key: cord-0937062-kt46gwgn authors: Edwards, Michael R.; Strong, Katherine; Cameron, Aoife; Walton, Ross P.; Jackson, David J.; Johnston, Sebastian L. title: Viral infections in allergy and immunology: How allergic inflammation influences viral infections and illness date: 2017-10-31 journal: Journal of Allergy and Clinical Immunology DOI: 10.1016/j.jaci.2017.07.025 sha: f5a4c7e110e00fe0dc001ce61df2ebddb50757cc doc_id: 937062 cord_uid: kt46gwgn Viral respiratory tract infections are associated with asthma inception in early life and asthma exacerbations in older children and adults. Although how viruses influence asthma inception is poorly understood, much research has focused on the host response to respiratory viruses and how viruses can promote; or how the host response is affected by subsequent allergen sensitization and exposure. This review focuses on the innate interferon-mediated host response to respiratory viruses and discusses and summarizes the available evidence that this response is impaired or suboptimal. In addition, the ability of respiratory viruses to act in a synergistic or additive manner with TH2 pathways will be discussed. In this review we argue that these 2 outcomes are likely linked and discuss the available evidence that shows reciprocal negative regulation between innate interferons and TH2 mediators. With the renewed interest in anti-TH2 biologics, we propose a rationale for why they are particularly successful in controlling asthma exacerbations and suggest ways in which future clinical studies could be used to find direct evidence for this hypothesis. IL-4, IL-5, IL-13, and mucin production. 2, 3 Furthermore, cells or tissues from asthmatic patients can respond to infection with a delayed and suboptimal antiviral response exemplified by delayed and deficient production of the innate interferons. [4] [5] [6] Although both processes are undoubtedly important in determining disease outcomes, one idea is that these 2 outcomes are linked and that current anti-T H 2 biologics can function not only by reducing harmful T H 2-related inflammation but also by restoring an impaired antiviral response. This review will examine this hypothesis and discuss the evidence that supports not only impaired interferon production in asthmatic patients but also that impaired interferon production could be caused at least in part by increased T H 2 pathways and allergic inflammation. For simplicity, we will group allergic inflammation and T H 2 pathways together, although we accept that T H 2 cytokines might be made or might function independently of allergen exposure. This review will be limited to the discussion of impaired innate type I (IFN-a/IFN-b) and type III (IFN-ls) interferons in asthmatic patients. Some studies have shown a defective or impaired type II IFN-g response in cells of asthmatic patients or lower T H 1/T H 2 ratios in atopic asthmatic patients versus control subjects 7 ; however, these studies will not be discussed further. Additionally, this review will focus on discussing the role of innate interferons in patients with established GLOSSARY CCL5: A chemokine also known as RANTES, it has been shown to be chemotactic for T cells, eosinophils, and basophils and plays an active role in recruiting leukocytes into inflammatory sites. With the help of particular cytokines that are released by T cells, CCL5 also induces the proliferation and activation of certain natural killer cells to form CC chemokine-activated killer (CHAK) cells. It has also been shown to be an HIV-suppressive factor released from CD8 1 T cells. A type of human type I interferon protein that helps regulate the activity of the immune system. IFN-a is produced by leukocytes and has been shown to be mainly involved in the innate immune response against viral infection. A type of human type I interferon protein produced in large quantities by fibroblasts. IFN-b displays antiviral activity in the innate immune response. IFN-g: A type II interferon, IFN-g is a cytokine required for innate and adaptive immunity against viral, bacterial, and protozoal infections. IFN-g has been shown to be an important activator of macrophages and inducer of class II major histocompatibility complex molecule expression. IFN-g is produced predominantly by natural killer (NK) and NK T cells as part of the innate immune response and by CD4 T H 1 and CD8 cytotoxic T-lymphocyte effector T cells once antigen-specific immunity develops. This recently classified type III interferon group consists of 3 molecules called IFN-l1, IFN-l2, and IFN-l3 (also called IL-29, IL-28A and IL-28B, respectively). IL-28 (IFN-l2/3) is a cytokine that comes in 2 isoforms, IL-28A and IL-28B, which play a role in innate immune defense against viruses. IL-28A and IL-28B are highly similar (in amino acid sequence) to IL-29. Both IL-28A and IL-28B have been shown to exhibit antiviral qualities and inhibit tumor cell proliferation while promoting antitumor immune responses. IL-29 is a potent antiviral cytokine through upregulation of MHC class I expression on the cell surface. to T H 2 cells. IL-4 subsequently produces additional IL-4 in a positive feedback loop. IL-4 is a ligand for the IL-4 receptor that also binds to IL-13, which contributes to many overlapping functions of IL-4 and IL-13. A major maturation and differentiation cytokine expressed by T H 2 cells and eosinophils in mice and human subjects. IL-5 has been shown to play an instrumental role in eosinophilic inflammation in patients with allergic diseases. IL-6: A cytokine also known as IFN-b2 is implicated in a wide variety of inflammation-associated disease states. IL-6 has been associated with maturation of B cells and has been shown to act as an endogenous pyrogen capable of inducing fever in patients with autoimmune diseases or infections. IL-13: A cytokine produced primarily by T H 2 cells that is involved in several stages of B-cell maturation and differentiation and is critical to the pathogenesis of allergen-induced asthma but operates through mechanisms independent of IgE and eosinophils. A proinflammatory cytokine that shares sequence similarity with IL-17 and has been shown to favor the T H 2-type immune response IL-25 activates dendritic cells and innate lymphoid cells (ILC2s). A member of the IL-1 family of cytokines that potently drives production of T H 2-associated cytokines. IL-33 is produced from epithelial cells and activates T H 2 cells and ILC2s. INTERFERON REGULATORY FACTOR (IRF) 3: A member of the IRF factor family, which plays an important role in the innate immune system's response to viral infection through induction of type I interferons. MELANOMA DIFFERENTIATION FACTOR 5: A RIG-I-like receptor double-stranded RNA (dsRNA) helicase enzyme that functions as a pattern recognition receptor (recognizing long dsRNA) that is a sensor for viruses. A universal adapter protein that functions as an essential signal transducer in the IL-1 and Toll-like receptor (TLR) signaling pathways (except TLR3). These pathways regulate activation of numerous proinflammatory genes, including the transcription factor nuclear factor kB. RETINOIC ACID-INDUCIBLE GENE (RIG) I: A RIG-I-like receptor doublestranded RNA (dsRNA) helicase enzyme that functions as a pattern recognition receptor for short dsRNA and 59 phosphorylated singlestranded RNA. RIG-I is a sensor for viruses, specifically influenza A, Sendai virus, and flavivirus, resulting in induction of members of the type I interferon family. SUPPRESSOR OF CYTOKINE SIGNALING 1 (SOCS1): Negative regulators of cytokine signaling that are members of the STAT-induced STAT inhibitor (SSI) family. Expression of this gene has been shown to be induced by a subset of cytokines, including IL-2, IL-3, erythropoietin, CSF2/GM-CSF, type I interferon, and IFN-g. Negative regulator of cytokines that signal through the Janus kinase/signal transducer and activator of transcription pathway. Expression of the SOCS3 gene is induced by various cytokines, including IL-6, IL-10, and IFN-g. TOLL-LIKE RECEPTOR (TLR) 3: A member of the TLR family that plays a fundamental role in pathogen recognition and activation of innate immunity. TLR3 recognizes dsRNA associated with viral infection and induces the activation of interferon regulatory factor 3 and nuclear factor kB. TOLL-LIKE RECEPTOR (TLR) 7: A TLR that recognizes single-stranded RNA (ssRNA) in endosomes, which is a common feature of viral genomes. TLR7 recognizes ssRNA of viruses, such as HIV and hepatitis C virus, and GU-rich ssRNA. TOLL-LIKE RECEPTOR (TLR) 8: Similar to TLR7, TLR8 endosomal receptor that recognizes single-stranded RNA (ssRNA) and can recognize ssRNA viruses, such as influenza, Sendai, and Coxsackie B viruses. TLR8 binding to the viral RNA signals MyD88 and leads to activation of the transcription factor nuclear factor kB and an antiviral response. TLR8 also recognizes ssRNA of viruses, such as HIV and HCV. The Editors wish to acknowledge Kristina Bielewicz, MS, for preparing this glossary. asthma and will not consider the possible role of innate interferon deficiency in asthma inception in any detail. The respiratory viruses associated with asthma include the same viruses that cause the common cold, influenza-like illnesses, and wheeze and bronchiolitis in children. Respiratory viruses are the main triggers of AEs in adults and school-aged children, accounting for around 85%. [8] [9] [10] [11] Viruses associated with AEs include respiratory syncytial virus (RSV), influenza viruses, and human rhinoviruses. Coronaviruses, parainfluenza viruses, adenoviruses, and the more recently identified metapneumoviruses and bocaviruses are also involved; however, they are less common. 12, 13 Both RSV and rhinoviruses have been associated with asthma inception and are strongly associated with wheeze in infants; however, the exact mechanisms remain elusive. There is some argument whether rhinoviruses and RSV cause asthma or might be markers of asthma susceptibility; it is also unclear whether both viruses act in the same manner or are subtly different. In young children RSV bronchiolitis has been linked to increased allergic sensitization 14 and immune skewing to increased T H 2 responses. 15 The skewing to T H 2 responses might reflect a way of evading protective T H 1 responses. Although related, these topics have been thoroughly discussed elsewhere and will not be the main focus of this article; interested readers are directed to other helpful reviews. 1, 16, 17 Respiratory viruses are capable of infecting the bronchial epithelium of the lower airway, resulting in a local inflammatory reaction characterized by airway neutrophilia, attraction of T lymphocytes, activation of macrophages, and damage to the integrity of the bronchial epithelium. 7 In an effort to limit viral spread, host cells also mount a type I (IFN-a/b) and type III (IFN-l) interferon response. Interferons are antiviral cytokines made by a wide range of cells in response to viral infection and ligation of various pattern recognition receptors (PRRs). Type I IFN-as and IFN-b signal through the IFN-a receptor (IFNAR) 1 and IFNAR2 complex, whereas type III IFN-ls (IFN-l1/IL-29, IFN-l2/IL-28A, and IFN-l3/IL-28B in human subjects) act through the IL-10 receptor b chain and IFN-l1 receptor a chain. 18 Interferons are the first-line defense against viral infection 19 and induce expression of the distinct genes termed interferon-stimulated genes (ISGs) through signaling through the type I and type III interferon receptor complexes. 20, 21 ISGs can be extracellular proteins, such as the T H 1-attracting chemokines CXCL10 (interferoninducible protein 10) or CXCL11 (interferon-inducible T-cell alpha chemoattractant [ITAC]), or other interferons, with IFN-b inducing downstream IFN-a and IFN-ls. 22 Most ISGs are intracellular proteins that interfere with and thus attempt to contain viral protein trafficking, nucleic acid synthesis, or virion assembly and release. Currently, the number of known ISGs is thought to exceed 380 23 ; however, there is the possibility of cell type-specific ISGs that are yet to be identified. Rhinovirus infection induces rapid transcription of type I IFNa/b and type III IFN-l mRNAs; however, interferon expression remains in part cell type dependent. The important signaling pathways used by rhinovirus to induce IFN-b and IFN-ls have been studied in vitro and with mouse models 22, [24] [25] [26] and require the PRRs melanoma differentiation factor 5, retinoic acidinducible gene I, Toll-like receptor (TLR) 3, TLR7, and the transcription factor interferon regulatory factor (IRF) 3. An increased susceptibility to viral infections in asthmatic patients was first reported by Corne et al. 27 By studying asthmatic and nonasthmatic cohabiting spouse pairs, it was observed that both asthmatic patients and nonasthmatic subjects reported similar incidences of rhinovirus infections; however, asthmatic patients had prolonged lower respiratory tract symptoms and increased symptom severity. In 2005, Wark et al 5 cultured primary bronchial epithelial cells (HBECs) from bronchial brushings of allergic asthmatic adults and nonallergic nonasthmatic adults ex vivo and infected them with rhinovirus or treated them with the TLR3 ligand polyinosinic:polycytidylic acid (poly I:C). HBECs from asthmatic patients produced lower levels of IFN-b, had lower levels of virus-induced apoptosis, and higher rhinovirus loads over time. Proinflammatory cytokine levels (IL-6 and CCL5) between the 2 groups were similar, and the cells from asthmatic patients produced normal levels of rhinovirus replication when pretreated with exogenous IFN-b. The authors concluded that asthmatic patients had a specific deficiency in IFN-b production. An earlier study observed impaired IFN-a release in PBMCs from asthmatic patients. 4 This work has since been verified by other studies and expanded to include other cell types, including dendritic cells (DCs), 28,29 bronchoalveolar lavage fluid-derived macrophages, 30 alveolar macrophages (AMs), 31 and further confirmed in PBMCs. 32 Furthermore, ex vivo type III IFN-l1 levels from bronchoalveolar lavage macrophages of asthmatic patients have been shown to negatively correlate with total cold scores, changes in lung function, eosinophilia, and markers of inflammation after rhinovirus infection in vivo, strongly suggesting that IFN-ls are important in the pathogenesis of AEs and determining clinical outcomes of rhinovirus infection in asthmatic patients. 6 More recently, several other reports have confirmed these initial findings, 30, [33] [34] [35] including impaired interferon expression in children with severe asthma. 36 This work culminated in the first phase II placebo-controlled clinical trial of inhaled IFN-b for asthma. 37 This was a study of natural infections with the primary outcome of changes in Asthma Control Questionnaire (ACQ) and FEV 1 . Although the trial found no benefit in the primary outcome in the intent-to-treat population of 72 asthmatic patients receiving active treatment versus 75 receiving placebo, a beneficial effect was observed on peak expiratory flow (P 5.033), and in a subpopulation composed of patients with more severe asthma (n 5 27 active and n 5 31 placebo), benefit was observed in ACQ scores (P 5 .004) and peak expiratory flow (P 5 .029). This finding supports the premise that interferon production is indeed impaired in asthmatic patients and that restoring this defect then beneficially influences clinical disease. The impaired interferon responses seen in patients with atopic asthma to date are likely clinically important for 2 reasons. First, impaired interferon production in vivo would not adequately con-trol viral infection and could lead to greater or prolonged infection, including progression and potentially the establishment of infection in the lower airway and associated virus-induced inflammation. Second, impaired interferon production might not counterregulate ongoing T H 2-mediated inflammation, thus contributing to established T H 2-mediated inflammation and thereby increasing disease pathogenesis and increasing risk of hospitalization, which will be discussed at length in the next section. Not all studies investigating impaired interferon production in asthmatic patients are in agreement, with several studies failing to show any difference in interferon production between cells from asthmatic patients and those from control subjects. [38] [39] [40] [41] [42] [43] Reasons for this disparity mostly concern studies of primary HBECs, which are cultured in vitro for several weeks before infection and study. Differences in the above findings are likely due to several variables, including differences in viral strains used or their preparation, 44 cell-culture methods (eg, different components of medium), or methods of propagation, 39 and also recruitment and phenotype of asthmatic subjects, including the asthma severity, asthma control 43 and atopic status. 40 There also exist differing strengths of evidence for these phenomena between various studies and relevance to clinical findings, such as eosinophilia, atopy, or AE rate. The findings of these studies and a description of the level of supporting evidence are summarized in Table I . [4] [5] [6] [28] [29] [30] [31] [32] [33] [34] [35] [36] [44] [45] [46] [47] [48] [49] [50] [51] Although the majority of studies use rhinovirus as a test virus, differences in interferon expression using 48 PBMCs Atopic asthmatic children IFN-a, IFN-b *, k Impaired interferon group identified by using cluster analysis had higher rates of asthma. BAL, Bronchoalveolar lavage; NDV, Newcastle disease virus; NECs, nasal epithelial cells; pDCs, plasmacytoid dendritic cells; STRA, severe therapy-resistant asthma; TEC, tracheal epithelial cell; TSLP, thymic stromal lymphopoietin. Level of evidence: *Interferon levels in asthmatic donors were significantly lower than those of control subjects. Deficient interferon levels were related to clinical disease, such as number of AEs, eosinophilia, lung function, IgE levels, atopy, and T H 2 markers. àSignificantly higher viral loads in asthmatic patients or significantly lower interferon levels that were negatively correlated with higher viral loads in asthmatic patients. §Exogenous IFN-b restored the antiviral response. kOther noninterferon cytokines were not significantly different between asthmatic donors and control subjects. other viruses have produced mixed results. Newcastle disease virus and RSV have both been used to show differences in interferon levels between groups when studying PBMCs 4,32 ; however, a recent side-by-side comparison of interferon responses in asthmatic patients and control subjects with RSV and influenza virus did not show impaired interferon production in HBECs from asthmatic patients. 40 In another study differences in IFN-a responses in plasmacytoid dendritic cells have also been shown with influenza virus. 28 More studies with comparisons of rhinovirus with other respiratory viruses clearly need to be performed to better understand whether the defect in interferon expression is virus specific. Likewise, studies of impaired interferon production in nasal epithelial cells from asthmatic patients have produced mixed results, 41, 45, 46 suggesting that deficiency might be more profound in lower airway cells, yet deficiency is still observable in peripheral leukocyte populations. 28, 32, 47 The role of atopy is perplexing; Baraldo et al 34 showed that deficient interferon production was a feature of atopic asthmatic, nonatopic asthmatic, and nonasthmatic atopic children, 34 and studies focusing on allergic rhinitis without asthma have produced mixed results, 4, 52 furthering the debate about whether it is asthma or atopy (or both) that drives this defect. Some argue that there is controversy regarding interferon deficiency in asthmatic patients because studies investigating interferon responses in patients with stable disease 53 or during AEs in vivo 54,55 find increased rather than decreased interferon levels in asthmatic patients. However, we would argue that studies of interferon levels in vivo will be confounded by differences in viral loads and that if one wishes to investigate the presence of interferon deficiency in a disease state, this can only be done by infecting cells with a standardized quantity of virus ex vivo or stimulating cells with a standard quantity of a viral mimic ex vivo. However, enhanced rhinovirus replication in asthmatic patients in vivo during experimental challenge studies has been observed. 2, 7 Gaps in our knowledge relating to the argument for impaired interferon production in asthmatic patients include (1) a lack of studies testing whether the defect is stable and repeatable in the same samples, (2) few data on asthma phenotypes other than atopic asthma, (3) a paucity of studies with viruses other than rhinovirus or a rhinovirus versus other comparison with other viruses, and (4) a lack of studies relating interferon deficiency to either asthma control at the time samples were taken or underlying asthma severity. Another limitation is that ex vivo studies use small patient numbers and a definite study of interferon expression in larger studies, such as birth cohort studies, is only beginning to be applied with impaired antiviral immunity taken into consideration. Of note, a recent large birth cohort study identified a population of asthmatic patients with impaired interferon responses to rhinovirus at age 11 years by using cluster analysis. 48 This group had increased wheeze in infancy and asthma at adolescence, increased AE rate, and allergic sensitization in infancy. Conversely, a subgroup of children with very low prevalence of asthma were also identified, and this group had robust interferon responses and antiviral immunity. Overall, the data strongly suggest that at least a subgroup of asthmatic patients exists with impaired interferon and antiviral immunity. However, it is possible, although currently unclear, whether asthmatic patients with impaired interferon production are a new and separate endophenotype or an important subgroup of an existing endophenotype. Despite the above differences in study design, the conflicting nature of some findings, and the above limitations, the number of studies that support the original work of Bufe et al 4 and Wark et al 5 continue to grow and now number approximately 20 (Table I) . Our mechanistic understanding of impaired interferon responses in asthmatic patients remains poor, despite much effort. The inability to identify detailed mechanisms likely points to a complex mechanism that is not easily understood. Initially, researchers considered 2 possible explanations. First was that impaired interferon production in asthmatic patients was due to a single gene defect, microRNA, or epigenetic determinant present in asthmatic patients. This hypothesis would describe the defect as static, unchanging, and likely identifiable in genome-wide association studies (GWASs) or perhaps large gene expression data sets from next-generation sequencing approaches or protein activation data sets. The second hypothesis described a more complex gene-environment interaction that could involve a variable or even transient phenotype, immune imprinting, the actions of more than 1 gene, microRNA, or an epigenetic determinant. Thus far, databases of next-generation sequencing 56, 57 and microarray data 38, 58 have generally all failed to show a reproducible single gene defect that would explain the specific decreased expression of innate interferons observed in asthmatic patients. With the exception of TLR7 59 and the highaffinity receptor for the IgE a chain (FcεRIa) at 1q23, 60 large published GWAS data sets on asthma again showed no obvious clue that explains these findings. 61 The possibility still exists that a single gene defect resides in a very specific subpopulation of asthmatic patients but has been diluted in the larger studies that define asthma based on the clinical attributes or FEV 1 alone and often involve several hundred donors. Considering the different cell types involved (leukocytes vs structural cells), it is possible that the mechanism responsible for the defect is cell type specific. It is also possible that HBECs, because of the culture associated with their study, are more prone to lose this defect, explaining the conflicting studies that have arisen. Considering the available evidence, coupled with the failure of several HBEC studies to reproduce this defect, 38, 39, 43 overall, the data argue against the single gene-hypothesis and encourages alternative hypotheses to be considered. Two studies have identified suppressor of cytokine signaling (SOCS) 1, a negative regulator of type I and type II interferon signaling, as potentially important in determining impaired interferons in asthmatic patients. 62 ,63 SOCS1 expression in T cells and airway smooth muscle had been linked previously to asthma 64, 65 ; however, studies of airway epithelial cells and effects on viral responses were few. 66 In HBECs and rhinovirus infection, T H 2 cytokines, and proinflammatory cytokines all induced SOCS1 mRNA and protein in a time-dependent manner. SOCS1, but not the closely related family member SOCS3, was observed to be of greater abundance in bronchial biopsy specimens from patients with atopic asthma, and abundance was related to PC 20 data and atopy. In a small cohort of asthmatic children with severe therapy-resistant asthma, 36 levels of SOCS1 mRNA in unstimulated cells negatively correlated with impaired IFN-b and IFNl expression after rhinovirus infection and poly I:C treatment. 63 SOCS1 can also function as a nuclear protein through a conserved J ALLERGY CLIN IMMUNOL VOLUME 140, NUMBER 4 nuclear localization sequence and might interfere with gene transcription. 67 In the same bronchial biopsy specimens, SOCS1 nuclear staining was increased in asthmatic patients, and functional studies in vitro with mutants lacking the ability to home to the nucleus did not suppress rhinovirus-induced IFN-b and IFN-l1 induction. 63 The data strongly suggest that SOCS1, a virus and T H 2 cytokine-inducible negative regulator that is overexpressed in asthmatic patients, might be at least in part responsible for the impaired interferon levels observed. Further studies are required to support these findings, including bigger studies of SOCS1 expression in bronchial epithelium of asthmatic patients. A recent study showed reduced SOCS1 mRNA expression in bronchial biopsy specimens from patients with severe asthma compared with patients with mild-to-moderate asthma; however, immunostaining was not performed. 68 The idea of increased SOCS1 expression in asthmatic patients remains attractive and provides a mechanistic link of how T H 2 pathways can negatively regulate innate interferons. Studying purified AMs from patients with severe asthma, Rupani et al 31 observed impaired TLR7 expression levels with reduced rhinovirus and TLR7 ligand-induced IFN-b and IFN-a mRNA and protein levels; however, expression of other PRRs, such as TLR8, TLR3, retinoic acid-inducible gene I, and melanoma differentiation factor 5, was not impaired. TLR7 is a single-stranded RNA receptor that uses the adaptor protein myeloid differentiation primary response 88 (MyD88) to engage a signaling apparatus leading to IRF3/7 activation and hence IFNa transcription. Furthermore, TLR7 expression was inversely related to AE rate and ACQ scores, strongly suggesting that TLR7 expression levels are related to clinical asthma outcomes. Studying expression patterns of microRNAs identified several mi-croRNAs that differed between AMs from healthy subjects and asthmatic patients, and 4 of these showed identity to the untranslated region of TLR7. Further expression analysis identified that 3 of these microRNAs, 150, 152, and 375, were significantly upregulated in AMs from asthmatic patients versus control subjects. Each microRNA was not induced by the T H 2 cytokines IL-4 and IL-13, the proinflammatory cytokine TNF, and also IFN-g and were not affected by steroid treatment. The importance of each microRNA in reducing TLR7 expression was then shown by using TLR7-specific luciferase reporters, and targeting each microRNA resulted in significantly increased interferon levels and ISG induction. Together, the data offer an important mechanistic explanation for impaired interferon in AMs from asthmatic patients in that decreased abundance of TLR7, an important PRR, is due to increased expression of 3 microRNAs that target the mRNA of TLR7. Although the article did not discuss why these microRNAs have increased abundance in asthmatic patients, the 3 microRNAs represent new therapeutic targets aiming to restore defective AM antiviral immunity to rhinovirus. The significance of TLR7 has been appreciated in other studies also. Hatchwell et al 49 identified reduced TLR7 expression in asthmatic patients in endobronchial biopsy specimens, which was related to impaired IL-28 (IFN-l2/3) mRNA levels. Work with a mouse model showed that TLR7 can be downregulated specifically by IL-5, and TLR7 staining in endobronchial biopsy specimens was inversely related to sputum eosinophil levels. Together, these studies suggest that in leukocytes TLR7 expression or function might be a key factor determining impaired interferon production in asthmatic patients. Although these studies show clearly the important role of virus sensing by TLR7 on AMs, leading to innate interferon induction, the role of T H 2 pathways on this remain incompletely resolved. The T H 2 cytokines IL-4, IL-5, and IL-13 are well established as effector molecules in patients with atopic asthma. Their roles are well described elsewhere 69 Evidence for synergy between viral infections and allergen sensitization and exposure was first identified in epidemiologic studies, 70, 71 and evidence for this has been extended to both human challenge studies 2,7 and mouse models of rhinovirus. 3, 22, 49, 50, 72 For RSV, several studies in mice have shown that prior exposure to RSV G protein leads to a strong T H 2 response and airway eosinophilia on infection. 73, 74 Furthermore, mice sensitized to ovalbumin through the airway route after RSV infection had increased airway responsiveness and lung eosinophilia associated with T H 2 cytokines, notably IL-5. 75 In asthmatic patients rhinovirus infection of the bronchial epithelium also likely has additive and synergistic effects on allergen sensitization and challenge that are important in AE pathogenesis 72 ; however, a detailed mechanistic insight into these processes is not complete. The discovery of important pro-T H 2 factors (IL-33, IL-25, and thymic stromal lymphopoietin) produced from the epithelium that induce type 2 innate lymphoid cells (ILC2s) now allows a more precise understanding of how respiratory viruses can promote T H 2 pathways because the mediators are induced by viral infection and their expression is consistent with clinical manifestations of asthma, asthma severity, or features of AE pathogenesis. 2,3 How viruses induced pro-T H 2 factors from the epithelium and how this relates to establishing T H 2 immunity are depicted in Fig 1. No study has yet linked impaired interferon responses to a known asthma phenotype; however, most studies observed these phenomena to date in patients with atopic asthma. 6, 28, 30, [34] [35] [36] In the past, sputum eosinophil counts have been related to AE risk 76 and impaired IFN-ls in cultured cells negatively correlated with sputum eosinophilia 6 and both serum IgE and IL-4 staining levels in biopsy specimens. 34 Mechanistically, much evidence shows definitively that type I and III interferons and T H 2 cytokines can exhibit potent negative regulation on the expression or actions of each other. IFN-a or TLR7 agonists can suppress T H 2 cell polarization in pure T-cell and mixed leukocyte culture systems, downregulating levels of GATA-3, IL-4, IL-13, and IL-5. [77] [78] [79] In PBMCs from atopic children, IFN-a can downregulate FcεRIa mRNA levels on both unstimulated and IL-4/IL-13-stimulated cells, highlighting a negative regulatory capacity for IFN-a on FcεRI expression. In the same experiments IFN-a increased FcεRIg chain expression; the g chain is involved in stabilizing FcεRIa on the cell surface and together suggests a more complex role for IFN-a in regulating atopy and IgE-mediated responses. 80 In fact, viral induction of IFN-a in early life is thought by some to enhance FcεRIa expression on DCs and other antigen-presenting cells, likely leading to enhancement of allergic sensitization, and has been suggested as another potential mechanism how viruses contribute to the establishment of atopy. 16 Mouse models have also been able to replicate the negative regulation on T H 2 responses; IFN-l2 (IL-28), when administered through the airways, can suppress the generation of allergic airways inflammation in an ovalbumin sensitization and challenge model in mice that is T H 2 driven. 81 This mechanism involved downregulation of OX40 ligand on DCs, leading to less T H 2 cell polarization. Recently, in a mouse model of influenza virus infection, infection of Ifnar1 2/2 mice resulted in higher levels of virusinduced lung eosinophilia, ILC2 recruitment, and higher levels of serum IgE. 82 In comparative human studies, ILC2s cultured with IL-33 and IL-17 produced the T H 2 cytokines IL-4, IL-5, IL-13, and IL-9, which were downregulated in the presence of IFN-b. 82 This was the first study to show that, in a similar manner to T H 2 cells, innate lymphoid cells are also affected by innate interferons and that their type 2 function might be increased in an environment that lacks interferon and hence this important negative regulatory mechanism. These mechanisms are shown in Fig 2. Conversely, ex vivo and mouse model evidence strongly suggests that T H 2 pathways have potent negative effects on the induction of innate interferons by viruses. For example, cross-linking FcεRI on DCs before influenza virus or rhinovirus challenge downregulates IFN-a production. 28, 29 Blocking IgE function with omalizumab restored this defect and reduced the rate of AEs, showing for the first time outside an experimental infection setting 6 that ex vivo interferon measurements can predict clinical outcomes. 51 In a mouse model of pneumovirus infection, cockroach allergen exposure in early life followed by infection led to an asthma-like phenotype with impaired lung type I and type III interferon production. 83 Ex vivo, cultured, plasmacytoid dendritic cells pretreated with IL-33 showed reduced IFN-a levels on infection, along with reduced levels of IRF7, viperin, and IL-1 receptor-associated kinase 1, an important signaling molecule in TLR7 function. These data further support the importance of TLR7 expression or function in impaired interferon production in immune cells. In HBECs pretreatment with TGF-b, 59,84 IL-4, or IL-13 85 can reduce rhinovirus-induced IFN-b and/or IFN-l levels; this suppression is specific for interferons and does not inhibit proinflammatory cytokines, such as IL-8, 85 which is consistent with studies of impaired interferon production in HBECs. 5, 36 Additionally, other non-T H 2 cytokines, such as IL-2, do not have this inhibitory effect on interferons, again highlighting the unique relationship between T H 2 cytokines and innate interferons, which is not simply an effect of any cytokine impairing the effect of another. Although the mechanism was not studied, it is possibly related to induced SOCS1 because SOCS1 is T H 2 inducible, its levels are increased in asthmatic patients, and it is a potent negative regulator of virus-induced interferon production. 63 Therefore establishing how T H 2 cytokines can impair virus-induced interferons is a priority, and the mechanism could be an important therapeutic target. A summary of mechanisms responsible for T H 2 pathway impairment of innate interferon production is shown in Fig 2. Because the overall evidence strongly underscores many potential mechanisms of counterregulation of T H 2 cytokines and type I and III interferons, it could be proposed that in patients with atopic or eosinophilic asthma, impaired interferon production is due to ongoing and uncontrolled T H 2-mediated inflammation. The observations of increased SOCS1 levels in biopsy specimens of asthmatic patients and that SOCS1 can directly suppress interferon induction 63 suggest a mechanism explaining why interferon levels ex vivo can be inversely associated with markers of T H 2 pathways. However, it is possible that impaired interferon production might not be limited strictly to patients with eosinophilic or T H 2-high asthma 31 and that other mechanisms could explain impaired interferon expression in asthmatic patients. Considering the evidence that impaired interferon production in asthmatic patients can be associated with atopic asthma and the growing wealth of studies using anti-T H 2 biologics, the above idea is a testable hypothesis, and future research should endeavor to examine the effects of T H 2 biologics in clinical trials on interferon expression in asthmatic patients in vivo. The new generation of anti-T H 2 biologics have shown impressive efficacy in reducing the rate of AEs in a number of studies. The different biologics and small molecules currently undergoing clinical trials are summarized in Table II . 51, [86] [87] [88] [89] [90] [91] [92] [93] [94] [95] [96] The anti-IL-13 targeting mAb lebrikizumab produced improvements in the AE rate in a subset of periostin-high asthmatic patients. In the high-T H 2 subgroup, the rate of protocol-defined exacerbations was 60% lower in the lebrikizumab group than in the placebo group (P 5 .03). 86 Dupilumab, an mAb specific for the IL-4 receptor a chain, reduced AE rates by 87% in a study of patients with moderate-to-severe asthma with eosinophilia. 87 Mepolizumab, an anti-IL-5 antibody, reduced blood and sputum eosinophil numbers in a study focusing on prednisolone-taking asthmatic patients with persistent sputum eosinophilia. Mepolizumab also significantly reduced rates of AEs and prednisolone use and improved the median time to AEs by 8 weeks. 88 In another study of patients with eosinophilic asthma with a history of severe AEs, mepolizumab significantly reduced AE rates versus placebo up to 52%. 89 The anti-IL-5 receptor a antibody benralizumab significantly reduced rates of AEs up to 57% in asthmatic patients with high baseline levels of eosinophils. 90 Furthermore reslizumab, another anti-IL-5 antibody, showed a 58% reduction in AE rates, which was not significant versus placebo in patients with poorly controlled eosinophilic asthma, 91 and pitrakinra, an IL-4 variant that inhibits IL-4/IL-13 signaling, reduced the decrease in FEV 1 versus placebo during allergen challenge in atopic asthmatic patients. 92 Small molecules targeting chemoattractant receptor-homologous molecule expressed on T H 2 cells have additionally shown promise increasing FEV 1 93 and reducing eosinophilic airways inflammation, 94 and a DNAzyme-targeted GATA-3 has improved FEV 1 and reduced sputum eosinophil numbers and plasma IL-5 levels versus placebo in patients with eosinophilic asthma. 95 These results are encouraging and show proof of concept that targeting T H 2 pathways in atopic asthmatic patients or asthmatic patients with eosinophilia can lead to improvements in AEs. Targeting IgE has also been shown to be effective. In a study of children and young adults with persistent asthma, omalizumab significantly reduced the proportion of participants who had 1 or more exacerbations by 18% and also reduced the number of days with asthma symptoms. 96 Another study in asthmatic children investigated the effects of omalizumab on AE rates, and omalizumab was found to improve AE rates by approximately 5-fold. 51 IFN-a from rhinovirus-infected PBMC cultures was also sampled before and after omalizumab treatment (4 months). Although there was no difference between omalizumab and placebo before treatment commenced, after randomization, the omalizumab group had improved IFN-a induction after rhinovirus infection, and this increase was related to the AE rate. Based on median IFN-a production in the omalizumab group, those subjects with IFN-a levels of less than the median had an almost 6-fold increase in AE rates compared with those who displayed a higher than median IFN-a response. This observation is of importance because it is the first evidence that measuring IFN-a response ex vivo can predict clinical outcomes in a clinical trial and provocatively suggests that a benefit of anti-T H 2 pathway treatment might be due to restoring an impaired interferon response and increasing antiviral immunity. Considering the number of studies investigating innate interferon responses in asthmatic patients, interferon therapy for asthma, and the renewed appreciation for anti-T H 2 pathway biologics as therapeutic interventions, we have provided a timely summary and offered important insights that might help explain the current state of this field and what will likely follow in future studies. Increased viral replication accompanied by impaired interferon production in lung cells of asthmatic patients was first observed in 2005, 5 and since then, a number of studies have repeated these initial observations, although some have not observed this. This work culminated in the first trial of inhaled IFN-b for asthma in 2014, 37 which showed a significant effect in a subpopulation with more severe disease. Although the importance of impaired interferon production in asthmatic patients remains contentious and the benefit of interferon therapy is in need of confirmation, a number of important discussion points can be made from these studies. The inconsistency of some studies investigating interferon levels in asthmatic patients is likely due to subtle differences in laboratory techniques, including preparation and use of both viruses and cells, as well as patient recruitment and characterization. Furthermore, this phenotype in HBECs might be unstable. There is a paucity of studies measuring interferon levels ex vivo, and the study by Teach et al 51 has shown that this is possible and meaningful. A further point is that future investigations of inhaled IFN-b could benefit from identifying those with impaired interferon responses as a preferred study population; however, whether this is a stable phenotype remains unproved. The identification of SOCS1 63 and microRNAs 150, 152 and 375 controlling TLR7 expression 31 remains of interest but requires confirmation. TLR7's role in determining interferon induction in immune cells is also encouraging, tempting speculation that a dedicated GWAS in poor interferon responders versus good responders might be a worthwhile pursuit. Although the effects of IgE cross-linking on virus-induced interferon 28 Regarding anti-T H 2 biologics, we have proposed herein that the benefit of these therapies on AEs, which largely have a viral cause, could be through restoration of defective antiviral immunity, thus protecting the host from more severe infections that progress to the lower airway. Ex vivo studies, mouse models, and now clinical trials 51 provide conclusive evidence that type I and III innate interferons and T H 2 pathways have antagonistic effects on each other, providing a mechanism or mechanisms explaining how asthmatic patients might benefit from T H 2-targeting therapies. The restoration of a defective innate interferon response could be a novel mechanism of action, and future studies of anti-IL-5, anti-IL-4, and anti-IL-13 biologics should endeavor to examine this in detail. This is a testable hypothesis; however, without conclusive human evidence linking interferon responses to AE-related clinical outcomes, this novel hypothesis will remain underappreciated. The microbiology of asthma IL-33-dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo Rhinovirus-induced IL-25 in asthma exacerbation drives type 2 immunity and allergic pulmonary inflammation Atopic phenotype in children is associated with decreased virus-induced interferon-alpha release Asthmatic bronchial epithelial cells have a deficient innate immune response to infection with rhinovirus Role of deficient type III interferon-lambda production in asthma exacerbations Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production The September epidemic of asthma exacerbations in children: a search for etiology Community study of role of viral infections in exacerbations of asthma in 9-11 year old children The relationship between upper respiratory infections and hospital admissions for asthma: a time-trend analysis Association between human rhinovirus C and severity of acute asthma in children Prevalence of viral respiratory tract infections in children with asthma Newly identified respiratory viruses in children with asthma exacerbation not requiring admission to hospital Respiratory syncytial virus bronchiolitis in infancy is an important risk factor for asthma and allergy at age 7 Respiratory syncytial virus infection in infants is associated with predominant Th-2-like response Viral infections and atopy in asthma pathogenesis: new rationales for asthma prevention and treatment Respiratory syncytial virus(RSV)-induced allergy may be controlled by IL-4 and CX3C fractalkine antagonists and CpG ODN as adjuvant: hypothesis and implications for treatment IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex Virus interference. I. The interferon Functional classification of interferon-stimulated genes identified using microarrays Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics Defining critical roles for NF-kappaB p65 and type I interferon in innate immunity to rhinovirus A diverse range of gene products are effectors of the type I interferon antiviral response Co-ordinated role of TLR3, RIG-I and MDA5 in the innate response to rhinovirus in bronchial epithelium MDA5 and TLR3 initiate pro-inflammatory signaling pathways leading to rhinovirus-induced airways inflammation and hyperresponsiveness Role of double-stranded RNA pattern recognition receptors in rhinovirus-induced airway epithelial cell responses Frequency, severity, and duration of rhinovirus infections in asthmatic and nonasthmatic individuals: a longitudinal cohort study Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells Innate immune responses to rhinovirus are reduced by the high-affinity IgE receptor in allergic asthmatic children Rhinovirus 16-induced IFN-a and IFN-b are deficient in bronchoalveolar lavage cells in asthmatic patients Toll-like receptor 7 is reduced in severe asthma and linked to an altered mi-croRNA profile Impaired virus-induced interferon-alpha2 release in adult asthmatic patients Impaired airway epithelial cell responses from children with asthma to rhinoviral infection Deficient antiviral immune responses in childhood: distinct roles of atopy and asthma Double-stranded RNA induces disproportionate expression of thymic stromal lymphopoietin versus interferon-beta in bronchial epithelial cells from donors with asthma Impaired innate interferon induction in severe therapy resistant atopic asthmatic children The effect of inhaled IFN-beta on worsening of asthma symptoms caused by viral infections. A randomized trial Rhinovirus-induced modulation of gene expression in bronchial epithelial cells from subjects with asthma In vitro susceptibility to rhinovirus infection is greater for bronchial than for nasal airway epithelial cells in human subjects Interferon response and respiratory virus control are preserved in bronchial epithelial cells in asthma Human metapneumovirus impairs apoptosis of nasal epithelial cells in asthma via HSP70 Exogenous IFN-b has antiviral and anti-inflammatory properties in primary bronchial epithelial cells from asthmatic subjects exposed to rhinovirus Rhinovirus-induced interferon production is not deficient in well controlled asthma Diversity in the bronchial epithelial cell response to infection with different rhinovirus strains Viral and host factors determine innate immune responses in airway epithelial cells from children with wheeze and atopy dsRNA-induced changes in gene expression profiles of primary nasal and bronchial epithelial cells from patients with asthma, rhinitis and controls Peripheral blood mononuclear cells from patients with bronchial asthma show impaired innate immune responses to rhinovirus in vitro Phenotyping immune responses in asthma and respiratory infections Toll-like receptor 7 governs interferon and inflammatory responses to rhinovirus and is suppressed by IL-5-induced lung eosinophilia The E3 ubiquitin ligase midline 1 promotes allergen and rhinovirus-induced asthma by inhibiting protein phosphatase 2A activity Preseasonal treatment with either omalizumab or an inhaled corticosteroid boost to prevent fall asthma exacerbations Impaired virus replication and decreased innate immune responses to viral infections in nasal epithelial cells from patients with allergic rhinitis Raised interferon-beta, type 3 interferon and interferon-stimulated genes -evidence of innate immune activation in neutrophilic asthma IFN-a/ IFN-l responses to respiratory viruses in paediatric asthma A comprehensive evaluation of nasal and bronchial cytokines and chemokines following experimental rhinovirus infection in allergic asthma: increased interferons (IFN-g and IFN-l) and type 2 inflammation (IL-5 and IL-13) Gene expression profiles during in vivo human rhinovirus infection: insights into the host response Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways Association analysis identifies TLR7 and TLR8 as novel risk genes in asthma and related disorders Finemapping of IgE-associated loci 1q23, 5q31, and 12q13 using 1000 Genomes Project data A large-scale, consortium-based genomewide association study of asthma Transforming growth factor-Beta promotes rhinovirus replication in bronchial epithelial cells by suppressing the innate immune response Increased nuclear suppressor of cytokine signaling 1 in asthmatic bronchial epithelium suppresses rhinovirus induction of innate interferons Pulmonary suppressor of cytokine signaling-1 induced by IL-13 regulates allergic asthma phenotype Functional polymorphism in the suppressor of cytokine signaling 1 gene associated with adult asthma Cutting edge: innate immune response triggered by influenza A virus is negatively regulated by SOCS1 and SOCS3 through a RIG-I/IFNAR1-dependent pathway Identification of a nuclear localization signal in suppressor of cytokine signaling 1 Reduced epithelial suppressor of cytokine signalling 1 in severe eosinophilic asthma Type 2 inflammation in asthma-present in most, absent in many Synergism between allergens and viruses and risk of hospital admission with asthma: case-control study Study of modifiable risk factors for asthma exacerbations: virus infection and allergen exposure increase the risk of asthma hospital admissions in children Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation Eliminating a region of respiratory syncytial virus attachment protein allows induction of protective immunity without vaccine-enhanced lung eosinophilia Atypical pulmonary eosinophilia is mediated by a specific amino acid sequence of the attachment (G) protein of respiratory syncytial virus Respiratory syncytial virus infection results in airway hyperresponsiveness and enhanced airway sensitization to allergen Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial Innate IFNs and plasmacytoid dendritic cells constrain Th2 cytokine responses to rhinovirus: a regulatory mechanism with relevance to asthma Cutting edge: type I IFN reverses human Th2 commitment and stability by suppressing GATA3 TLR7 Stimulation of APCs results in inhibition of IL-5 through type I IFN and Notch signaling pathways in human peripheral blood mononuclear cells Interactions between innate antiviral and atopic immunoinflammatory pathways precipitate and sustain asthma exacerbations in children IL-28A (IFN-lambda2) modulates lung DC function to promote Th1 immune skewing and suppress allergic airway disease Type I interferon restricts type 2 immunopathology through the regulation of group 2 innate lymphoid cells Aeroallergeninduced IL-33 predisposes to respiratory virus-induced asthma by dampening antiviral immunity Transforming growth factor-beta enhances rhinovirus infection by diminishing early innate responses Th2-cytokines impair innate immune responses to rhinovirus in respiratory epithelial cells Lebrikizumab treatment in adults with asthma Dupilumab in persistent asthma with elevated eosinophil levels Mepolizumab for prednisone-dependent asthma with sputum eosinophilia Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial Benralizumab, an anti-interleukin 5 receptor alpha monoclonal antibody, versus placebo for uncontrolled eosinophilic asthma: a phase 2b randomised dose-ranging study Reslizumab for poorly controlled, eosinophilic asthma: a randomized, placebo-controlled study Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies Heightened response of eosinophilic asthmatic patients to the CRTH2 antagonist OC000459 Fevipiprant, a prostaglandin D2 receptor 2 antagonist, in patients with persistent eosinophilic asthma: a single-centre, randomised, double-blind, parallel-group, placebo-controlled trial Allergen-induced asthmatic responses modified by a GATA3-specific DNAzyme Randomized trial of omalizumab (anti-IgE) for asthma in inner-city children