spiral.imperial.ac.uk › bitstream › 10044 › 1 › ... · Web viewAirway Epithelial...

43
Airway Epithelial Orchestration of Innate Immune Function in Response to Virus Infection: a Focus on Asthma Andrew I. Ritchie, 1,2,3 David J. Jackson, 4 Michael R. Edwards, 1,2,3 and Sebastian L. Johnston. 1,2,3* 1 Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, UK. 2 MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK. 3 Imperial College Healthcare NHS Trust, London, UK. 4 Guy's and St Thomas' NHS Trust, London, UK. Corresponding author: Professor Sebastian L Johnston, Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom. [email protected]. Tel: +442075943764, fax: +442072628913. Author contributions

Transcript of spiral.imperial.ac.uk › bitstream › 10044 › 1 › ... · Web viewAirway Epithelial...

Airway Epithelial Orchestration of Innate Immune Function in Response to Virus Infection: a Focus on Asthma

Andrew I. Ritchie,1,2,3 David J. Jackson,4 Michael R. Edwards,1,2,3 and Sebastian L. Johnston.1,2,3*

1Airway Disease Infection Section, National Heart and Lung Institute, Imperial College, London, UK.

2MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.

3Imperial College Healthcare NHS Trust, London, UK.

4Guy's and St Thomas' NHS Trust, London, UK.

Corresponding author: Professor Sebastian L Johnston, Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom. [email protected]. Tel: +442075943764, fax: +442072628913.

Author contributions

Writing of manuscript and critical review of content: All authors.

Word Count: 2796

Abstract

Asthma is a very common respiratory condition with worldwide prevalence predicted to increase in the future. There are significant differences in airway epithelial responses in asthma which are of particular interest during exacerbations. Preventing exacerbations is a primary aim when treating asthma as they often necessitate unscheduled healthcare visits, hospitalizations and are a significant cause of morbidity and mortality. The commonest aetiology of asthma exacerbations is a respiratory virus infection of which the most likely type is rhinovirus infection. This article focuses on the role played by the epithelium in orchestrating the innate immune responses to respiratory virus infection. Recent studies show impaired bronchial epithelial cell innate antiviral immune responses, as well as augmentation of a novel bronchial epithelial cell – IL-25/IL-33 – Th2/type 2 innate lymphoid cell – Th2 cytokine response to virus infection in asthma. A better understanding of the mechanisms of these abnormal immune responses has the potential to lead to the development of novel therapeutic targets for virus-induced exacerbations. The aim of this article is to highlight current knowledge regarding the role of viruses and immune modulation in the asthmatic epithelium and discuss exciting areas for future research and novel treatments.

Keywords

Asthma, respiratory viruses, rhinovirus, interferon

Introduction

In excess of 300 million people worldwide suffer from asthma making it a significant source of morbidity and mortality worldwide. Asthma prevalence has increased over recent decades, such that in many European countries up to 30% of children report wheeze in the last year [1]. The pathogenesis of asthma is complex and varies across clinical phenotypes. Complex interactions between genetic, epigenetic, and environmental factors predispose patients to develop a limited number of dysfunctional immunologic regulatory patterns, which in turn dictate the presentation of clinical phenotypes.

Asthma has a typical disease course of chronic respiratory symptoms, such as wheeze and breathlessness, punctuated by periods of markedly increased symptomatology, termed ‘acute exacerbations’ [2]. Acute exacerbations are significant events as they impair quality of life and are the predominant cause of mortality. Moreover, they often necessitate unscheduled healthcare visits, treatment costs and hospitalizations[3].

Preventing exacerbations is a major, currently incompletely met, therapeutic goal. A crucial step towards this goal is the recognition that acute exacerbations are most commonly due to respiratory virus infection [4, 5]. The response to virus infection differs between healthy and asthmatic individuals as highlighted by a study of co-habiting partners, one of whom had asthma, which found that asthmatics exhibit significantly more severe and longer lasting lower respiratory symptoms and greater changes in airway physiology than non-asthmatics [6]. Therefore, it follows that the balance between protective anti-viral responses and inflammatory responses may be skewed in asthmatics, while likely account for the differences in symptom duration and severity. Underpinning this is the airway epithelium, which is highly dynamic and orchestrates the immune, inflammatory and host defence response observed in both stable and exacerbated disease.

This article discusses current knowledge of the airway epithelium’s orchestration of innate immunity and the current understanding of how this differs in asthma since a better understanding has the potential to lead to the development of new therapies.

The Epithelial Response to Rhinovirus Infection

The majority of experimental work on epithelial responses in asthma focus on rhinovirus infections since they are the most commonly detected virus type during exacerbations in epidemiological studies [4, 5, 7]. Rhinoviruses are small (30 nanometer), non-enveloped, single-stranded RNA viruses, that are members of the Picornaviridae family [8]. Rhinoviruses can be classified according to genetic similarity, as rhinovirus-A, -B and –C giving an estimated ~160 species [9]. It is estimated that a substantial proportion of these viruses are currently in circulation. The A & C subtypes are most often associated with wheezing illnesses. Historically the RV-C subtype has proved difficult grow and therefore characterise but recent work suggests that human cadherin-related family member 3 (CDHR3) is a functional binding receptor facilitating cell entry [10].

A cascade of immune and inflammatory responses is triggered when rhinoviruses enter and replicate in epithelial cells . The airway epithelium is of great interest during viral respiratory infections because it serves as the major host cell for viral replication The airway epithelium isare of great interest during viral respiratory infections because they serve as the major host cell for viral replication [11].

A product of rhinovirus replication is synthesised viral RNA, which is recognised by innate immune pattern recognition receptors. These receptors include the cytosolic RNA helicases, retinoic acid inducible gene I (RIG-I), melanoma differentiation-associated protein-5 (MDA-5), dsRNA/protein kinase receptor (PKR) and the toll-like receptors (TLR)-3, -7 and -8 [12-15]. The resulting ligand and receptor binding triggers a signalling cascade which leads to the activation of transcription factors including interferon regulatory factors (IRF)-3 and-7, nuclear factor-κB (NF-κB), p65/p50, activating transcription factor 2 (ATF2) and c-Jun [16-18]. Following activation, the transcription factors translocate to the nucleus of bronchial epithelial cells to induce transcription of type I and type III interferons (IFN-α/-β and IFN-λ1, 2, 3 respectively) and a plethora of pro-inflammatory cytokines and chemokines including interleukin(IL)-6, IL-8 (also known as C-X-C motif ligand 8, CXCL8), epithelial-derived neutrophil-activating peptide 78 (ENA-78, or CXCL5), C-C motif ligand 5 (CCL-5, also known as Regulated on Activation, Normal T cell Expressed and Secreted, RANTES), and IFN-γ-induced protein 10 kDa (IP-10, or CXCL10)[11, 19-26].

IFNs are key components in the innate immune response of the airway epithelium to viral infection[27]. Their antiviral properties are brought about directly through inhibition of viral replication in cells, and indirectly through stimulation of innate and adaptive immune responses. Type I IFNs effect their antiviral activity by a number of mechanisms including blocking viral entry into cells, control of viral transcription, cleavage of RNA, blocking translation, and induction of apoptosis [28]. Underpinning these protective effects are the induction of IFN-stimulated genes (ISGs) and the production of antiviral proteins[27]. The indirect effect of IFNs is brought about through induction of cytokines and chemokines leading to recruitment of natural killer cells and CD4 and CD8 T cells [29], up-regulation of the expression of MHC-I on cells and up-regulation of antigen-presenting cell co-stimulatory molecules. Therefore, the epithelium’s IFN response is central to effective antiviral responses and resolution of virus infection.

Deficient innate anti-viral immunity in the airway epithelium

In 2005, Wark and colleagues published novel work showing mechanistically, the impaired innate immune response to virus infection in the asthmatic epithelium. Primary human bronchial epithelial cells (HBECs) taken from normal and asthmatic subjects at bronchoscopy demonstrated increased rhinovirus replication in the HBECs obtained from asthmatic subjects [30]. Furthermore, they concluded induction of IFN-β was both delayed and deficient in asthmatics and administration of exogenous IFN-β resulted in induced apoptosis and reduced virus replication, thereby demonstrating a causal link between deficient IFN-β and increased virus replication. The observation of a restored antiviral response with administration of type I IFN was validated in a subsequent study [31]. Similarly deficient IFN-λ induction was shown by Contoli et al [23] in HBECs and alveolar macrophages from atopic asthmatics infected ex vivo with rhinovirus-16. This study utilised a human rhinovirus induced asthma exacerbation model to demonstrate that exacerbation severity was inversely proportional to IFN-λ generation. These early studies have provided the stimulus for further work to examine, characterise and understand the mechanisms of IFN deficiency in asthma (see table 1).

However, not all studies have found deficient epithelial IFN responses in asthma [32-35] although it must also be noted that the in vitro studies discussed are small, with different experimental conditions, such as cell culture techniques and virus dose (see table 2). Therefore, the role of the epithelium in the IFN deficient state remains an extremely interesting and plausible explanation for the observed increased severity of virus infection in asthma.

Interestingly there are multiple studies that report relationships between markers of asthma/allergy severity and IFN responses [36-39], and it may be that IFN delay/deficiency is more readily detected in more severe/less well-controlled disease. Analysis of the pre-specified subgroup, containing subjects with more severe disease and ongoing symptoms despite treatment (BTS Steps 4–5), in a recent inhaled IFN-β clinical trial demonstrated IFN-β effectively prevented virus-induced worsening of asthma symptoms, leading authors to conclude that future studies of IFN-β should target patients with moderate or severe disease.[40]. It is also possible that the exact nature of IFN deficiency is specific to certain asthmatic phenotypes. Further studies which involve greater subject numbers and careful patient selection and characterisation are indicated to better understand these mechanisms.

Table 1: Studies supporting type I and/or III IFN deficiency in the asthmatic airway epithelium:

Studies demonstrating type I and/or III IFN deficiency in asthma

Study

Asthmatic subtype studied (vs. healthy control unless stated)

Cell type

Stimulating Virus/analogue

IFN studied

Wark et al, 2005 [30]

Atopic asthmatic

HBECs

Rhinovirus

↓ IFN-β

Contoli et al, 2006 [23]

Atopic asthmatics

HBECs and alveolar macrophages

Rhinovirus

↓ IFN-λ

Gill et al, 2010 [41]

Allergic asthmatics

Plasmacytoid dendritic cells

Influenza A

↓IFN-α

Uller et al, 2010 [42]

Asthma (GINA criteria). Mild to severe disease studied.

HBECs

dsRNA

↓ IFN-β

Edwards et al, 2013 [36]

Children with severe therapy resistant atopic asthma

HBECs

Rhinovirus

↓ IFN-β and IFN-λ

Baraldo et al, 2012 [37]

Asthmatics (atopic and non-atopic) and atopic non-asthmatics

HBECs

Rhinovirus

↓ IFN-β and IFN-λ

Parsons et al, 2014 [43]

Asthma (GINA criteria)

HBECs

Rhinovirus

↓ IFN-λ

Spann et al, 2014 [44]

Children with wheeze and/or atopy.

Nasal epithelial cells

Tracheal epithelial cells

Respiratory syncytial virus

↓ IFN-β***

Wagener et al, 2014 [45]

allergic asthma with concomitant rhinitis and allergic rhinitis.

Primary nasal and HBECs

dsRNA

↓ several interferon-related genes

*** but not IFN-λ

Table 2: Studies not supporting type I and/or III IFN deficiency in the asthmatic airway epithelium:

Studies not demonstrating type I and/or III IFN deficiency in asthma

Study

Asthmatic subtype studied

Cell type

Stimulating Virus/analogue

IFN studied

Lopez-Souza et al, 2009 [33]

Allergic asthma

Nasal epithelial cells and HBECs

Rhinovirus

↔ IFN-β

Bochkov et al, 2010 [32]

atopic asthmatics and atopic-non asthmatics

HBECs

Rhinovirus

↔ Type I or III IFN expression

Sykes et al, 2014[34]

Mild, well controlled atopic asthma

HBECs

Rhinovirus

↔ IFN-λ and, to a lesser degree, IFN-β

Patel et al, 2014 [46]

Asthma (9 of 11 recruits atopic)

HBECs

Influenza A and RSV

↔ Type I or III IFN expression

↓ IFN- λ (in response to RSV)

↑ IFN- λ (in response to influenza virus in asthmatic groups

Sykes et al, 2014 [35]

Atopic asthma

HBECs and PBMCs

TLR agonist

↔ Type I or III IFN expression

Spann et al, 2014 [47]

Children with wheeze and/or atopy.

Tracheal epithelial cells

RSV or hMPV

↔ IFN-β or -λ production

The mechanisms underpinning impaired induction of IFN in response to virus infection in asthmatics are not fully understood. Suppressor of cytokine signalling (SOCS) 1 and SOCS3 are proteins known to act on the respiratory epithelium as negative regulators of cytokines. In murine models, SOCS1 and 2 negatively feedback on Th2 immunity, [48-51], whilst in human studies a genetic polymorphism enhancing SOCS1 is associated with asthma [52] and T cell SOCS3 mRNA levels are increased in asthmatic patients [53]. Edwards et al have shown that rhinovirus infection and Th2 and pro-inflammatory cytokines increase levels of SOCS1 and SOCS3 mRNA in HBECs, while epithelial SOCS1, but not SOCS3, was increased in bronchial biopsies in asthmatic patients [54]. Increased SOCS1 mRNA expression was associated with impaired IFN induction and increased virus replication in HBECs from children with severe asthma. As SOCS1 inhibits both virus-stimulated and IFN signalling dependant IFN induction, antagonism of SOCS1 is an attractive therapeutic target.

Transforming growth factor (TGF)-β is a multifunctional cytokine of interest because it mediates suppression of both IFN-λ and IFN-β in primary bronchial epithelial cells from healthy subjects exposed to rhinovirus [55]. In asthmatics, rhinovirus replication and virus release into supernatants of primary fibroblast cultures incubated with TGF-beta1 is enhanced [56]. Work by Mathur and colleagues further investigated the role of TGF-β mediated IFN suppression by co-culturing rhinovirus infected BEAS-2B monolayers with eosinophils to demonstrate enhanced suppression of IFN induction [57]. This fits with the observation that airway eosinophilia is associated with an increased risk of exacerbation in asthma.

An alternative mechanism of IFN suppression is through the prominent Th2 cytokine, IL-13, which induces IL-1 receptor associated kinase M (IRAK-M). IRAK-M over-expression is present in the asthmatic airway, where it appears to promote lung epithelial rhinovirus replication and autophagy but crucially inhibits rhinovirus-induced IFN-β and IFN-λ1 expression [58]. In vitro, recombinant IL-13 suppressed dsRNA-induced expression of IFN-λs in airway epithelial cells whilst a Janus kinase inhibitor prevented the IL-13 mediated suppression [59]. IL-13 can also reduce RV induced IFN in cell culture experiments [60] again suggesting a link between the actions of Th2 cytokines and IFN expression.

The effects of asthmatic treatment on innate anti-viral immunity were investigated in peripheral blood mononuclear cells infected with rhinovirus and pre-treated with budesonide with and without formoterol. There was reduced type I IFN induction in budesonide treated cells from both healthy and asthmatic donors [61]. Whilst examining theses responses in the epithelium, an influenza A murine infection model demonstrated more severe disease in the steroid treated group and interestingly went on to show adjuvant IFN treatment markedly reduced glucocorticosteroid-amplified infections in human airway cells and in mouse lung [62].

Virus induction of type 2 immunity

Virus infection typically promotes a type 1 immune response however there is clear evidence to support it induces a type 2 inflammatory pattern which is coordinated by the asthmatic epithelium(see figure 1). For example, in a recently completed study, our group has utilised novel methods to sample airway lining fluid from the nose (nasosorption) and the bronchus (bronchosorption) which in combination with low volume protein detection methods demonstrate that the type 2 cytokines IL-4, IL-5, and IL-13 are all induced in vivo in asthmatics, but not in healthy control subjects in a rhinovirus-induced exacerbation model [63]. Moreover, the work demonstrated that levels of IL-5 and IL-13 during infection correlated significantly with exacerbation severity, suggesting that the induction of type 2 cytokines might be functionally important.

Recently important evidence that epithelial-derived mediators including thymic stromal lymphopoietin (TSLP), IL-25, and IL-33, are capable of promoting the type 2 immune response has emerged from animal studies of helminth infections and allergen driven inflammation. Their importance in human asthma exacerbations was, until very recently, unknown. Investigating the role of IL-25 in this context, our group found that rhinovirus-infected HBECs from asthmatic donors had greater IL-25 induction, which correlated with donor atopic status. Human IL-25 levels were induced by experimental rhinovirus infection in vivo and expression was greater in asthmatics at baseline and during infection. In mice, rhinovirus infection also induced IL-25 and augmented allergen-induced IL-25 and blockade of the IL-25 receptor markedly suppressed many rhinovirus-induced exacerbation-specific responses, including type 2 cytokines and chemokines, mucus production, and recruitment of eosinophils, lymphocytes and neutrophils, IL-4+ basophils and Th2 cells, as well as ILC2s. Therefore, asthmatic epithelial cells have an increased intrinsic capacity for IL-25 expression in response to a viral infection, and IL-25 is a key mediator of rhinovirus induced exacerbations of pulmonary inflammation [64].

Our group also recently investigated the mechanisms underpinning induction of type 2 inflammation by IL-33. IL-33 was induced by rhinovirus in the asthmatic airway in vivo and levels in asthmatic subjects related to rhinovirus-induced asthma exacerbation severity. Furthermore, induction of IL-33 correlated with viral load and levels of IL-5 and IL-13 induced by infection. Rhinovirus infection of human primary BECs in vitro strongly induced IL-33, and culture of human T cells and ILC2s with supernatants of rhinovirus-infected BECs (but not with supernatants of mock-infected BECs) strongly induced type 2 cytokines (IL-4, IL-5 and IL-13 in T cells, and IL-5 and IL-13 in ILC2s). These inductions were entirely dependent on IL-33, as blocking the IL-33 receptor in these co-cultures completely suppressed the inductions observed in the cultures with supernatants of rhinovirus infected BECs. Thus, virus-induced IL-33 released from BECs and IL-33-responsive T cells and ILC2s are key mechanistic links between viral infection and exacerbation of asthma (figure 2) [63].

In summary, there is evidence that anti-viral immune responses in the asthmatic respiratory epithelium are impaired (delayed and deficient) and this likely underlies increased disease severity following virus infection in asthma. Further studies are required to determine whether these impairments are common to all asthmatics or whether they are unique to a phenotypic cluster or to more severe and less well controlled asthma. A novel virus infection/epithelial cell-derived cytokine/type 2 cytokine pathway has been identified, in which IL-25 and IL-33 and their receptors appear exciting new targets for development of novel therapies for asthma exacerbations.

Augmented Th2 responses are very clearly associated with the pathogenesis of allergies and asthma, as IL-4 and IL-13 are required for IgE synthesis and IL-5 is required for eosinophil recruitment, maturation and survival. Their importance in the context of virus induced asthma exacerbations is much more controversial however, as Th1 responses not Th2 responses are classically associated with viral infections. Despite this, it is reasonable to hypothesise that a relatively strong Th2 bias at the time of a viral infection might impair Th1 responses to the viral infection and thereby increase the severity of outcomes.

Until now measurement of many soluble mediators (including IL-4, -5, and -13) in airway samples has been difficult with conventional techniques (such as nasal and bronchoalveolar lavage) that wash the airway with large volumes of saline. The subsequent dilution of mediators below detection limits of assays has prevented direct measurement of protein levels in vivo, leading to a reliance on in vitro approaches as described above.

Therapeutic approaches

As the understanding of the key airway epithelial pro-inflammatory mediators in asthma increases, advances have led to development of novel therapeutic approaches (see Table 3). The demonstrable deficient and delayed IFN responses found in asthma, has led to the investigation of inhaled IFN-β as a novel approach to therapy [40].

The inflammatory milieu

At present, anti-inflammatory approaches have been more successful in asthma. The monoclonal antibody class of treatment for asthma is expanding with anti-IgE (omaluzimab) an approved treatment option for selected patients, and anti-IL-4, anti-IL-5 and anti-IL-13 in development. Future targets are likely to include IL-25 and IL-33.

Table 3: Emerging biological therapeutic agents targeting the Th2 pathway in asthma:

Therapeutic approach

Specific treatment

Status

Interferon treatment to restore the immune response with

Inhaled IFN-β

In development, benefit in moderate asthma in early trial [40].

Attenuating IgE-mediated allergic responses

Anti-IgE (omalizumab)

Reduce exacerbations and steroid use [65, 66].

Inhibiting type 2 cytokines in asthma

Anti-IL-5 (mepolizumab, reslizumab, benralizumab)

In refractory eosinophilic asthma, reduces exacerbations [67-69].

Anti-IL-13 (lebrikizumab, tralokinumab)

Currently In late phase clinical trials [70].

Anti -IL-4 /-13 receptor (dupilumab)

Currently In late phase clinical trials [71].

Alternative inflammatory targets

Therapies targeting alternative pro-inflammatory cytokines, chemokines and their receptors are also under investigation, although as these changes are less specific to airways diseases, many candidates are being trialled in other conditions such as rheumatoid arthritis [72-74]. However manipulating the inflammatory response risks impairing the immune system, thus research must proceed cautiously. A good example is the trial of an anti-TNF agent in severe asthma which was stopped early due to increased serious infections, malignancies and one death in the treatment arm [75].

Direct virus targeting

An alternative strategy is to target the virus. Anti-rhinovirus compounds, such as vapendavir, targeting blockade of viral attachment, internalization and/or replication are currently in phase 2 clinical trials (NCT02367313; http://www.clinicaltrials.gov/). Pleconaril, targeting the virus capsid, produced symptomatic relief in naturally acquired colds [76], but was rejected by the US FDA on safety grounds. However the risk-benefit ratio may be different in chronic lung disease, thus trials are ongoing in asthma (pleconaril: ClinicalTrials.gov identifier NCT00394914; vapendavir: NCT01175226; http://www.clinicaltrials.gov/). Rupintrivir is an inhibitor of rhinovirus protease 3C and thus targets rhinovirus replication. However, following successful outcomes in early phase, experimental rhinovirus studies development was ultimately halted because of a lack of efficacy in natural infection studies [77].

Offering annual seasonal influenza vaccines is well established in national asthma treatment guidelines, but development of vaccines against other respiratory viruses, particularly rhinoviruses, is complicated by antigenic diversity and high mutation and recombination rates. Advances made within the field of Bioinformatics is enabling identification of conserved regions across serotypes that could form suitable vaccine targets [78].

An alternative strategy is to target the virus. Offering annual seasonal influenza vaccines is well established in national asthma treatment guidelines, but development of vaccines against other respiratory viruses, particularly rhinoviruses, is complicated by antigenic diversity and high mutation and recombination rates. Advances made within the field of Bioinformatics is enabling identification of conserved regions across serotypes that could form suitable vaccine targets. Compounds, such as vapendavir, targeting blockade of viral attachment, internalization and/or replication are currently in phase 2 clinical trials (NCT02367313; HYPERLINK "http://www.clinicaltrials.gov/" http://www.clinicaltrials.gov/).

Despite optimal use of currently available therapies that are effective at suppressing allergic inflammation the great majority of asthma exacerbations continue to occur. Therefore, current therapies have limited efficacy and development of better treatment options is urgently indicated.

Conclusion

There are significant differences in the airway epithelial orchestrated responses to virus infection in asthma which are of particular interest during exacerbations. Emerging studies highlight the mechanisms underpinning inherent innate immune deficiencies and better characterise the enhanced Th2 inflammatory profile of the asthmatic epithelium. Clarification of the exact mechanisms of increased susceptibility to virus infection in asthma, and how these impact on host immune responses has the potential to lead to the development of novel treatment targets to reduce the impact of virus induced acute exacerbations. Targeting a novel virus infection/epithelial cell-derived cytokine/type 2 cytokine pathway is highly promising for development of new therapies for asthma exacerbations.

Figure 1: Host responses orchestrated by the airway epithelium in the pathogenesis of asthma. Rhinovirus infection induces Il-25, IL-33 and TLSP in the asthmatic epithelium. This figure provides an overview of how the Th2 pathway is induced by this process.

Figure 2: Schematic highlighting the role of rhinovirus infection in promoting IL-25/33 release from the asthmatic airway epithelium. The inflammatory cascade leads to increased eosinophil, Th2 and B cells as well as mucus hypersecretion and increased airway hyperresponsiveness.

Abbreviations

ATF2 = activating transcription factor 2

BEC = bronchial epithelial cells

CCL = Chemokine (C-C motif) ligand

CCR = CC chemokine receptor

CDHR3 = human cadherin-related family member 3 

CXCL = Chemokine (C-X-C motif) ligand

dsRNA = double stranded

FEV1 = forced expiratory volume in one-second

GINA = Global Initiative for Asthma

HBEC = human bronchial epithelial cells

hMPV = human metapneumovirus

ICAM-1 = intercellular adhesion molecule-1

ICS = Inhaled corticosteroids

IFN = Interferon

IL = Interleukin

IP-10 = IFN-γ-Inducible Protein 10 (also known as CXCL10)

ILC2 = Innate lymphoid cells

IRAK-M = IL-1 receptor associated kinase M

IRF = interferon regulatory factor

ISGs = interferon stimulated genes

I-TAC = Interferon-inducible T-cell Alpha Chemoattractant (also known as CXCL11)

LDLR = low density lipoprotein receptors

MDA-5 = melanoma differentiation-associated protein-5 MCP1

MHC-I = major histocompatibility complex class I

mRNA = messenger ribonucleic acid

NDV = Newcastle disease virus

NF-κB = nuclear factor-κB

pBEC = primary bronchial epithelial cells

PBMC = Peripheral blood mononuclear cells

PCR = polymerase chain reaction

PKR = protein kinase receptor

qPCR = quantitative polymerase chain reaction

RANTES = Regulated on Activation, Normal T cell Expressed and Secreted (also known as CCL5)

RIG-I = retinoic acid inducible gene I

RV = Rhinovirus

RSV = Respiratory syncytial virus

SOCs = Suppressor of cytokine signalling

TGF = Transforming growth factor

Th2 = T helper 2 lymphocytes

Th17 = T helper 17 lymphocytes

TLR = Toll Like Receptor

TNF = tumour necrosis factor

TSLP = Thymic stromal lymphoprotein

WHO = World health organisation

References

1.Asher, M.I., et al., Worldwide time trends in the prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and eczema in childhood: ISAAC Phases One and Three repeat multicountry cross-sectional surveys. Lancet, 2006. 368(9537): p. 733-43.

2.Beasley, V., et al., Lung microbiology and exacerbations in COPD. Int J Chron Obstruct Pulmon Dis, 2012. 7: p. 555-69.

3.Donaldson, G.C., et al., Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax, 2002. 57(10): p. 847-52.

4.Johnston, S.L., et al., Community study of role of viral infections in exacerbations of asthma in 9-11 year old children. Bmj, 1995. 310(6989): p. 1225-9.

5.Jackson, D.J. and S.L. Johnston, The role of viruses in acute exacerbations of asthma. J Allergy Clin Immunol, 2010. 125(6): p. 1178-87; quiz 1188-9.

6.Corne, J.M., et al., Frequency, severity, and duration of rhinovirus infections in asthmatic and non-asthmatic individuals: a longitudinal cohort study. Lancet, 2002. 359(9309): p. 831-4.

7.Khetsuriani, N., et al., Prevalence of viral respiratory tract infections in children with asthma. J Allergy Clin Immunol, 2007. 119(2): p. 314-21.

8.Winther, B., et al., Viral-induced rhinitis. Am J Rhinol, 1998. 12(1): p. 17-20.

9.McIntyre, C.L., N.J. Knowles, and P. Simmonds, Proposals for the classification of human rhinovirus species A, B and C into genotypically assigned types. J Gen Virol, 2013. 94(Pt 8): p. 1791-806.

10.Bochkov, Y.A., et al., Cadherin-related family member 3, a childhood asthma susceptibility gene product, mediates rhinovirus C binding and replication. Proc Natl Acad Sci U S A, 2015.

11.Papadopoulos, N.G., et al., Rhinoviruses Infect the Lower Airways. Journal of Infectious Diseases, 2000. 181(6): p. 1875-1884.

12.Slater, L., et al., Co-ordinated role of TLR3, RIG-I and MDA5 in the innate response to rhinovirus in bronchial epithelium. PLoS Pathog, 2010. 6(11): p. e1001178.

13.Yoneyama, M., et al., The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol, 2004. 5(7): p. 730-7.

14.Hewson, C.A., et al., Toll-like receptor 3 is induced by and mediates antiviral activity against rhinovirus infection of human bronchial epithelial cells. J Virol, 2005. 79(19): p. 12273-9.

15.Chen, Y., et al., Rhinovirus induces airway epithelial gene expression through double-stranded RNA and IFN-dependent pathways. Am J Respir Cell Mol Biol, 2006. 34(2): p. 192-203.

16.Johnston, S.L., et al., Low grade rhinovirus infection induces a prolonged release of IL-8 in pulmonary epithelium. J Immunol, 1998. 160(12): p. 6172-81.

17.Korpi-Steiner, N.L., et al., Human rhinovirus induces robust IP-10 release by monocytic cells, which is independent of viral replication but linked to type I interferon receptor ligation and STAT1 activation. J Leukoc Biol, 2006. 80(6): p. 1364-74.

18.Laza-Stanca, V., et al., Rhinovirus replication in human macrophages induces NF-kappaB-dependent tumor necrosis factor alpha production. J Virol, 2006. 80(16): p. 8248-58.

19.Zhu, Z., et al., Rhinovirus stimulation of interleukin-8 in vivo and in vitro: role of NF-kappaB. Am J Physiol, 1997. 273(4 Pt 1): p. L814-24.

20.Edwards, M.R., et al., Corticosteroids and beta2 agonists differentially regulate rhinovirus-induced interleukin-6 via distinct Cis-acting elements. J Biol Chem, 2007. 282(21): p. 15366-75.

21.Spurrell, J.C., et al., Human airway epithelial cells produce IP-10 (CXCL10) in vitro and in vivo upon rhinovirus infection. Am J Physiol Lung Cell Mol Physiol, 2005. 289(1): p. L85-95.

22.Zhu, Z., et al., Rhinovirus stimulation of interleukin-6 in vivo and in vitro. Evidence for nuclear factor kappa B-dependent transcriptional activation. J Clin Invest, 1996. 97(2): p. 421-30.

23.Contoli, M., et al., Role of deficient type III interferon-lambda production in asthma exacerbations. Nat Med, 2006. 12(9): p. 1023-6.

24.Bossios, A., et al., Rhinovirus infection and house dust mite exposure synergize in inducing bronchial epithelial cell interleukin-8 release. Clin Exp Allergy, 2008. 38(10): p. 1615-26.

25.Sheppard, P., et al., IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol, 2003. 4(1): p. 63-8.

26.Ieki, K., et al., Double-stranded RNA activates RANTES gene transcription through co-operation of nuclear factor-kappaB and interferon regulatory factors in human airway epithelial cells. Clin Exp Allergy, 2004. 34(5): p. 745-52.

27.Isaacs, A. and J. Lindenmann, Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci, 1957. 147(927): p. 258-67.

28.Fensterl, V. and G.C. Sen, Interferons and viral infections. Biofactors, 2009. 35(1): p. 14-20.

29.Biron, C.A., et al., Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol, 1999. 17: p. 189-220.

30.Wark, P.A., et al., Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus. J Exp Med, 2005. 201(6): p. 937-47.

31.Cakebread, J.A., et al., Exogenous IFN-beta has antiviral and anti-inflammatory properties in primary bronchial epithelial cells from asthmatic subjects exposed to rhinovirus. J Allergy Clin Immunol, 2011. 127(5): p. 1148-54.e9.

32.Bochkov, Y.A., et al., Rhinovirus-induced modulation of gene expression in bronchial epithelial cells from subjects with asthma. Mucosal Immunol, 2010. 3(1): p. 69-80.

33.Lopez-Souza, N., et al., In vitro susceptibility to rhinovirus infection is greater for bronchial than for nasal airway epithelial cells in human subjects. J Allergy Clin Immunol, 2009. 123(6): p. 1384-90.e2.

34.Sykes, A., et al., Rhinovirus-induced interferon production is not deficient in well controlled asthma. Thorax, 2014. 69(3): p. 240-246.

35.Sykes, A., et al., TLR3, TLR4 and TLRs7-9 Induced Interferons Are Not Impaired in Airway and Blood Cells in Well Controlled Asthma. PLoS One, 2013. 8(6): p. e65921.

36.Edwards, M.R., et al., Impaired innate interferon induction in severe therapy resistant atopic asthmatic children. Mucosal Immunol, 2013. 6(4): p. 797-806.

37.Baraldo, S., et al., Deficient antiviral immune responses in childhood: distinct roles of atopy and asthma. J Allergy Clin Immunol, 2012. 130(6): p. 1307-14.

38.Pritchard, A.L., et al., Asthma is associated with multiple alterations in anti-viral innate signalling pathways. PLoS One, 2014. 9(9): p. e106501.

39.Sykes, A., et al., Rhinovirus 16-induced IFN-alpha and IFN-beta are deficient in bronchoalveolar lavage cells in asthmatic patients. J Allergy Clin Immunol, 2012. 129(6): p. 1506-1514.e6.

40.Djukanovic, R., et al., The effect of inhaled IFN-beta on worsening of asthma symptoms caused by viral infections. A randomized trial. Am J Respir Crit Care Med, 2014. 190(2): p. 145-54.

41.Gill, M.A., et al., Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells. J Immunol, 2010. 184(11): p. 5999-6006.

42.Uller, L., et al., Double-stranded RNA induces disproportionate expression of thymic stromal lymphopoietin versus interferon-beta in bronchial epithelial cells from donors with asthma. Thorax, 2010. 65(7): p. 626-32.

43.Parsons, K.S., A.C. Hsu, and P.A. Wark, TLR3 and MDA5 signalling, although not expression, is impaired in asthmatic epithelial cells in response to rhinovirus infection. Clin Exp Allergy, 2014. 44(1): p. 91-101.

44.Spann, K.M., et al., Viral and host factors determine innate immune responses in airway epithelial cells from children with wheeze and atopy. Thorax, 2014. 69(10): p. 918-25.

45.Wagener, A.H., et al., dsRNA-induced changes in gene expression profiles of primary nasal and bronchial epithelial cells from patients with asthma, rhinitis and controls. Respiratory Research, 2014. 15(1): p. 9-9.

46.Patel, D.A., et al., Interferon response and respiratory virus control are preserved in bronchial epithelial cells in asthma. J Allergy Clin Immunol, 2014. 134(6): p. 1402-1412.e7.

47.Singanayagam, A., et al., Viruses exacerbating chronic pulmonary disease: the role of immune modulation. BMC Med, 2012. 10: p. 27.

48.Ballard, P.L., et al., Regulated gene expression in cultured type II cells of adult human lung. Am J Physiol Lung Cell Mol Physiol, 2010. 299(1): p. L36-50.

49.Wenzel, S.E., et al., Subepithelial basement membrane immunoreactivity for matrix metalloproteinase 9: association with asthma severity, neutrophilic inflammation, and wound repair. J Allergy Clin Immunol, 2003. 111(6): p. 1345-52.

50.Bleecker, E.R., et al., Effect of ADRB2 polymorphisms on response to longacting beta2-agonist therapy: a pharmacogenetic analysis of two randomised studies. Lancet, 2007. 370(9605): p. 2118-25.

51.Sears, M.R., et al., Long-acting beta-agonists: a review of formoterol safety data from asthma clinical trials. Eur Respir J, 2009. 33(1): p. 21-32.

52.Harada, M., et al., Functional polymorphism in the suppressor of cytokine signaling 1 gene associated with adult asthma. Am J Respir Cell Mol Biol, 2007. 36(4): p. 491-6.

53.Seki, Y., et al., SOCS-3 regulates onset and maintenance of T(H)2-mediated allergic responses. Nat Med, 2003. 9(8): p. 1047-54.

54.Gielen, V., et al., Increased nuclear suppressor of cytokine signaling 1 in asthmatic bronchial epithelium suppresses rhinovirus induction of innate interferons. Journal of Allergy and Clinical Immunology.

55.Bedke, N., et al., Transforming Growth Factor-Beta Promotes Rhinovirus Replication in Bronchial Epithelial Cells by Suppressing the Innate Immune Response. PLoS ONE, 2012. 7(9): p. e44580.

56.Thomas, B.J., et al., Transforming growth factor-beta enhances rhinovirus infection by diminishing early innate responses. Am J Respir Cell Mol Biol, 2009. 41(3): p. 339-47.

57.Mathur, S.K., et al., Interaction between allergy and innate immunity: model for eosinophil regulation of epithelial cell interferon expression. Ann Allergy Asthma Immunol, 2013. 111(1): p. 25-31.

58.Wu, Q., et al., Interleukin-1 receptor-associated kinase M (IRAK-M) promotes human rhinovirus infection in lung epithelial cells via the autophagic pathway. Virology, 2013. 446(1-2): p. 199-206.

59.Moriwaki, A., et al., IL-13 suppresses double-stranded RNA-induced IFN-lambda production in lung cells. Biochem Biophys Res Commun, 2011. 404(4): p. 922-7.

60.Contoli, M., et al., Th2 cytokines impair innate immune responses to rhinovirus in respiratory epithelial cells. Allergy: p. n/a-n/a.

61.Davies, J.M., et al., Budesonide and formoterol reduce early innate anti-viral immune responses in vitro. PLoS One, 2011. 6(11): p. e27898.

62.Thomas, B.J., et al., Glucocorticosteroids enhance replication of respiratory viruses: effect of adjuvant interferon. Sci Rep, 2014. 4: p. 7176.

63.Jackson, D.J., et al., IL-33-dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am J Respir Crit Care Med, 2014. 190(12): p. 1373-82.

64.Beale, J., et al., Rhinovirus-induced IL-25 in asthma exacerbation drives type 2 immunity and allergic pulmonary inflammation. Sci Transl Med, 2014. 6(256): p. 256ra134.

65.Busse, W., et al., Omalizumab, anti-IgE recombinant humanized monoclonal antibody, for the treatment of severe allergic asthma. J Allergy Clin Immunol, 2001. 108(2): p. 184-90.

66.Soler, M., et al., The anti-IgE antibody omalizumab reduces exacerbations and steroid requirement in allergic asthmatics. Eur Respir J, 2001. 18(2): p. 254-61.

67.Pavord, I.D., et al., Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet, 2012. 380(9842): p. 651-9.

68.Castro, M., et al., Benralizumab, an anti-interleukin 5 receptor α monoclonal antibody, versus placebo for uncontrolled eosinophilic asthma: a phase 2b randomised dose-ranging study. The Lancet Respiratory Medicine, 2014. 2(11): p. 879-890.

69.Haldar, P., et al., Mepolizumab and Exacerbations of Refractory Eosinophilic Asthma. New England Journal of Medicine, 2009. 360(10): p. 973-984.

70.Corren, J., et al., Lebrikizumab treatment in adults with asthma. N Engl J Med, 2011. 365(12): p. 1088-98.

71.Wenzel, S., et al., Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med, 2013. 368(26): p. 2455-66.

72.Laragione, T., et al., CXCL10 and its receptor CXCR3 regulate synovial fibroblast invasion in rheumatoid arthritis. Arthritis Rheum, 2011. 63(11): p. 3274-83.

73.Yellin, M., et al., A phase II, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of MDX-1100, a fully human anti-CXCL10 monoclonal antibody, in combination with methotrexate in patients with rheumatoid arthritis. Arthritis Rheum, 2012. 64(6): p. 1730-9.

74.Yao, X., et al., Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther, 2014. 141(2): p. 125-39.

75.Wenzel, S.E., et al., A randomized, double-blind, placebo-controlled study of tumor necrosis factor-alpha blockade in severe persistent asthma. Am J Respir Crit Care Med, 2009. 179(7): p. 549-58.

76.Hayden, F.G., et al., Efficacy and safety of oral pleconaril for treatment of colds due to picornaviruses in adults: results of 2 double-blind, randomized, placebo-controlled trials. Clin Infect Dis, 2003. 36(12): p. 1523-32.

77.Thibaut, H.J., A.M. De Palma, and J. Neyts, Combating enterovirus replication: state-of-the-art on antiviral research. Biochem Pharmacol, 2012. 83(2): p. 185-92.

78.Glanville, N., et al., Cross-serotype immunity induced by immunization with a conserved rhinovirus capsid protein. PLoS Pathog, 2013. 9(9): p. e1003669.

13