfurther impact on adaptive immune response, epithelialand smooth muscle cells through NF-kB. Polymorph-ism in TLR-4 is related to asthma and it is proposedthat the other TLRs (e.g. TLR-9 and -3) present thesimilar polymorphic associations with other environ-mental stimuli, such as CpG methylation of TLR-9 anddouble-stranded RNA (dsRNA) for TLR-3 [89, 92].These reports point out to the importance of early lifeenvironmental factors, such as passive smoking, pollu-tant exposure and viral infections, as a perverse factoron the developing asthma in childhood.However little is known about the effect ofenvironmental-gene interactions in airway epitheliumof asthmatics. It has been shown that particulate matterwith a diameter of <10 μm diameter (known as PM10) in-creases HAT activity and the level of acetylated histone 4(H4) through oxidative stress. PM10 induced histoneacetylation is associated with promoter region of the IL-8resulting in increased IL-8 gene and protein release fromalveolar epithelial (A549) cells [93]. Interestingly, butyrate;a fermentation product of intestinal bacteria, also showedto enhance histone acetylation by inhibition of HDACenzymes leading to an increase in gene expression ofinflammatory cytokines in intestinal epithelial cells [94].Cigarette smoke-induced oxidative stress also reducesHDAC2 and increases cytokines expression in alveolarmacrophages [95] but the effect on airway epithelium isyet to be determined.Most importantly, many of the indoor and outdoorasthma triggers also have demonstrable reprogrammingeffects on the immature airway during early life, leadingto altered asthma risk in later life. Asthma hence is not ahomogeneous disease but a condition influenced byMoheimani et al. Respiratory Research (2016) 17:119 Page 5 of 15interactions between genetic and environmental factorsthrough epigenetic mechanisms that influence geneexpression.Epigenetic regulatory factors in airway epitheliumEnvironmental challenges can affect gene expressionthrough epigenetic mechanisms. Epigenetics is describedas a heritable regulation of gene transcription that doesnot require alterations in gene sequence [16]. Epigeneticchanges may form stable heritable changes in gene ex-pression and in a tissue-specific fashion [96, 97]. Par-ticularly, epigenetic regulation affects gene expressionthrough three main mechanisms, including DNA modi-fications, histone modifications, and non-coding RNAs(Fig. 1 and Table 2).DNA modificationsModification of DNA occurs through addition or re-moval of small covalent molecules such as methyl oracetyl groups. DNA methylation occurs when methylgroups are added by a DNA methyltransferase (DNMT),onto cytosine nucleotides that are followed by guanineresidues (CpG sites). Gene promoters are relatively richin CpG sites which are known as CpG islands. DNAmethylation can lead to gene expression silencing throughformation of 5-methyl-cytosine (5mC) [98].Recent studies have characterised a number of DNAmethylation signatures in epithelial cells of asthmatics,including cytokeratin 5 (KRT5) [99], signal transducer andactivator of transcription 5A (STAT5A) [99], cysteine-richprotein 1 (CRIP1) [99], arginase 2 (ARG2) [100], IL-6[98], inducible nitric oxide synthase (iNOS) [98] andADAM33 [101] (Table 2).KRT5 is a marker of basal cells and its expression is in-creased in the epithelium of asthmatics [5, 36]. In asthmaticchildren, KRT5 exhibits reduced methylation resulting inincreased expression of this gene [97, 99]. Increased KRT5may hence be associated with dysregulated epithelium dif-ferentiation. The STAT5A transcription factor is activatedby different pro-Th2 cytokines (e.g. IL2, IL7, or TSLP) sug-gesting its significant role in promoting Th2 cell differenti-ation and responses [102] and epithelial cell proliferation[103]. CRIP1 has been reported to play a role in cell motil-ity, adhesion, and structure through interaction with thecytoskeletal protein actin [104] and also translocates to thenucleus to facilitate protein interactions important for tran-scriptional regulation [105]. The promoters of STAT5A andCRIP1 are hyper-methylated in epithelium of asthmaticchildren [99], resulting in decreased expression of STAT5A,contrary to increased CRIP1 expression [99]. Further stud-ies are therefore needed to understand the roles of STAT5Aand CRIP1 in epithelial function.Fig. 1 Epigenetic regulatory factors in airway epithelium. a DNA methylation; white circles represent unmethylated CpGs that induces geneexpression (e.g. KRT5) while black circles represent methylated CpGs that suppresses gene expression (e.g. STAT5A). b Histone acetylation; greencircles refer to acetylated histone tail that stimulate gene expression (e.g. ΔNp63) while red circles indicate free histone tails that suppressed geneexpression. c Noncoding RNA; miRNAs affect gene expression by either RNA degradation or translational inhibition. miRNAs hence (e.g. miR-19a)may suppress mRNA expression (e.g. TGF-β receptor 2)Moheimani et al. Respiratory Research (2016) 17:119 Page 6 of 15ARG2, IL-6 and iNOS are three methylated genes thathave been related to fractional exhalation of nitric oxide(FeNO) in asthmatic children [98, 100, 106]. Methylationof the ARG2 promoter in asthmatic children is associ-ated with reduced FeNO [100]. However, asthmaticchildren with lower DNA methylation of the IL-6 andiNOS promoters in nasal epithelial cells had higher air-way inflammation, as measured by increased FeNO[98]. Therefore, further investigation is required to de-termine the underlying biological mechanisms drivingthe association of these DNA methylations with FeNOand whether children with different degrees of asthmaseverity and symptom management have different levelsof DNA methylation.Furthermore, hyper-methylation on ADAM33 in bron-chial epithelial cells is strongly associated with BHR,irrespective of asthma status [101]. This is in contrastto ADAM33 hypo-methylation in fibroblasts, which isspeculated to be involved in airway remodelling [107].This highlights the importance of cell specific epigeneticchanges, as well as the potential challenges in developingnovel therapeutics.Recently, DNA methylation has been shown to occurin airway epithelial cells isolated from asthmatics after asingle 24 h exposure to IL-13 [108]. Intriguingly, areasof methylation were mainly adjacent to asthma suscepti-bility genes and in particular genes related to fibroticand inflammatory pathways (e.g. neutrophil cytosolic fac-tor 2; NCF2, and MMP14) [108]. Global and gene specificmethylation status in the airway epithelium however stillrequires further investigation before potential targets canbe identified and trialled as therapies for asthma.Histone modificationsThe DNA of each cell is packaged into nucleosomeswhere its 147 base pairs wrap around an octamer of fourcore histone (H2A, H2B, H3, and H4). The covalentalterations of the amino acid residues of core histoneN-terminal tails are essential for modification of thechromatin structure and regulate gene expression. ForTable 2 Epigenetic regulatory factors associated with asthma in airway epithelial cellsDNA modification signaturesGene Status Function ReferencesKRT5a Hypo-methylation Epithelial homeostasis [97, 99]STAT5Aa Hyper-methylation Immune system, Cell proliferation [99]CRIP1a Hyper-methylation Epithelial homeostasis, transcription [99]ARG2a Hyper-methylation Reduced FeNO [100]IL-6a Hypo-methylation Increased FeNO [98]iNOSa Hypo-methylation Increased FeNO [98]ADAM33 Hyper-methylation BHR [101]Histone modification signaturesHDAC/HAT Status Function ReferencesH3K18 Acetylation Increases the expression of ΔNp63, EGFR and STAT6 affecting epithelial homeostasis [109]HDAC2a De-acetylation Anti-inflammatory [112]miRNA signaturesmiRNA Status Function Referenceslet-7fb Overexpressed unknown [121]miR-487bbmiR-181cbmiR-203b Suppressed Targeting p63 and c-Abl [121–123]miR-34/449 family Suppressed Targeting NOTCH1 mRNA and affecting cell homeostasis [125]miR-18a Suppressed activation/signalling of IL-6 and IL-8 [129]miR-27amiR-128miR-155miR-19ac Overexpressed Targeting TGF-β receptor 2 mRNA and affecting cell homeostasis [130]aChildrenbMild asthmacSevere asthmaMoheimani et al. Respiratory Research (2016) 17:119 Page 7 of 15example, the acetylation of lysine residues on histonetails via histone acetyltransferases (HATs), generally re-sults in increased gene transcription whereas removal ofthe acetyl group via deacetylases (HDACs) leads to genesuppression. In contrast, methylation of histone tails canbe both activating and suppressing depending on the par-ticular residue. Methyl groups are added to lysine or argin-ine residues by histone methyltransferases (HMTs) andremoved by histone demethylases (HDMs) [109, 110].Increased HAT activity and reduced HDAC activity inbiopsies from mild asthmatics have been reported tolead to the increased expression of multiple inflammatorygenes [111]. Interestingly, these activities may be partiallyreversed by treatment with inhaled corticosteroids [111].Furthermore, within the adult airway epithelium, elevatedhistone H3 lysine 18 (H3K18) acetylation and histone H3lysine 9 trimethylation (H3K9me3) have been shown inasthmatics [109]. H3K18 acetylation increases the expres-sion of ΔNp63, EGFR, and STAT6, which, are known to bealtered in the epithelium of asthmatics [109]. Very fewstudies have investigated histone modifications in childrenwith asthma. In one study, passive cigarette smoke re-duced HDAC2 activity and protein expression via PI3Ksignalling in children with severe asthma. This is believedto suppress the anti-inflammatory effects of corticosteroidtreatment [112] (Table 2).Whether histone modifications in epithelial cells aremajor contributors in conferring asthma susceptibilityand/or severity remains to be determined.Non-coding RNAsA number of classes of noncoding RNAs have been dis-covered in mammalian cells including long non-codingRNAs (lncRNAs), Piwi-interacting RNAs (piRNAs), andmiRNAs.lncRNAs are non-protein coding RNA transcripts lon-ger than 200 nucleotides. There are approximately15,000 lncRNAs discovered so far although only a smallnumber of which have been shown to be biologicallyrelevant. piRNAs are small non-coding RNAs of 26–31nucleotides long, predominantly found in spermatogenicand ovarian cells [113]. Their functions have been linkedto both epigenetic and post-transcriptional gene silencing[113]. piRNAs interact with piwi protein, a RNA-bindingprotein, which degrades target mRNAs to prevent proteintranslation [113]. There have been no studies that exten-sively investigate profiles of lncRNAs and piRNAs in airwayepithelium of asthmatics. Only one study has shown thatpiR30840 directly targets and degrades IL-4 mRNAs lead-ing to inhibition of the development of Th2 T-lymphocytes.Furthermore, the level of piR30840 is significantly reducedin serum from patients with asthma [113].miRNAs are proposed to control expression of 30–60 %of human genes [114] and hence are crucial in mostbiological and pathological processes including cellproliferation, differentiation, apoptosis, carcinogenesisand immune responses [115–117]. miRNAs are 20–24nucleotides long and bind to the 3′ untranslated region(UTR) of target mRNAs resulting in their degradation ortranslational inhibition [118–120]. Currently, there are over1000 miRNAs identified in miRbase (www.mirbase.org),many of which have multiple binding partners and thusaffect multiple pathways [118]. miRNAs may modulate pro-tein synthesis at both initiation and post-initiation of trans-lation [118]. miRNAs have also been shown to up-regulatesome mRNA targets [120]. The balance between up- anddown-regulation of miRNA plays a pivotal role in cell cycleand process of cell proliferation and regeneration, a processthat is dysregulated in asthmatic epithelium. miRNAsare therefore interesting regulatory factors which maycontribute substantially to airway epithelium abnormal-ities in asthmatics.miRNA expression in epithelium of asthmaticsThere are a limited number of studies examining miRNAexpression in epithelium of asthmatics or non-asthmatics(Table 2). miRNA microarray performed on primary bron-chial epithelial cells cultured at air-liquid interface (ALI)showed higher expression of let-7f, miR-181c* and miR-487b but lower expression of miR-203 in mild asthmaticscompared with healthy controls [121]. miR-203 has beenshown to play a potent role in the (keratinocyte) self-renewal program during epidermal differentiation by target-ing p63, facilitating cell cycle exit and promoting differenti-ation [122]. Given that the epithelium of asthmaticsexpresses higher levels of p63 [36], it is intriguing to specu-late that there is a direct link between miR-203 and p63(Fig. 2). Recently, miR-203 has also been shown to inhibitairway smooth cell proliferation through targeting the non-receptor tyrosine kinase c-Abl (Abelson tyrosine kinase,Abl, ABL1) [123]. In particular, Abl kinases regulate cell-cell adhesion in epithelial cells and fibroblasts throughcadherin-mediated adhesion signals via regulating the ac-tivities of the Rac and Rho GTPases [124]. Since, c-Ab1plays important roles in regulation of the actin cytoskel-eton and hence different cellular functions such as prolif-eration, cell adhesion and migration as well as growth anddevelopment [123], the reduced level of miR-203 in epi-thelial cells of asthmatics may promote cell proliferation,increase goblet cell hyperplasia and/or decrease ciliatedcells. Genome wide profiling of bronchial epithelial brush-ings also revealed four members of the miR-34/449 family(miR-34b-5p, miR-34c-5p, miR-449a, and miR-449b-5p)were significantly suppressed in asthma [125]. Interest-ingly, no clear relationship was observed betweenthese differentially expressed miRNA and serum IgElevel in asthmatics [125], which indicates the role ofthese miRNAs at cellular and molecular levels ratherMoheimani et al. Respiratory Research (2016) 17:119 Page 8 of 15than inflammatory and allergic responses. Furthermore,inhaled corticosteroids showed only minor effects onmiRNA expression, and failed to restore miRNA levelsto healthy control levels [125]. The miR-34/449 familyare closely associated with regulation of epithelial cellproliferation and differentiation. Specifically, miR-449is essential in regulation of airway ciliated cells by tar-geting NOTCH1 [126]. Notch signaling triggers airwaymucous metaplasia and inhibits alveolar development(ciliated cells) [127]. The low level of miR-449 in epi-thelial cells of asthmatics may therefore shift the fate ofthese cells toward more mucous production (Fig. 2).Furthermore, miR-34/449-deficient mice suffer fromprimary ciliary dyskinesia (PCD). These abnormalitieshave been shown to be mediated by Cp110, a centriolarprotein suppressing cilia assembly, that is a target ofmiR-34/449 [128]. A panel of miRNAs including miR-18a,miR-27a, miR-128 and miR-155 were also down-regulatedin the epithelium of asthmatics. These miRNAs are in-volved in activation/signaling of IL-6 and IL-8 [129].Recently, miR-19a was reported to be the only miRNAthat differentiates severe from mild asthma. miR-19a isup-regulated in severe asthmatic epithelial cells and isnot restored by corticosteroids [130]. Elevated levels ofmiR-19a further stimulate cell proliferation of epithelialcells by targeting TGF-β receptor 2 mRNA. Overexpres-sion of miR-19a results in reduction in phosphorylatedSMAD3 whereas suppression of miR-19a facilitatesSMAD3 phosphorylation through TGF-β receptor 2signaling and restore epithelial cells proliferation [130].These may suggest the potential of miR-19a as a thera-peutic target in airway epithelium of asthmatics to re-establish epithelial regeneration.There are limited numbers of investigations on miR-NAs expression in asthmatic epithelium of children,possibly due to difficulty in obtaining epithelial samples.Higher level of miR-148b; a member of miR-152 family,was reported in airway epithelial cells of adults with anasthmatic mother and correlates with sHLA-G levels inthe BAL fluid of these subjects [131]. The prenatal effectsof maternal asthma on the regulation of foetal genes inairway cells may persist well into adulthood throughmiRNA regulatory mechanisms.Other miRNAs in airway epithelium with potential role inasthma developmentAirway epithelial cells display dysregulted differentiationin asthma. It is therefore important to investigate po-tential miRNAs fundamental for lung developmentand epithelial cell homeostasis such as miR-17 family[132, 133]. miR-17 family consist of three paralog clustersof miR-17–92 (miR-17-5p, miR-18, miR-19b, miR-20a,miR-92, miR-19a and miR-17-3p), miR-106a–363 (miR-106a, miR-18b, miR-20b, miR-19b-2, miR-92–2, andmiR-363), and miR-106b–25 (miR-106b, miR-93, andmiR-25) [134]. Among the miR-17 family, the miR-17-92cluster and miR-106b have been shown to be essential inmaintaining of the structural homeostasis of developinglung epithelium [132, 134]. miR-17-5p, miR-19b and miR-20 are increased in the epithelium and mesenchyme of theembryonic lung compared with fully developed lung [132].Furthermore, miR-17, miR-20a and miR-106b modulatefibroblast growth factor (FGF)10–FGFR2b signaling byspecifically targeting STAT3 and MAPK14, and alteringE-cadherin distribution. This is vital for epithelial budmorphogenesis in response to FGF10 signaling [134].Fig. 2 Role of miRNAs in airway epithelial cells regeneration. a In healthy airway epithelial cells, miR-449 suppresses NOTCH1 mRNA and encouragesdifferentiation of ciliated cells compared with goblet cells. miR-203 may paly essential role in epithelial cell homeostasis by suppressing p63 which isexpressed in basal cells. b The level of miR-449 and miR-203 are reduced in asthmatic airway epithelial cells, which may result in increase in goblet cellsand compromising epithelial cells regeneration, respectively. Solid lines and bold fonts represent strong effect and dashed lines and normal fontrepresent weak effectMoheimani et al. Respiratory Research (2016) 17:119 Page 9 of 15STAT3 and MAPK14 signalling play important roles inairway homeostasis. STAT3 stimulates regenerationand multiciliogenesis by inhibition of the Notch path-way and direct regulation of genes such as Mcidas andFoxj1 [135]. MAPK14 (known also as p38α) has alsobeen shown to regulate lung stem or progenitor cellproliferation and differentiation [136]. MAPK14 regu-lates C/EBP and HNF3b which are necessary for thedifferentiation of the stem cells into AT2 and Claracells, while coordinately suppressing the regulators ofstem and progenitor cells proliferation; cyclin D1 andEGFR [136]. These findings emphasise the role of thesesignalling pathways as well as their regulatory miRNAs inairway epithelial cells homeostasis, which is dysregulatedin asthma. These pathways may initiate development andprogression of asthma.ConclusionsOverall, studies on asthma susceptibility genes (Table 1)and epigenetic regulatory mechanisms (Table 2) of air-way epithelial cells provide important insights in the de-velopment and progression of asthma (Fig. 3).Among susceptibility genes detected by positionalcloning (Table 1), IRAKM may represent a potential bio-marker for early onset of asthma [26] whereas PCDH1may be a potential biomarker in both children andadults [22]. Although ADAM33 protein increases in epi-thelium of asthmatics, it is not related to severity of dis-ease [18]. None of the genes listed above can predictspecific endotypes of asthma. Some genes detected byGWAS (Table 1) have shown the potential as asthmabiomarkers in a specific age group, e.g. ORMDL3/GSDMBin children [60]. However, there is little overlap betweenasthma susceptibility genes and their products detectedwith positional cloning and GWAS. Epithelial eQTL coulddetect more specific biomarkers for different phenotype ofasthma, such as CDHR3 associated with asthma in chil-dren with severe exacerbation [78, 85], and CST1 [78],which can differentiate asthmatic with EIB from thosewith no EIB [84].Asthma is a complex disease and it is unlikely thatlimited functional genes are driving the entire patho-physiological (immunological and structural) events.Other challenges for genetic assessments are neglectingFig. 3 Overview of the key genes and epigenetic regulatory mechanisms associated with asthma in airway epithelial cells. Environmental insults(e.g. allergens or viruses) may damage the integrity of airway epithelial cells. Some of the susceptibility genes expressed in the airway epithelialcells (e.g. ADAM33) may further deteriorate this structural damage through the process of epithelial–mesenchymal trophic unit (EMTU) and henceencouraging airway remodelling and hyper-responsiveness. Whereas other asthma susceptibility genes (e.g. IRAKM) may promote (Th2)-immunityresult in activation of inflammatory responses. Some of the genes have more protective roles (e.g. SPINK5 and TSLP). Furthermore, epigeneticregulatory mechanisms may affect some of these genes (e.g. hyper-methylation of ADAM33, and HLA-G suppression by miRNAs)Moheimani et al. Respiratory Research (2016) 17:119 Page 10 of 15information for rare variants in some populations, clin-ical heterogeneity of asthma and the effects of variousimportant environmental factors including smoking, airpollution, and microbial exposures. Recent studies onepigenetic regulatory factors (Table 2) have added newinsights to the field. Some of the DNA methylation sig-natures in epithelial cells of asthmatics may be involvedin epithelial homeostasis (KRT5 and STAT5A), BHR(ADAM33) and FeNO regulation (ARG2, IL-6 and iNOS)[98–101]. Notably, ADAM33 is also a target of methylation(Fig. 3) [101], which emphasises the importance of con-comitant assessment genetic and epigenetic regulatory fac-tors in airway epithelium of asthmatics. More profoundly,miRNA networks have been shown to regulate a major por-tion of post-transcriptional gene regulation [130].There are however some challenges in interpretationof miRNAs findings from different studies. One import-ant factor is the origin of samples; for example bronchialbiopsies [137] compared with cultured primary epithelialcells [121] or bronchial epithelial brushing that may in-clude other cells which may affect yield and type of de-tectable miRNAs [125]. Severity of disease [137] andvarying technologies and methods of analysis may alsoplay essential roles in outcomes of miRNAs quantifica-tions. The most common technology used is microarraywhich may ignore less abundant miRNAs and may notdistinguish miRNAs from other RNAs with similar se-quences, such as other members of the same miRNAfamily [125]. quantitative PCR is an alternative, whichdoes not necessarily measure all potentially biologicallyimportant miRNAs. Additionally, the method to analyseand present data; ΔΔCT [121, 129, 130] versus 2-ΔΔCT[137], can affect the outcomes. One potential way toovercome these inconsistencies is assessing miRNA ex-pression in primary airway epithelial cells at air liquidinterface culture; a mimic of pseudostratified physio-logical model, using technology such as NanoString thatcounts individual miRNA with high accuracy.Future prospects of miRNA researchWhile a number of miRNAs have been associated withabnormalities in asthmatic epithelium and disease pro-gression, current asthma treatments (e.g. corticosteroids)show no major effect on them [125, 130, 137]. Seeking anovel approach to target abnormally expressed miRNAsand hence restoring their normal functions may providea novel asthma intervention strategy. Specific targetingof miRNA clusters (e.g. miR-17-92 cluster with proposedproliferative roles) may restore normal epithelial homeo-stasis although off-target effects should to be carefullyevaluated. Further studies to identify specific miRNA-mRNA interactions and validation of target proteinsimportant in asthma pathogenesis may provide potentialsteps forward to providing important insights into thedevelopment of potential intervention to reverse the epithe-lial abnormities in asthmatics. Additionally, manipulatingepigenetic factors regulating miRNAs (e.g. re-expression ofmiRNAs using demethylating agents to inhibit DNAmethylation of the miRNA promoter) may provide anotherapproach to restore miRNA abnormalities in asthmatics. Itis also important to clarify whether these differences inmiRNAs are the major factors driving asthma or if thepathology of the disease induces these changes. Hence, add-itional studies in larger cohorts are essential to distinguishthe effects of different asthma medications on the expres-sion of miRNAs in bronchial epithelial cells.Overall, there are interactions between genetic factorsand epigenetic regulatory mechanisms and assessmentof only one factor may not provide enough information.miRNAs expression in conjunction with other epigeneticregulatory factors may be an essential contributing factorto asthma. Understanding the mechanisms that initiatethe development and progression of asthma, includingregulation of gene transcription or translation, are essen-tial to identify potential targets in airway epithelium forasthma intervention.Abbreviations5mC: 5-methyl-cytosine; ADAM33: A disintegrin and metalloprotease 33;ALI: Air-liquid interface; ARG2: Arginase 2; BHR: Bronchial hyperresponsiveness;CDHR3: Cadherin-related family member 3; CDK12: Cyclin-dependent kinase 12;CNVs: Copy number variants; CRIP1: Cysteine-rich protein 1; CST1: Cystatin SN;DNMT: DNA methyltransferase; DPP10: Dipeptidyl-peptidase 10; dsRNA: Double-stranded RNA; ECM: Extracellular matrix; EIB: Exercise-induced bronchoconstriction;EMT: Epithelial–mesenchymal transition; EMTU: Epithelial–mesenchymal trophicunit; eQTLs: Expression quantitative trait loci; FeNO: Fractional exhalation of nitricoxide; FGF: Fibroblast growth factor; GSDMB: Gasdermin B; GSTM1: Glutathione-S-transferase M1; GWAS: Genome-wide association studies; H3K18: Histone H3 lysine18; H3K9me3: Histone H3 lysine 9 trimethylation; HAT: Histone acetyltransferase;HDAC: Histone deacetylase; HDM: Histone demethylase; HMT: Histonemethyltransferase; IFN: Interferon; IL18R1: IL18 receptor 1; IL1RL1: Interleukin (IL) 1receptor-like 1; iNOS: inducible nitric oxide synthase; IRAKM: IL-1 receptorassociated kinase-M; KRT5: Cytokeratin 5; LEKTI: Lympho-epithelial Kazal-type-related inhibitor; lncRNA: Long non-coding RNA; MHC: Major histocompatibility;miRNAs: microRNAs; NK: Natural killer; NPSR1: Neuropeptide S Receptor 1;ORMDL3: ORM1-like 3; PCDH1: Protocadherin-1; piRNA: Piwi-interacting RNA;PM10: Particulate matter with a diameter of <10 μm diameter; PPP1R1B: Proteinphosphatase 1 regulatory subunit 1B; SNAI1: SMAD3-dependent transcriptionfactor snail1; SPINK5: Serine protease inhibitor Kazal type-5; STARD3: StAR-relatedlipid transfer (START) Domani containing 3; START: StAR-related lipid transfer;STAT5A: Signal transducer and activator of transcription 5A; TCAP: Titin-cap;TGF: Transforming growth factor; Th: T-helper; TLR: Toll like receptor;TSLP: Thymic stromal lymphopoietin; UPR: Unfolded protein response;WGS: Whole genome sequencingAcknowledgementsThe authors acknowledge the research funding from the Early CareerResearcher (ECR) Grant and New Staff Grant to FM, the University ofNewcastle; McDonald Jones Homes Group Philanthropy Grant - HMRI to FMand DK, and the National Health and Medical Research Council (NHMRC: APP1064405), Australia to DK.FundingThe Early Career Researcher (ECR) Grant and New Staff Grant to FM, theUniversity of Newcastle; McDonald Jones Homes Group PhilanthropyGrant - HMRI to FM and DK; and the National Health and MedicalResearch Council (NHMRC: APP 1064405), Australia to DK.Moheimani et al. Respiratory Research (2016) 17:119 Page 11 of 15Availability of data and materialNot applicable.Authors’ contributionsFM and DK designed the concept and organized the review. FM, AH, AR andTW participated in drafting the manuscript. AK, SS, PH, PW and DK criticallyevaluated and improved the manuscript. All authors read and approved thefinal manuscript.Competing interestsThe authors declare that they have no competing interests.Consent for publicationNot applicable.Ethics approval and consent to participateNot applicable.Author details1School of Biomedical Sciences and Pharmacy, Faculty of Health andMedicine, HMRI building, The University of Newcastle, Callaghan, NSW 2308,Australia. 2Priority Research Centre for Healthy Lungs, Hunter MedicalResearch Institute, The University of Newcastle, New South Wales, Australia.3Department of Biochemistry and Microbiology, University of Victoria,Victoria, Canada. 4Telethon Kids Institute, Centre for Health Research, TheUniversity of Western Australia, Nedlands 6009, Western Australia, Australia.5Department of Respiratory Medicine, Princess Margaret Hospital forChildren, Perth 6001, Western Australia, Australia. 6School of Paediatrics andChild Health, The University of Western Australia, Nedlands 6009, WesternAustralia, Australia. 7Centre for Cell Therapy and Regenerative Medicine,School of Medicine and Pharmacology, The University of Western Australia,Nedlands 6009, Western Australia, Australia. 8Department of Respiratory andSleep Medicine, John Hunter Hospital, New South Wales, Australia.9Department of Anesthesiology, Pharmacology and Therapeutics, Universityof British Columbia, Vancouver, Canada.Received: 22 June 2016 Accepted: 17 September 2016References1. Fanta CH. Asthma. N Engl J Med. 2009;360(10):1002–14.2. Roth HM, Wadsworth SJ, Kahn M, Knight DA. The airway epithelium inasthma: developmental issues that scar the airways for life? Pulm PharmacolTher. 2012;25(6):420–6.3. Hirota JA, Knight DA. Human airway epithelial cell innate immunity:relevance to asthma. Curr Opin Immunol. 2012;24(6):740–6.4. Hackett TL, Singhera GK, Shaheen F, Hayden P, Jackson GR, Hegele RG, VanEeden S, Bai TR, Dorscheid DR, Knight DA. Intrinsic phenotypic differencesof asthmatic epithelium and its inflammatory responses to respiratorysyncytial virus and air pollution. Am J Respir Cell Mol Biol. 2011;45(5):1090–100.5. Kicic A, Sutanto EN, Stevens PT, Knight DA, Stick SM. Intrinsic biochemicaland functional differences in bronchial epithelial cells of children withasthma. Am J Respir Crit Care Med. 2006;174(10):1110–8.6. Warner SM, Hackett TL, Shaheen F, Hallstrand TS, Kicic A, Stick SM, Knight DA.Transcription factor p63 regulates key genes and wound repair in humanairway epithelial basal cells. Am J Respir Cell Mol Biol. 2013;49(6):978–88.7. Freishtat RJ, Watson AM, Benton AS, Iqbal SF, Pillai DK, Rose MC, HoffmanEP. Asthmatic airway epithelium is intrinsically inflammatory and mitoticallydyssynchronous. Am J Respir Cell Mol Biol. 2011;44(6):863–9.8. Wark PA, Johnston SL, Bucchieri F, Powell R, Puddicombe S, Laza-Stanca V,Holgate ST, Davies DE. Asthmatic bronchial epithelial cells have a deficientinnate immune response to infection with rhinovirus. J Exp Med. 2005;201(6):937–47.9. Wark PA, Grissell T, Davies B, See H, Gibson PG. Diversity in the bronchialepithelial cell response to infection with different rhinovirus strains.Respirology. 2009;14(2):180–6.10. Parsons KS, Hsu AC, Wark PA. TLR3 and MDA5 signalling, although notexpression, is impaired in asthmatic epithelial cells in response to rhinovirusinfection. Clin Exp Allergy. 2014;44(1):91–101.11. Stevens PT, Kicic A, Sutanto EN, Knight DA, Stick SM. Dysregulated repair inasthmatic paediatric airway epithelial cells: the role of plasminogen activatorinhibitor-1. Clin Exp Allergy. 2008;38(12):1901–10.12. Kicic A, Hallstrand TS, Sutanto EN, Stevens PT, Kobor MS, Taplin C, Pare PD,Beyer RP, Stick SM, Knight DA. Decreased fibronectin productionsignificantly contributes to dysregulated repair of asthmatic epithelium. AmJ Respir Crit Care Med. 2010;181(9):889–98.13. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, vonMutius E, Farrall M, Lathrop M, Cookson WO. A large-scale, consortium-basedgenomewide association study of asthma. N Engl J Med. 2010;363(13):1211–21.14. Akhabir L, Sandford AJ. Genome-wide association studies for discovery ofgenes involved in asthma. Respirology. 2011;16(3):396–406.15. Campbell CD, Mohajeri K, Malig M, Hormozdiari F, Nelson B, Du G, PattersonKM, Eng C, Torgerson DG, Hu D, Herman C, Chong JX, Ko A, O’Roak BJ,Krumm N, Vives L, Lee C, Roth LA, Rodriguez-Cintron W, Rodriguez-Santana J,Brigino-Buenaventura E, Davis A, Meade K, LeNoir MA, Thyne S, Jackson DJ,Gern JE, Lemanske Jr RF, Shendure J, Abney M, Burchard EG, Ober C, Eichler EE.Whole-genome sequencing of individuals from a founder population identifiescandidate genes for asthma. PLoS One. 2014;9(8):e104396.16. Knight D, Hirota JA. Airway epithelial cells. In: Adkinson Jr NF, Bochner BS,Burks AW, Busse WW, Holgate ST, editors. Adkinson: Middleton’s allergy:principles and practice. 8th ed. An Imprint of Elsevier: Saunders; 2013. p. 302–12.17. Van Eerdewegh P, Little RD, Dupuis J, Del Mastro RG, Falls K, Simon J, Torrey D,Pandit S, McKenny J, Braunschweiger K, Walsh A, Liu Z, Hayward B, Folz C,Manning SP, Bawa A, Saracino L, Thackston M, Benchekroun Y, Capparell N,Wang M, Adair R, Feng Y, Dubois J, FitzGerald MG, Huang H, Gibson R, AllenKM, Pedan A, Danzig MR, Umland SP, Egan RW, Cuss FM, Rorke S, Clough JB,Holloway JW, Holgate ST, Keith TP. Association of the ADAM33 gene withasthma and bronchial hyperresponsiveness. Nature. 2002;418(6896):426–30.18. Tripathi P, Awasthi S, Husain N, Prasad R, Mishra V. Increased expression ofADAM33 protein in asthmatic patients as compared to non-asthmaticcontrols. Indian J Med Res. 2013;137(3):507–14.19. Laitinen T, Polvi A, Rydman P, Vendelin J, Pulkkinen V, Salmikangas P, MakelaS, Rehn M, Pirskanen A, Rautanen A, Zucchelli M, Gullsten H, Leino M, AleniusH, Petays T, Haahtela T, Laitinen A, Laprise C, Hudson TJ, Laitinen LA, Kere J.Characterization of a common susceptibility locus for asthma-related traits.Science. 2004;304(5668):300–4.20. Vendelin J, Pulkkinen V, Rehn M, Pirskanen A, Raisanen-Sokolowski A,Laitinen A, Laitinen LA, Kere J, Laitinen T. Characterization of GPRA, a novelG protein-coupled receptor related to asthma. Am J Respir Cell Mol Biol.2005;33(3):262–70.21. Pietras CO, Vendelin J, Anedda F, Bruce S, Adner M, Sundman L, Pulkkinen V,Alenius H, D’Amato M, Soderhall C, Kere J. The asthma candidate gene NPSR1mediates isoform specific downstream signalling. BMC Pulm Med. 2011;11:39.22. Koppelman GH, Meyers DA, Howard TD, Zheng SL, Hawkins GA, AmplefordEJ, Xu J, Koning H, Bruinenberg M, Nolte IM, van Diemen CC, Boezen HM,Timens W, Whittaker PA, Stine OC, Barton SJ, Holloway JW, Holgate ST,Graves PE, Martinez FD, van Oosterhout AJ, Bleecker ER, Postma DS.Identification of PCDH1 as a novel susceptibility gene for bronchialhyperresponsiveness. Am J Respir Crit Care Med. 2009;180(10):929–35.23. Koning H, Sayers I, Stewart CE, de Jong D, Ten Hacken NH, Postma DS, vanOosterhout AJ, Nawijn MC, Koppelman GH. Characterization ofprotocadherin-1 expression in primary bronchial epithelial cells: associationwith epithelial cell differentiation. FASEB J. 2012;26(1):439–48.24. Faura Tellez G, Nawijn MC, Koppelman GH. Protocadherin-1: epithelialbarrier dysfunction in asthma and eczema. Eur Respir J. 2014;43(3):671–4.25. Meng JF, Rosenwasser LJ. Unraveling the genetic basis of asthma andallergic diseases. Allergy, Asthma Immunol Res. 2010;2(4):215–27.26. Balaci L, Spada MC, Olla N, Sole G, Loddo L, Anedda F, Naitza S, ZunchedduMA, Maschio A, Altea D, Uda M, Pilia S, Sanna S, Masala M, Crisponi L, FattoriM, Devoto M, Doratiotto S, Rassu S, Mereu S, Giua E, Cadeddu NG, Atzeni R,Pelosi U, Corrias A, Perra R, Torrazza PL, Pirina P, Ginesu F, Marcias S, SchintuMG, Del Giacco GS, Manconi PE, Malerba G, Bisognin A, Trabetti E, Boner A,Pescollderungg L, Pignatti PF, Schlessinger D, Cao A, Pilia G. IRAK-M isinvolved in the pathogenesis of early-onset persistent asthma. Am J HumGenet. 2007;80(6):1103–14.27. Adcock IM, Kirkham PA. PHF11 and DPP10: a tale of two genes in asthma.Respiration. 2010;79(1):14–6.28. Qi SY, Riviere PJ, Trojnar J, Junien JL, Akinsanya KO. Cloning andcharacterization of dipeptidyl peptidase 10, a new member of an emergingsubgroup of serine proteases. Biochem J. 2003;373(Pt 1):179–89.Moheimani et al. Respiratory Research (2016) 17:119 Page 12 of 1529. Zhou H, Hong X, Jiang S, Dong H, Xu X, Xu X. Analyses of associationsbetween three positionally cloned asthma candidate genes and asthmaor asthma-related phenotypes in a Chinese population. BMC Med Genet.2009;10:123.30. Nicolae D, Cox NJ, Lester LA, Schneider D, Tan Z, Billstrand C, Kuldanek S,Donfack J, Kogut P, Patel NM, Goodenbour J, Howard T, Wolf R, KoppelmanGH, White SR, Parry R, Postma DS, Meyers D, Bleecker ER, Hunt JS, Solway J,Ober C. Fine mapping and positional candidate studies identify HLA-G asan asthma susceptibility gene on chromosome 6p21. Am J Hum Genet.2005;76(2):349–57.31. Tan Z, Randall G, Fan J, Camoretti-Mercado B, Brockman-Schneider R, Pan L,Solway J, Gern JE, Lemanske RF, Nicolae D, Ober C. Allele-specific targeting ofmicroRNAs to HLA-G and risk of asthma. Am J Hum Genet. 2007;81(4):829–34.32. Cakebread JA, Haitchi HM, Holloway JW, Powell RM, Keith T, Davies DE,Holgate ST. The role of ADAM33 in the pathogenesis of asthma. SpringerSemin Immunopathol. 2004;25(3–4):361–75.33. Bezerra GA, Dobrovetsky E, Seitova A, Fedosyuk S, Dhe-Paganon S, Gruber K.Structure of human dipeptidyl peptidase 10 (DPPY): a modulator ofneuronal Kv4 channels. Sci Rep. 2015;5:8769.34. Kocmalova M, Oravec M, Adamkov M, Sadlonova V, Kazimierova I,Medvedova I, Joskova M, Franova S, Sutovska M. Potassium ion channelsand allergic asthma. Adv Exp Med Biol. 2015;838:35–45.35. Gros F, Cabillic F, Toutirais O, Maux AL, Sebti Y, Amiot L. Soluble HLA-Gmolecules impair natural killer/dendritic cell crosstalk via inhibition ofdendritic cells. Eur J Immunol. 2008;38(3):742–9.36. Hackett TL, Warner SM, Stefanowicz D, Shaheen F, Pechkovsky DV, MurrayLA, Argentieri R, Kicic A, Stick SM, Bai TR, Knight DA. Induction of epithelial-mesenchymal transition in primary airway epithelial cells from patients withasthma by transforming growth factor-beta1. Am J Respir Crit Care Med.2009;180(2):122–33.37. Mitsudo K, Jayakumar A, Henderson Y, Frederick MJ, Kang Y, Wang M,El-Naggar AK, Clayman GL. Inhibition of serine proteinases plasmin, trypsin,subtilisin A, cathepsin G, and elastase by LEKTI: a kinetic analysis.Biochemistry. 2003;42(13):3874–81.38. Chavanas S, Bodemer C, Rochat A, Hamel-Teillac D, Ali M, Irvine AD, Bonafe JL,Wilkinson J, Taieb A, Barrandon Y, Harper JI, de Prost Y, Hovnanian A. Mutationsin SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome.Nat Genet. 2000;25(2):141–2.39. Deraison C, Bonnart C, Lopez F, Besson C, Robinson R, Jayakumar A,Wagberg F, Brattsand M, Hachem JP, Leonardsson G, Hovnanian A. LEKTIfragments specifically inhibit KLK5, KLK7, and KLK14 and control desquamationthrough a pH-dependent interaction. Mol Biol Cell. 2007;18(9):3607–19.40. Slager RE, Hawkins GA, Li X, Postma DS, Meyers DA, Bleecker ER. Genetics ofasthma susceptibility and severity. Clin Chest Med. 2012;33(3):431–43.41. Zhao YF, Luo YM, Xiong W, Wu XL. Genetic variation in ORMDL3 genemay contribute to the risk of asthma: a meta-analysis. Hum Immunol.2014;75(9):960–7.42. Hui CC, Yu A, Heroux D, Akhabir L, Sandford AJ, Neighbour H, Denburg JA.Thymic stromal lymphopoietin (TSLP) secretion from human nasal epitheliumis a function of TSLP genotype. Mucosal Immunol. 2015;8(5):993–9.43. Hirota T, Takahashi A, Kubo M, Tsunoda T, Tomita K, Doi S, Fujita K, Miyatake A,Enomoto T, Miyagawa T, Adachi M, Tanaka H, Niimi A, Matsumoto H, Ito I,Masuko H, Sakamoto T, Hizawa N, Taniguchi M, Lima JJ, Irvin CG, Peters SP,Himes BE, Litonjua AA, Tantisira KG, Weiss ST, Kamatani N, Nakamura Y, TamariM. Genome-wide association study identifies three new susceptibility loci foradult asthma in the Japanese population. Nat Genet. 2011;43(9):893–6.44. Imaoka H, Gauvreau GM, Watson RM, Smith SG, Dua B, Baatjes AJ, Howie K,Hoshino T, Killian KJ, Aizawa H, O’Byrne PM. Interleukin-18 and interleukin-18receptor-alpha expression in allergic asthma. Eur Respir J. 2011;38(4):981–3.45. Akhabir L, Sandford A. Genetics of interleukin 1 receptor-like 1 in immuneand inflammatory diseases. Curr Genomics. 2010;11(8):591–606.46. Grotenboer NS, Ketelaar ME, Koppelman GH, Nawijn MC. Decoding asthma:translating genetic variation in IL33 and IL1RL1 into diseasepathophysiology. J Allergy Clin Immunol. 2013;131(3):856–65.47. Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P,Jonsdottir GM, Thorleifsson G, Helgadottir H, Steinthorsdottir V, StefanssonH, Williams C, Hui J, Beilby J, Warrington NM, James A, Palmer LJ,Koppelman GH, Heinzmann A, Krueger M, Boezen HM, Wheatley A,Altmuller J, Shin HD, Uh ST, Cheong HS, Jonsdottir B, Gislason D, Park CS,Rasmussen LM, Porsbjerg C, Hansen JW, Backer V, Werge T, Janson C,Jonsson UB, Ng MC, Chan J, So WY, Ma R, Shah SH, Granger CB, QuyyumiAA, Levey AI, Vaccarino V, Reilly MP, Rader DJ, Williams MJ, van Rij AM,Jones GT, Trabetti E, Malerba G, Pignatti PF, Boner A, Pescollderungg L,Girelli D, Olivieri O, Martinelli N, Ludviksson BR, Ludviksdottir D, Eyjolfsson GI,Arnar D, Thorgeirsson G, Deichmann K, Thompson PJ, Wjst M, Hall IP,Postma DS, Gislason T, Gulcher J, Kong A, Jonsdottir I, Thorsteinsdottir U,Stefansson K. Sequence variants affecting eosinophil numbers associatewith asthma and myocardial infarction. Nat Genet. 2009;41(3):342–7.48. Prefontaine D, Nadigel J, Chouiali F, Audusseau S, Semlali A, Chakir J, MartinJG, Hamid Q. Increased IL-33 expression by epithelial cells in bronchialasthma. J Allergy Clin Immunol. 2010;125(3):752–4.49. Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, ZurawskiG, Moshrefi M, Qin J, Li X, Gorman DM, Bazan JF, Kastelein RA. IL-33, aninterleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2and induces T helper type 2-associated cytokines. Immunity. 2005;23(5):479–90.50. Moussion C, Ortega N, Girard JP. The IL-1-like cytokine IL-33 is constitutivelyexpressed in the nucleus of endothelial cells and epithelial cells in vivo: anovel ‘alarmin’? PLoS One. 2008;3(10), e3331.51. Moffatt MF, Schou C, Faux JA, Abecasis GR, James A, Musk AW, CooksonWO. Association between quantitative traits underlying asthma and theHLA-DRB1 locus in a family-based population sample. Eur J Hum Genet.2001;9(5):341–6.52. Andreatta M, Nielsen M. Characterizing the binding motifs of 11 commonhuman HLA-DP and HLA-DQ molecules using NNAlign. Immunology.2012;136(3):306–11.53. Kalb TH, Chuang MT, Marom Z, Mayer L. Evidence for accessory cellfunction by class II MHC antigen-expressing airway epithelial cells. Am JRespir Cell Mol Biol. 1991;4(4):320–9.54. Chapoval SP, Nabozny GH, Marietta EV, Raymond EL, Krco CJ, Andrews AG,David CS. Short ragweed allergen induces eosinophilic lung disease inHLA-DQ transgenic mice. J Clin Invest. 1999;103(12):1707–17.55. Anthoni M, Wang G, Leino MS, Lauerma AI, Alenius HT, Wolff HJ. Smad3-signalling and Th2 cytokines in normal mouse airways and in a mousemodel of asthma. Int J Biol Sci. 2007;3(7):477–85.56. Moheimani F, Roth HM, Cross J, Reid AT, Shaheen F, Warner SM, Hirota JA,Kicic A, Hallstrand TS, Kahn M, Stick SM, Hansbro PM, Hackett TL, Knight DA.Disruption of beta-catenin/CBP signaling inhibits human airway epithelial-mesenchymal transition and repair. Int J Biochem Cell Biol. 2015;68:59–69.57. Gibbs JD, Ornoff DM, Igo HA, Zeng JY, Imani F. Cell cycle arrest bytransforming growth factor beta1 enhances replication of respiratorysyncytial virus in lung epithelial cells. J Virol. 2009;83(23):12424–31.58. McCann KL, Imani F. Transforming growth factor beta enhances respiratorysyncytial virus replication and tumor necrosis factor alpha induction inhuman epithelial cells. J Virol. 2007;81(6):2880–6.59. Bedke N, Sammut D, Green B, Kehagia V, Dennison P, Jenkins G, Tatler A,Howarth PH, Holgate ST, Davies DE. Transforming growth factor-betapromotes rhinovirus replication in bronchial epithelial cells by suppressingthe innate immune response. PLoS One. 2012;7(9), e44580.60. Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, Depner M,von Berg A, Bufe A, Rietschel E, Heinzmann A, Simma B, Frischer T, Willis-Owen SA, Wong KC, Illig T, Vogelberg C, Weiland SK, von Mutius E, AbecasisGR, Farrall M, Gut IG, Lathrop GM, Cookson WO. Genetic variants regulatingORMDL3 expression contribute to the risk of childhood asthma. Nature.2007;448(7152):470–3.61. Miller M, Tam AB, Cho JY, Doherty TA, Pham A, Khorram N, Rosenthal P,Mueller JL, Hoffman HM, Suzukawa M, Niwa M, Broide DH. ORMDL3 is aninducible lung epithelial gene regulating metalloproteases, chemokines,OAS, and ATF6. Proc Natl Acad Sci U S A. 2012;109(41):16648–53.62. Zehethofer N, Bermbach S, Hagner S, Garn H, Muller J, Goldmann T, LindnerB, Schwudke D, Konig P. Lipid analysis of airway epithelial cells for studyingrespiratory diseases. Chromatographia. 2015;78(5–6):403–13.63. Breslow DK, Collins SR, Bodenmiller B, Aebersold R, Simons K, ShevchenkoA, Ejsing CS, Weissman JS. Orm family proteins mediate sphingolipidhomeostasis. Nature. 2010;463(7284):1048–53.64. Maceyka M, Spiegel S. Sphingolipid metabolites in inflammatory disease.Nature. 2014;510(7503):58–67.65. Nixon GF. Sphingolipids in inflammation: pathological implications andpotential therapeutic targets. Br J Pharmacol. 2009;158(4):982–93.66. Levy BD. Sphingolipids and susceptibility to asthma. N Engl J Med. 2013;369(10):976–8.67. Verlaan DJ, Berlivet S, Hunninghake GM, Madore AM, Lariviere M, MoussetteS, Grundberg E, Kwan T, Ouimet M, Ge B, Hoberman R, Swiatek M, Dias J,Moheimani et al. Respiratory Research (2016) 17:119 Page 13 of 15Lam KC, Koka V, Harmsen E, Soto-Quiros M, Avila L, Celedon JC, Weiss ST,Dewar K, Sinnett D, Laprise C, Raby BA, Pastinen T, Naumova AK. Allele-specific chromatin remodeling in the ZPBP2/GSDMB/ORMDL3 locusassociated with the risk of asthma and autoimmune disease. Am J HumGenet. 2009;85(3):377–93.68. Zhao CN, Fan Y, Huang JJ, Zhang HX, Gao T, Wang C, Wang T, Hou LF. Theassociation of GSDMB and ORMDL3 gene polymorphisms with asthma: ameta-analysis. Allergy, Asthma Immunol Res. 2015;7(2):175–85.69. Giovannini-Chami L, Marcet B, Moreilhon C, Chevalier B, Illie MI, Lebrigand K,Robbe-Sermesant K, Bourrier T, Michiels JF, Mari B, Crenesse D, Hofman P,de Blic J, Castillo L, Albertini M, Barbry P. Distinct epithelial gene expressionphenotypes in childhood respiratory allergy. Eur Respir J. 2012;39(5):1197–205.70. Genomes Project C, Abecasis GR, Altshuler D, Auton A, Brooks LD, DurbinRM, Gibbs RA, Hurles ME, McVean GA. A map of human genome variationfrom population-scale sequencing. Nature. 2010;467(7319):1061–73.71. Torgerson DG, Ampleford EJ, Chiu GY, Gauderman WJ, Gignoux CR, GravesPE, Himes BE, Levin AM, Mathias RA, Hancock DB, Baurley JW, Eng C, SternDA, Celedon JC, Rafaels N, Capurso D, Conti DV, Roth LA, Soto-Quiros M,Togias A, Li X, Myers RA, Romieu I, Van Den Berg DJ, Hu D, Hansel NN,Hernandez RD, Israel E, Salam MT, Galanter J, Avila PC, Avila L, Rodriquez-Santana JR, Chapela R, Rodriguez-Cintron W, Diette GB, Adkinson NF, AbelRA, Ross KD, Shi M, Faruque MU, Dunston GM, Watson HR, Mantese VJ,Ezurum SC, Liang L, Ruczinski I, Ford JG, Huntsman S, Chung KF, Vora H, LiX, Calhoun WJ, Castro M, Sienra-Monge JJ, del Rio-Navarro B, DeichmannKA, Heinzmann A, Wenzel SE, Busse WW, Gern JE, Lemanske Jr RF, Beaty TH,Bleecker ER, Raby BA, Meyers DA, London SJ, Mexico City ChildhoodAsthma S, Gilliland FD, Children’s Health S, study H, Burchard EG, Geneticsof Asthma in Latino Americans Study SoG-E, Admixture in Latino A, Studyof African Americans AG, Environments, Martinez FD, Childhood Asthma R,Education N, Weiss ST, Childhood Asthma Management P, Williams LK,Study of Asthma P, Pharmacogenomic Interactions by R-E, Barnes KC,Genetic Research on Asthma in African Diaspora S, Ober C, Nicolae DL.Meta-analysis of genome-wide association studies of asthma in ethnicallydiverse North American populations. Nat Genet. 2011;43(9):887–92.72. He JQ, Hallstrand TS, Knight D, Chan-Yeung M, Sandford A, Tripp B, ZamarD, Bosse Y, Kozyrskyj AL, James A, Laprise C, Daley D. A thymic stromallymphopoietin gene variant is associated with asthma and airwayhyperresponsiveness. J Allergy Clin Immunol. 2009;124(2):222–9.73. Ying S, O’Connor B, Ratoff J, Meng Q, Mallett K, Cousins D, Robinson D,Zhang G, Zhao J, Lee TH, Corrigan C. Thymic stromal lymphopoietin expression isincreased in asthmatic airways and correlates with expression of Th2-attractingchemokines and disease severity. J Immunol. 2005;174(12):8183–90.74. Semlali A, Jacques E, Koussih L, Gounni AS, Chakir J. Thymic stromallymphopoietin-induced human asthmatic airway epithelial cellproliferation through an IL-13-dependent pathway. J Allergy ClinImmunol. 2010;125(4):844–50.75. Ziegler SF. Thymic stromal lymphopoietin and allergic disease. J Allergy ClinImmunol. 2012;130(4):845–52.76. Wjst M, Sargurupremraj M, Arnold M. Genome-wide association studies inasthma: what they really told us about pathogenesis. Curr Opin Allergy ClinImmunol. 2013;13(1):112–8.77. Ober C, Yao TC. The genetics of asthma and allergic disease: a 21st centuryperspective. Immunol Rev. 2011;242(1):10–30.78. Luo W, Obeidat M, Di Narzo AF, Chen R, Sin DD, Pare PD, Hao K. Airwayepithelial expression quantitative trait loci reveal genes underlying asthmaand other airway diseases. Am J Respir Cell Mol Biol. 2016;54(2):177–87.79. Emilsson V, Thorleifsson G, Zhang B, Leonardson AS, Zink F, Zhu J, Carlson S,Helgason A, Walters GB, Gunnarsdottir S, Mouy M, Steinthorsdottir V,Eiriksdottir GH, Bjornsdottir G, Reynisdottir I, Gudbjartsson D, Helgadottir A,Jonasdottir A, Jonasdottir A, Styrkarsdottir U, Gretarsdottir S, Magnusson KP,Stefansson H, Fossdal R, Kristjansson K, Gislason HG, Stefansson T, LeifssonBG, Thorsteinsdottir U, Lamb JR, Gulcher JR, Reitman ML, Kong A, Schadt EE,Stefansson K. Genetics of gene expression and its effect on disease. Nature.2008;452(7186):423–8.80. Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, KosekiM, Pirruccello JP, Ripatti S, Chasman DI, Willer CJ, Johansen CT, Fouchier SW,Isaacs A, Peloso GM, Barbalic M, Ricketts SL, Bis JC, Aulchenko YS,Thorleifsson G, Feitosa MF, Chambers J, Orho-Melander M, Melander O,Johnson T, Li X, Guo X, Li M, Shin Cho Y, Jin Go M, Jin Kim Y, Lee JY, Park T,Kim K, Sim X, Twee-Hee Ong R, Croteau-Chonka DC, Lange LA, Smith JD,Song K, Hua Zhao J, Yuan X, Luan J, Lamina C, Ziegler A, Zhang W, Zee RY,Wright AF, Witteman JC, Wilson JF, Willemsen G, Wichmann HE, WhitfieldJB, Waterworth DM, Wareham NJ, Waeber G, Vollenweider P, Voight BF,Vitart V, Uitterlinden AG, Uda M, Tuomilehto J, Thompson JR, Tanaka T,Surakka I, Stringham HM, Spector TD, Soranzo N, Smit JH, Sinisalo J, SilanderK, Sijbrands EJ, Scuteri A, Scott J, Schlessinger D, Sanna S, Salomaa V,Saharinen J, Sabatti C, Ruokonen A, Rudan I, Rose LM, Roberts R, Rieder M,Psaty BM, Pramstaller PP, Pichler I, Perola M, Penninx BW, Pedersen NL,Pattaro C, Parker AN, Pare G, Oostra BA, O’Donnell CJ, Nieminen MS,Nickerson DA, Montgomery GW, Meitinger T, McPherson R, McCarthy MI,McArdle W, Masson D, Martin NG, Marroni F, Mangino M, Magnusson PK,Lucas G, Luben R, Loos RJ, Lokki ML, Lettre G, Langenberg C, Launer LJ,Lakatta EG, Laaksonen R, Kyvik KO, Kronenberg F, Konig IR, Khaw KT, KaprioJ, Kaplan LM, Johansson A, Jarvelin MR, Janssens AC, Ingelsson E, Igl W, KeesHovingh G, Hottenga JJ, Hofman A, Hicks AA, Hengstenberg C, Heid IM,Hayward C, Havulinna AS, Hastie ND, Harris TB, Haritunians T, Hall AS,Gyllensten U, Guiducci C, Groop LC, Gonzalez E, Gieger C, Freimer NB,Ferrucci L, Erdmann J, Elliott P, Ejebe KG, Doring A, Dominiczak AF, DemissieS, Deloukas P, de Geus EJ, de Faire U, Crawford G, Collins FS, Chen YD,Caulfield MJ, Campbell H, Burtt NP, Bonnycastle LL, Boomsma DI, BoekholdtSM, Bergman RN, Barroso I, Bandinelli S, Ballantyne CM, Assimes TL,Quertermous T, Altshuler D, Seielstad M, Wong TY, Tai ES, Feranil AB,Kuzawa CW, Adair LS, Taylor Jr HA, Borecki IB, Gabriel SB, Wilson JG, Holm H,Thorsteinsdottir U, Gudnason V, Krauss RM, Mohlke KL, Ordovas JM, MunroePB, Kooner JS, Tall AR, Hegele RA, Kastelein JJ, Schadt EE, Rotter JI,Boerwinkle E, Strachan DP, Mooser V, Stefansson K, Reilly MP, Samani NJ,Schunkert H, Cupples LA, Sandhu MS, Ridker PM, Rader DJ, van Duijn CM,Peltonen L, Abecasis GR, Boehnke M, Kathiresan S. Biological, clinical andpopulation relevance of 95 loci for blood lipids. Nature. 2010;466(7307):707–13.81. Leitch AE, Haslett C, Rossi AG. Cyclin-dependent kinase inhibitor drugs aspotential novel anti-inflammatory and pro-resolution agents. Br J Pharmacol.2009;158(4):1004–16.82. Puddicombe SM, Torres-Lozano C, Richter A, Bucchieri F, Lordan JL,Howarth PH, Vrugt B, Albers R, Djukanovic R, Holgate ST, Wilson SJ, DaviesDE. Increased expression of p21(waf) cyclin-dependent kinase inhibitor inasthmatic bronchial epithelium. Am J Respir Cell Mol Biol. 2003;28(1):61–8.83. Kim JT, Lee SJ, Kang MA, Park JE, Kim BY, Yoon DY, Yang Y, Lee CH, YeomYI, Choe YK, Lee HG. Cystatin SN neutralizes the inhibitory effect of cystatinC on cathepsin B activity. Cell Death Dis. 2013;4, e974.84. Hallstrand TS, Lai Y, Henderson Jr WR, Altemeier WA, Gelb MH. Epithelialregulation of eicosanoid production in asthma. Pulm Pharmacol Ther.2012;25(6):432–7.85. Bonnelykke K, Sleiman P, Nielsen K, Kreiner-Moller E, Mercader JM, BelgraveD, den Dekker HT, Husby A, Sevelsted A, Faura-Tellez G, Mortensen LJ,Paternoster L, Flaaten R, Molgaard A, Smart DE, Thomsen PF, Rasmussen MA,Bonas-Guarch S, Holst C, Nohr EA, Yadav R, March ME, Blicher T, Lackie PM,Jaddoe VW, Simpson A, Holloway JW, Duijts L, Custovic A, Davies DE, TorrentsD, Gupta R, Hollegaard MV, Hougaard DM, Hakonarson H, Bisgaard H. Agenome-wide association study identifies CDHR3 as a susceptibility locus forearly childhood asthma with severe exacerbations. Nat Genet. 2014;46(1):51–5.86. Bai J, Smock SL, Jackson Jr GR, MacIsaac KD, Huang Y, Mankus C, Oldach J,Roberts B, Ma YL, Klappenbach JA, Crackower MA, Alves SE, Hayden PJ.Phenotypic responses of differentiated asthmatic human airway epithelialcultures to rhinovirus. PLoS One. 2015;10(2), e0118286.87. Ho SM. Environmental epigenetics of asthma: an update. J Allergy ClinImmunol. 2010;126(3):453–65.88. Diette GB, McCormack MC, Hansel NN, Breysse PN, Matsui EC. Environmentalissues in managing asthma. Respir Care. 2008;53(5):602–15. discussion 616–607.89. Holgate ST, Davies DE, Powell RM, Howarth PH, Haitchi HM, Holloway JW.Local genetic and environmental factors in asthma disease pathogenesis:chronicity and persistence mechanisms. Eur Respir J. 2007;29(4):793–803.90. Koppelman GH. Gene by environment interaction in asthma. Curr AllergyAsthma Rep. 2006;6(2):103–11.91. Gilliland FD, Gauderman WJ, Vora H, Rappaport E, Dubeau L. Effects ofglutathione-S-transferase M1, T1, and P1 on childhood lung functiongrowth. Am J Respir Crit Care Med. 2002;166(5):710–6.92. Torres D, Dieudonne A, Ryffel B, Vilain E, Si-Tahar M, Pichavant M, Lassalle P,Trottein F, Gosset P. Double-stranded RNA exacerbates pulmonary allergicreaction through TLR3: implication of airway epithelium and dendritic cells.J Immunol. 2010;185(1):451–9.93. Gilmour PS, Rahman I, Donaldson K, MacNee W. Histone acetylationregulates epithelial IL-8 release mediated by oxidative stress fromMoheimani et al. Respiratory Research (2016) 17:119 Page 14 of 15environmental particles. Am J Physiol Lung Cell Mol Physiol.2003;284(3):L533–40.94. Fusunyan RD, Quinn JJ, Fujimoto M, MacDermott RP, Sanderson IR. Butyrateswitches the pattern of chemokine secretion by intestinal epithelial cellsthrough histone acetylation. Mol Med. 1999;5(9):631–40.95. Ito K, Lim S, Caramori G, Chung KF, Barnes PJ, Adcock IM. Cigarette smokingreduces histone deacetylase 2 expression, enhances cytokine expression,and inhibits glucocorticoid actions in alveolar macrophages. FASEB J.2001;15(6):1110–2.96. Bock C, Kiskinis E, Verstappen G, Gu H, Boulting G, Smith ZD, Ziller M, CroftGF, Amoroso MW, Oakley DH, Gnirke A, Eggan K, Meissner A. ReferenceMaps of human ES and iPS cell variation enable high-throughputcharacterization of pluripotent cell lines. Cell. 2011;144(3):439–52.97. Ji H, Zhang X, Oh S, Mayhew CN, Ulm A, Somineni HK, Ericksen M, Wells JM,Khurana Hershey GK. Dynamic transcriptional and epigenomicreprogramming from pediatric nasal epithelial cells to induced pluripotentstem cells. J Allergy Clin Immunol. 2015;135(1):236–44.98. Baccarelli A, Rusconi F, Bollati V, Catelan D, Accetta G, Hou L, Barbone F,Bertazzi PA, Biggeri A. Nasal cell DNA methylation, inflammation, lung functionand wheezing in children with asthma. Epigenomics. 2012;4(1):91–100.99. Stefanowicz D, Hackett TL, Garmaroudi FS, Gunther OP, Neumann S,Sutanto EN, Ling KM, Kobor MS, Kicic A, Stick SM, Pare PD, Knight DA. DNAmethylation profiles of airway epithelial cells and PBMCs from healthy,atopic and asthmatic children. PLoS One. 2012;7(9), e44213.100. Breton CV, Byun HM, Wang X, Salam MT, Siegmund K, Gilliland FD. DNAmethylation in the arginase-nitric oxide synthase pathway is associated withexhaled nitric oxide in children with asthma. Am J Respir Crit Care Med.2011;184(2):191–7.101. Yang Y, Haitchi HM, Cakebread J, Sammut D, Harvey A, Powell RM,Holloway JW, Howarth P, Holgate ST, Davies DE. Epigenetic mechanismssilence a disintegrin and metalloprotease 33 expression in bronchialepithelial cells. J Allergy Clin Immunol. 2008;121(6):1393–9. 1399 e1391-1314.102. Zhu J, Cote-Sierra J, Guo L, Paul WE. Stat5 activation plays a critical role inTh2 differentiation. Immunity. 2003;19(5):739–48.103. Liu J, Kern JA. Neuregulin-1 activates the JAK-STAT pathway and regulateslung epithelial cell proliferation. Am J Respir Cell Mol Biol. 2002;27(3):306–13.104. Kadrmas JL, Beckerle MC. The LIM domain: from the cytoskeleton to thenucleus. Nat Rev Mol Cell Biol. 2004;5(11):920–31.105. Chang DF, Belaguli NS, Iyer D, Roberts WB, Wu SP, Dong XR, Marx JG,Moore MS, Beckerle MC, Majesky MW, Schwartz RJ. Cysteine-rich LIM-onlyproteins CRP1 and CRP2 are potent smooth muscle differentiation cofactors.Dev Cell. 2003;4(1):107–18.106. Lane C, Knight D, Burgess S, Franklin P, Horak F, Legg J, Moeller A, Stick S.Epithelial inducible nitric oxide synthase activity is the major determinant ofnitric oxide concentration in exhaled breath. Thorax. 2004;59(9):757–60.107. Tripathi P, Awasthi S, Gao P. ADAM metallopeptidase domain 33 (ADAM33):a promising target for asthma. Mediat Inflamm. 2014;2014:572025.108. Nicodemus-Johnson J, Naughton KA, Sudi J, Hogarth K, Naurekas ET,Nicolae DL, Sperling AI, Solway J, White SR, Ober C. Genome-wideMethylation Study Identifies an IL-13 Induced Epigenetic Signature inAsthmatic Airways. Am J Respir Criti Care Med. 2016;193(4):376–85.109. Stefanowicz D, Lee JY, Lee K, Shaheen F, Koo HK, Booth S, Knight DA,Hackett TL. Elevated H3K18 acetylation in airway epithelial cells of asthmaticsubjects. Respir Res. 2015;16:95.110. Kouzarides T. Chromatin modifications and their function. Cell. 2007;128(4):693–705.111. Ito K, Caramori G, Lim S, Oates T, Chung KF, Barnes PJ, Adcock IM.Expression and activity of histone deacetylases in human asthmatic airways.Am J Respir Crit Care Med. 2002;166(3):392–6.112. Kobayashi Y, Bossley C, Gupta A, Akashi K, Tsartsali L, Mercado N, Barnes PJ,Bush A, Ito K. Passive smoking impairs histone deacetylase-2 in childrenwith severe asthma. Chest. 2014;145(2):305–12.113. Zhong F, Zhou N, Wu K, Guo Y, Tan W, Zhang H, Zhang X, Geng G, Pan T, LuoH, Zhang Y, Xu Z, Liu J, Liu B, Gao W, Liu C, Ren L, Li J, Zhou J, Zhang H. ASnoRNA-derived piRNA interacts with human interleukin-4 pre-mRNA andinduces its decay in nuclear exosomes. Nucleic Acids Res. 2015;43(21):10474–91.114. Wanet A, Tacheny A, Arnould T, Renard P. miR-212/132 expression andfunctions: within and beyond the neuronal compartment. Nucleic Acids Res.2012;40(11):4742–53.115. Lagos D, Pollara G, Henderson S, Gratrix F, Fabani M, Milne RS, Gotch F,Boshoff C. miR-132 regulates antiviral innate immunity through suppressionof the p300 transcriptional co-activator. Nat Cell Biol. 2010;12(5):513–9.116. Jiang X, Tsitsiou E, Herrick SE, Lindsay MA. MicroRNAs and the regulation offibrosis. FEBS J. 2010;277(9):2015–21.117. Buggele WA, Johnson KE, Horvath CM. Influenza A virus infection ofhuman respiratory cells induces primary microRNA expression. J BiolChem. 2012;287(37):31027–40.118. Pillai RS, Bhattacharyya SN, Filipowicz W. Repression of protein synthesis bymiRNAs: how many mechanisms? Trends Cell Biol. 2007;17(3):118–26.119. Liu J. Control of protein synthesis and mRNA degradation by microRNAs.Curr Opin Cell Biol. 2008;20(2):214–21.120. Chekulaeva M, Filipowicz W. Mechanisms of miRNA-mediated post-transcriptional regulation in animal cells. Curr Opin Cell Biol. 2009;21(3):452–60.121. Jardim MJ, Dailey L, Silbajoris R, Diaz-Sanchez D. Distinct microRNAexpression in human airway cells of asthmatic donors identifies a novelasthma-associated gene. Am J Respir Cell Mol Biol. 2012;47(4):536–42.122. Jackson SJ, Zhang Z, Feng D, Flagg M, O’Loughlin E, Wang D, Stokes N,Fuchs E, Yi R. Rapid and widespread suppression of self-renewal by microRNA-203 during epidermal differentiation. Development. 2013;140(9):1882–91.123. Liao G, Panettieri RA, Tang DD. MicroRNA-203 negatively regulates c-Abl,ERK1/2 phosphorylation, and proliferation in smooth muscle cells.Physiological reports. 2015;3(9):e12541.124. Zandy NL, Playford M, Pendergast AM. Abl tyrosine kinases regulate cell-celladhesion through Rho GTPases. Proc Natl Acad Sci U S A. 2007;104(45):17686–91.125. Solberg OD, Ostrin EJ, Love MI, Peng JC, Bhakta NR, Hou L, Nguyen C, SolonM, Nguyen C, Barczak AJ, Zlock LT, Blagev DP, Finkbeiner WE, Ansel KM,Arron JR, Erle DJ, Woodruff PG. Airway epithelial miRNA expression isaltered in asthma. Am J Respir Crit Care Med. 2012;186(10):965–74.126. Marcet B, Chevalier B, Luxardi G, Coraux C, Zaragosi LE, Cibois M, Robbe-Sermesant K, Jolly T, Cardinaud B, Moreilhon C, Giovannini-Chami L,Nawrocki-Raby B, Birembaut P, Waldmann R, Kodjabachian L, Barbry P.Control of vertebrate multiciliogenesis by miR-449 through direct repressionof the Delta/Notch pathway. Nat Cell Biol. 2011;13(6):693–9.127. Guseh JS, Bores SA, Stanger BZ, Zhou Q, Anderson WJ, Melton DA,Rajagopal J. Notch signaling promotes airway mucous metaplasia andinhibits alveolar development. Development. 2009;136(10):1751–9.128. Song R, Walentek P, Sponer N, Klimke A, Lee JS, Dixon G, Harland R, Wan Y,Lishko P, Lize M, Kessel M, He L. miR-34/449 miRNAs are required for motileciliogenesis by repressing cp110. Nature. 2014;510(7503):115–20.129. Martinez-Nunez RT, Bondanese VP, Louafi F, Francisco-Garcia AS, Rupani H,Bedke N, Holgate S, Howarth PH, Davies DE, Sanchez-Elsner T. A microRNAnetwork dysregulated in asthma controls IL-6 production in bronchialepithelial cells. PLoS One. 2014;9(10), e111659.130. Haj-Salem I, Fakhfakh R, Berube JC, Jacques E, Plante S, Simard MJ, Bosse Y,Chakir J. MicroRNA-19a enhances proliferation of bronchial epithelial cellsby targeting TGFbetaR2 gene in severe asthma. Allergy. 2015;70(2):212–9.131. Nicodemus-Johnson J, Laxman B, Stern RK, Sudi J, Tierney CN, Norwick L,Hogarth DK, McConville JF, Naureckas ET, Sperling AI, Solway J, Krishnan JA,Nicolae DL, White SR, Ober C. Maternal asthma and microRNA regulation ofsoluble HLA-G in the airway. J Allergy Clin Immunol. 2013;131(6):1496–503.132. Lu Y, Thomson JM, Wong HY, Hammond SM, Hogan BL. Transgenicover-expression of the microRNA miR-17-92 cluster promotesproliferation and inhibits differentiation of lung epithelial progenitorcells. Dev Biol. 2007;310(2):442–53.133. Dakhlallah D, Batte K, Wang Y, Cantemir-Stone CZ, Yan P, Nuovo G, MikhailA, Hitchcock CL, Wright VP, Nana-Sinkam SP, Piper MG, Marsh CB.Epigenetic regulation of miR-17 ~ 92 contributes to the pathogenesis ofpulmonary fibrosis. Am J Respir Crit Care Med. 2013;187(4):397–405.134. Carraro G, El-Hashash A, Guidolin D, Tiozzo C, Turcatel G, Young BM, DeLanghe SP, Bellusci S, Shi W, Parnigotto PP, Warburton D. miR-17 family ofmicroRNAs controls FGF10-mediated embryonic lung epithelial branchingmorphogenesis through MAPK14 and STAT3 regulation of E-Cadherindistribution. Dev Biol. 2009;333(2):238–50.135. Tadokoro T, Wang Y, Barak LS, Bai Y, Randell SH, Hogan BL. IL-6/STAT3promotes regeneration of airway ciliated cells from basal stem cells. ProcNatl Acad Sci U S A. 2014;111(35):E3641–9.136. Ventura JJ, Tenbaum S, Perdiguero E, Huth M, Guerra C, Barbacid M,Pasparakis M, Nebreda AR. p38alpha MAP kinase is essential in lung stem andprogenitor cell proliferation and differentiation. Nat Genet. 2007;39(6):750–8.137. Williams AE, Larner-Svensson H, Perry MM, Campbell GA, Herrick SE, AdcockIM, Erjefalt JS, Chung KF, Lindsay MA. MicroRNA expression profiling in mildasthmatic human airways and effect of corticosteroid therapy. PLoS One.2009;4(6), e5889.Moheimani et al. Respiratory Research (2016) 17:119 Page 15 of 15"@en ; edm:hasType "Article"@en ; edm:isShownAt "10.14288/1.0366283"@en ; dcterms:language "eng"@en ; ns0:peerReviewStatus "Reviewed"@en ; edm:provider "Vancouver : University of British Columbia Library"@en ; dcterms:publisher "BioMed Central"@en ; ns0:publisherDOI "10.1186/s12931-016-0434-4"@en ; dcterms:rights "Attribution 4.0 International (CC BY 4.0)"@en ; ns0:rightsURI "http://creativecommons.org/licenses/by/4.0/"@en ; ns0:scholarLevel "Faculty"@en ; dcterms:subject "Epithelial cells"@en, "Asthma"@en, "Genes"@en, "DNA methylation"@en, "Histone acetylation"@en, "microRNA"@en ; dcterms:title "The genetic and epigenetic landscapes of the epithelium in asthma"@en ; dcterms:type "Text"@en ; ns0:identifierURI "http://hdl.handle.net/2429/65851"@en .