Abstract
The respiratory epithelium is the major interface between the environment and the host. Sophisticated barrier, sensing, anti-microbial and immune regulatory mechanisms have evolved to help maintain homeostasis and to defend the lung against foreign substances and pathogens. During influenza virus infection, these specialised structural cells and populations of resident immune cells come together to mount the first response to the virus, one which would play a significant role in the immediate and long term outcome of the infection. In this review, we focus on the immune defence machinery of the respiratory epithelium and briefly explore how it repairs and regenerates after infection.
Keywords: Influenza, Airway epithelium, Defence, Sensing, Injury and repair
The respiratory epithelium
The respiratory epithelium starts as the ciliated pseudostratified nasal epithelial lining and continues as the trachea, bronchus, bronchiole (collectively known as the airway epithelium) and finally, the alveolar duct which feeds into the alveolus. The alveolar epithelium comprises a mix of type I and II alveolar epithelium, and has the added distinct adaptation for gas exchange. Both the airway and alveolar epithelial cells co-exist with numerous other specialised cells [Fig. 1] to form the first barrier to the outside world.
Fig. 1.
The respiratory epithelium, and relevant epithelial-associated immune cells and soluble factors involved in IAV infection.
The human respiratory epithelial layer is composed of mostly ciliated cells with some secretory cells and basal cells. Secretory cells including serous, neuroendocrine and goblet cells, generate mucus components and anti-microbial factors including oxidants, enzymes, anti-microbial peptides and protease inhibitors. Ciliated cells facilitate the removal of foreign particles and debris via the mucociliary elevator, sweeping mucus and trapped particles upwards and helping to expel them from the respiratory tract. Club cells (also known as bronchiolar non-mucous secretory cells or Clara cells), are found in distal bronchioles, are not ciliated and may be progenitors to other ciliated cells. Basal cells can also differentiate into other cells types found within the epithelium. Several immune cell populations are resident in epithelium including CD103+ CD8+ T cells and CD103+ conventional dendritic cell populations which act as sentinel cells. Other immune cell populations including innate lymphoid cells (ILCs), mucosal associated invariant T cell (MAIT), natural killer cells (NKT) and γδ T cells are in close proximity to the epithelium. The alveolar epithelium is composed of type I and type II alveolar epithelial cells-type I alveoli epithelial cells enable gas exchange and type II alveolar epithelial cells secrete surfactant lipids and proteins helping to maintain the function and structure of the alveoli. Alveolar macrophages are also resident in the alveolar space.
Different parts of the respiratory epithelium possess different methods of initial defence against pathogens and foreign substances. The airway epithelium utilizes structural and barrier defence provided by the muco-ciliary escalator and their incumbent anti-microbial proteins, and intra- or epithelial -associated immune cells like resident dendritic cells, invariant natural killer T (iNKT) cells, γδ T cells and intra-epithelial lymphocytes [1], [2], [3], [4]. In the alveolar space, a unique environment of surfactant-related antimicrobial proteins and resident immune cells, chiefly, the resident yolk-sac derived, self-renewing alveolar macrophages prevent debris deposition, pathogen colonisation or infection, and generally maintain a comparatively immunologically quiescent tone at homeostasis [5], [6].
The airway epithelium is the major point of defence during influenza A virus (IAV) infection, specified by the tropism of IAV for these structural cells. Human influenza viruses preferentially bind α2,6-linked sialic acid receptors (α2,6-SA) which are predominantly located on airway epithelium from the nose to respiratory bronchioles [7], [8], [9]. Dominance of α2,3-linked SA receptors in the lower respiratory tracts - alveolar ducts and alveoli, prevents initial infection of these sites by established IAV strains. In addition to this distribution of sialic acid receptors, the upper respiratory tracts have also evolved sentinel mechanisms to prevent pathogens from reaching the alveolar space. Failure to do so leads to disruption of the surface tension in alveoli, and the integrity of the closely apposed alveolar capillaries, causing serous leak and collapse of the airspace. Autopsies of fatal cases from all IAV pandemics showed diffuse alveolar damage in the lungs of these individuals [10] characterised by damage of the endothelial-epithelial barrier of the alveoli, fluid leakage and accumulation of cells and blood in the alveolar lumen [11]. In a case of fatal IAV infection with an H5N1 strain, a key cause of pathology was a mutation in the virus which changed the hemagglutinin (HA) binding preference from α2,3 to both α2,3 and α2,6 linked moieties [12], [13]. Thus, a major task of the airway epithelium is to protect pathogens from reaching or infecting the alveolar epithelium. In the following paragraphs, we explore how the airway epithelium and to a lesser extent, the alveolar epithelium, detect and eradicate IAV infection, and what happens to the epithelium after this defence exercise and infection.
The first line of defence against IAV
Tight junctions and polarity of airway epithelial cells
Epithelial cells are attached to their neighbours by cell–cell junctions, including tight junctions (found in the apical part of the lateral epithelial membrane), adherens junctions, gap junctions and desmosomes. These structures link with the cellular cytoskeleton via numerous adaptor proteins and form an impermeable mechanical barrier. They also maintain an apical to basal ionic gradient created by Na+, K+ and Cl− which facilitates directional secretion of many substances [14], enable communication between adjacent cells and regulate intercellular transport of soluble factors [15]. Interestingly, a high concentration of RIG-I, a key activator of type I interferon (IFN) antiviral signalling (see later) is found around tight junctions, suggesting that these junctions could act as apical foci for antiviral sensors [16]. These tight junctions are also resistant to change during infections - in well differentiated airway epithelial cell models, the trans-epithelial resistance conferred by intact tight junctions and the tight junction protein expression were retained throughout the infection period [17].
Antimicrobial proteins secreted by airway and alveolar epithelium
The pseudostratified columnar epithelium [Fig. 1] in the trachea, bronchi and bronchioles are interspersed by a further diverse group of cells – serous, club, neuroendocrine and goblet cells which secrete mucin and a variety of enzymes, protease inhibitors, oxidants, and antimicrobial peptides [1]. Not all of these proteins are relevant to influenza, but mucin, β-defensins and LL37 have roles. β-defensins are secreted by airway epithelial cells and bind to the membrane of pathogens forming a pore-like membrane defect through which essential ions and nutrients efflux. The β-defensin 1 (BD1) isoform is expressed constitutively by human airway epithelial cells whereas expression of BD2, 3 and 4 is inducible [18], [19]. Influenza infection increased expression of BD3 and BD4 in murine airway epithelium [20] and mice lacking BD-1 had a worse outcome after IAV infection [21]. LL37, a cathelicidin, which is released in neutrophil granules and by epithelial cells [22] seems also important in IAV infection. Administering LL37 to mice greatly improved outcome in severe influenza infection - mice had lower mortality, morbidity, viral titre and inflammatory cytokine levels in their lungs [22], [23].
Epithelial cells are covered by a layer of mucus which forms a physical barrier and serves as biophysical ‘rafts’ to bring pathogens in contact with antimicrobial proteins. Mucins make up this mucus layer and are secreted by specialised goblet, club and less abundantly, alveolar epithelial cells. Several isoforms exist. Most mucins are either tethered to epithelial cells (for example, MUC4, MUC13, MUC16 and MUC21) or secreted (MUC5B, MUC5AC and MUC2). In the context of IAV infection, MUC5AC has been shown to be induced in airway epithelial cells after infection [24], [25]. IAV infection of mice overexpressing MUC5AC in their epithelial cells resulted in significantly lower viral titres and neutrophil infiltration in the lungs compared with control mice [26]. Intriguingly, the sialic acid motifs on Muc5AC appear to act as a decoy, binding to SA receptors and so prevent binding of IAV to cell surface sialic acid receptors, and subsequently viral internalisation and infection [26]. Mucins can also communicate the presence of viruses. Airway epithelial cells can detect virus (e.g. herpes simplex virus) via perturbations in the mucin layer, triggering 'hyper-early' innate responses, including the release of CXCL-10 from epithelial cells and recruitment of neutrophils, independent of the early Type I IFN responses. This has not been demonstrated with IAV infection.
In the alveolar space, surfactant proteins contribute to the structure, regulation, and function of pulmonary surfactant and also have intrinsic host defence properties [27]. Pulmonary surfactant is mainly made up of lipids, as well as surfactant proteins (at lower concentrations), that together serve critical roles in reducing surface tension in the alveolus during the dynamic changes of the ventilatory cycle [27]. The surfactant proteins SP-A and SP-D are highly evolutionarily conserved members of the collection family of innate host defence proteins and highly expressed by type II alveolar epithelial cells. The carboxy-terminal lectin-like domains of SP-A and SP-D bind with varying specificity to a diverse range of pathogen-associated molecular patterns (PAMPs), including complex carbohydrate surfaces of common respiratory viruses, bacterial and fungal pathogens, and associated toxins to enhance their opsonization and phagocytosis by alveolar macrophages [28], [29]. During IAV infection, α(2,3)-linked sialic acid motifs on SP-A bind viral HA, inhibiting viral entry into cells [30], [31], [32]. SP-D can also bind to HA proteins on IAV and cluster the viral particles forming large aggregates of viral particles which reduce optimal functioning of the virus [33]. Both SP-D and SP-A increased viral particle binding and phagocytosis by neutrophils [30], [31], [33], [34]. Mice that are SP-D-deficient had reduced viral clearance, increased pro-inflammatory cytokines and elevated neutrophils after influenza infection [35]. Administering exogenous SP-D mutant protein deficient in a specific collagen domain did not correct the surfactant properties in the alveolar space but was able to reverse the reduced viral clearance in IAV-infected SP-D-deficient mice [36], indicating that SP-D had intrinsic anti-viral properties separate from their surface tension function. There is also evidence in humans that surfactant proteins have antiviral functions, fatal IAV infection appear associated with lower transcript levels of SP-D in lung tissue, although it is unclear whether this is the primary defect or consequence of infection [37]. The former is probably more likely, as SP-A, B and D gene polymorphisms were shown to be associated with severe Influenza in humans [38], [39].
Sensing the influenza virus
Three families of innate PRRs including the Toll like receptors (TLRs), RIG-I-like receptors (RLRs) and NOD-like receptors (NLRs) are known to cooperate for early recognition of IAV.
TLRs and epithelial cells
Once IAV crosses these first barriers to reach the airway epithelial cells, there are several key functions that the epithelial cell must perform to limit the spread of infection. They must first rapidly recognize their infected status through sensing viral factors. It then needs to limit viral replication and finally, primarily via soluble factors, they will initiate and influence the local and systemic immune response.
Upon binding sialic acid receptors on the epithelial cell surface, IAV are internalised via receptor-mediated endocytosis [40]. The low pH of the endosome triggers conformational changes and fusion of the viral and endosome membranes, allowing viral RNA and proteins to enter the cell cytoplasm (reviewed by Ref. [41]). Using nucleus localization signals, viral proteins then use the host cell's machinery to translocate into the nucleus where transcription and replication of the viral genome occurs [41]. The host cell begins sensing IAV as soon as it is internalised, utilising pathogen recognition receptors (PRRs), primarily the Toll-like receptors (TLRs) and RNA-sensing RIG-I–like receptors (RLRs), such as retinoic acid–inducible gene I (RIG-I) and melanoma differentiation-associated protein 5 (MDA-5) [Fig. 2].
Fig. 2.
Key sensors of IAV and dominant outcomes of viral sensing pathway activation.
IAV infects cells by first binding of the virion surface glycoprotein hemagglutinin (HA) to sialic acid that is expressed by a cell surface receptor, then is endocytosed. Upon acidification of the endosome, the viral HA undergoes a conformational rearrangement, enabling viral and endosomal membrane fusion. The H+ ions in the acidic environment of the endosome then translocate to the virion interior via the virion's M2 ion channel, causing the viral envelope to permeabilize. As a result, the virion particle opens, the viral RNA is liberated into the cytoplasm of the host cell and then imported into the nucleus to enable transcription of the viral genome and translation of viral proteins. TLR3 and TLR7 sense viral ds and ssRNA respectively within the endosome while RIG-I recognizes cytosolic ssRNA or viral RNA containing 5′-triphosphate and by interacting with MAVS, induces type I and III IFN responses through the transcriptional factors NF-κB and IRF. These cytokines bind to the epithelial's own IFNAR receptors or that expressed by myeloid cells causing expression of ISGs and an antiviral state. In parallel, cytosolic ssRNA and DAMPs can also interact with NLRP3 of the inflammasome complex to cause cleavage and activation of caspase-1 and induction of IL-1β and IL-18.
TLRs are the first PRR to encounter IAV [Fig. 2]. Multiple TLRs - TLR3, TLR7 and TLR8 (human) have been implicated [42], [43], [44], [45], [46], [47], [48], [49]. Upon recognition of the relevant pathogen-associated molecular patterns (PAMPs) – double stranded RNA for TLR3 and single stranded RNA for TLR 7 and 8, these TLRs recruit TIR domain-containing adaptor proteins such as MyD88 and TRIF, which initiate signal transduction pathways that culminate in the activation of NF-κB, IRFs, or MAP kinases to regulate the expression of cytokines, chemokines, and type I IFNs. IAV-infected MyD88−/− and MyD88/TRIF double-deficient mice show a marked reduction of pulmonary cytokine production compared to wild type mice [42], [48], [49], indicating the important role of these TLR signalling pathways in disease.
TLR3 and TLR7 are probably the most relevant respiratory tract TLRs during IAV infection. TLR3 is constitutively expressed by both bronchial and alveolar epithelial cells and recognises viral double-stranded RNA (dsRNA), small interfering RNAs, and self-RNAs derived from damaged cells generated during infection [50], [51], [52], [53]. Unlike other PRRs, TLR3 is expressed on both the cell surface and the endosome, allowing the epithelial cells to sense IAV both externally and internally in the endosome [43], [53], [54], [55], [56]. However, its contribution to host defence is not straightforward. Recognition of IAV by TLR3 leads to secretion of pro-inflammatory cytokines including type I IFNs and IL-6 [53] which can also lead to pathology [53], [56], [57]. Indeed, Tlr3−/−mice survive longer than wild-type mice following lethal IAV infection, despite sustaining higher viral loads in the lungs [57]. In the absence of TLR3, chemokine expression in the lungs, as well as the infiltration of leukocytes and CD8+ T cells, is reduced after IAV infection. However, Tlr3−/− mice generate normal antibody, CD4+ T cell and CD8+ T cell responses to sublethal doses of influenza virus infection [58], which indicates that this sensor is dispensable for generating T cell immunity. Studies have also shown that TLR3 signalling is detrimental to survival and pathology after H5N1 infection, but not after infection with pH1N1 infection, suggesting that the virus itself may utilise TLR3 signalling to subvert detection [42], [57]. However, in all these studies, no differentiation was made between TLR3 expressed by respiratory epithelial cells against those by myeloid cells, and therefore the actual significance of epithelial TLR3 in vivo is still unknown.
Another TLR expressed in human bronchial epithelial cells (in endosomes), TLR7, has also been reported to have a role in viral sensing (recognising single stranded genomic RNA) and immunopathology [59]. Here, its role appears somewhat greater than sensing alone. Activation of TLR7 in primary human bronchial epithelial cells, but not alveolar epithelial cells, in vitro, resulted in an increase in type III IFNs and IL-6 [60], [61]. Mice lacking TLR7 infected with IAV had similar viral titres to controls, but failed to generate germinal centre B centres and mount a B cell response upon re-challenge [62]. Mice deficient in the MAVS adaptor, which is central to RIG-I–like receptor sensing (see later), and TLR7 showed increased mortality independent of viral load [63]. Instead, these mice had increased bacterial burden, caspase-1/11, and neutrophil-dependent tissue damage, suggesting a key role for MAVS and TLR7 in control of immune response and immune pathology during IAV infection [63].
Therefore, viral sensing via TLR7 may have limited effect on viral control, but rather helps shape immune memory responses. However, lack of in vivo studies localising absence of TLR7 to epithelial cells limits interpretation of these studies.
RIG-I-like receptors (RLRs)
During their cytoplasmic phase, IAV RNA species exiting the endosome are detected by RIG-I-like receptors (RLRs) [64], [65]. The RLR family – including MDA-5 and RIG-I, are cytosolic double stranded RNA sensors [64], [66]. RIG-I is the prototypical member, and together with MDA-5, is constitutively expressed by human bronchial epithelial cells. Protein levels of RIG-I and MDA-5 in epithelial cells are relatively low but viral infection greatly increased this expression [53], [67]. The primary function of RLR signalling is to activate interferon genes by interacting with the adaptor mitochondrial anti-viral signalling (MAVS), which in turn engages the interferon regulatory factor (IRF) 3/7 transcription factor signalling pathway [68]. RIG-I signalling is important for activation of the inflammasome via the MAVS–CARD9– nuclear factor κB signalling pathway. In parallel, RIG-I can also directly activate the inflammasome complex by binding the adaptor, ASC [69], [70]. These viral sensing pathways are used by alveolar epithelial cells which upregulate RIG-I and MDA-5 upon IAV infection [71]. IFN responses are diminished when expression of RIG-I or MDA-5 is silenced in alveolar epithelial cell lines (A549) [71]. Thus far in vivo studies have been limited as RIG-I knockout mice are not viable on common laboratory background stains [72]. However, recent studies by Kandasamy et al. demonstrated that RIG-I knockout mice (which are viable when bred on ICR background) have prolonged morbidity due to higher viral loads and failure to clear IAV as quickly as controls [73]. Work by Benitez et al. suggested a hierarchy within the RIG-I-like receptor family with RIG-I the primary and MDA5 a secondary contributor to viral sensing [74]. The relative contribution of these sensing pathways to the generation of an IFN response may vary according to cell type. Crotta et al. demonstrated that in alveolar epithelial cell lines, knocking out TLR7 or MyD88 had little effect on type I or III IFN production, but MAVS ablation greatly reduced IFN levels [75]. Therefore, the MAVS sensing pathway may function as the dominant mechanism of viral sensing and IFN induction in alveolar epithelial cells.
NOD-like receptors and activation of the inflammasome during influenza
NOD-like receptors (NLRs) form part of the multi-protein inflammasome complexes that process caspase 1 and induces the cleavage of pro-IL-1β into its active form IL-1β [76], [77]. Activation of the inflammasome requires two signals; signal 1 results from detection of PAMPs and damage-associated molecular patterns (DAMPs) by PRRs which then induce signaling cascades related to the NF-kB transcription pathway; this leads to upregulation of the multiprotein factors that form the inflammasome complex and immature forms of IL-1β, IL-18 and caspase-1. Signal 2 from a multitude of different factors, leads to complex assembly and activation of caspase-1.
In the setting of IAV infection, NLRP3 is the most studied NLR family member. NLRP3 is expressed in the cytosol and recognises single stranded viral RNA [78], [79], [80], [81], [82]. Human airway epithelial cells have been shown to express NLRP3 and secrete IL-1β when challenged with IAV [83], [84]. Mice lacking NLRP3 (or inflammasome pathway components) showed reduced survival and impaired innate immune responses compared to control mice [83]. However, the majority of the NLRP3 response was derived from myeloid rather than respiratory epithelial cells in this murine model [83]; here too, there is still little to specify the importance of epithelial-expressed NLRP3. The exception is recent work by Kostadinova et al. in bacterial infection which suggests that NLRP3 expressed by epithelial cells could maintain the integrity of the alveolar barrier during Streptococcus pneumoniae infection by enhancing cellular adherence independent of the inflammasome, IL-1β and IL-18 [85].
Along with microbial products, such as viral nucleic acids and proteins, IAV infection results in the release of host cell constituents from both damaged or dying cells and from intact cells. Intracellular molecules (ie, ATP and HMGB1) serve as DAMPs during IAV, are released from infected epithelial cells, most often as a consequence of infection-induced apoptosis, necrosis, or pyroptosis [86], and accumulate in the extracellular space at a high concentration to act as signal 1 for inflammasome activation [87], [88], [89]. Recognition of DAMPs usually, but does not always result in an enhanced innate host response and accelerated viral clearance. For example, recognition of HMGB1 through the DAMP receptor known as receptor for advanced glycation end-products, reduced the host resistance to IAV infection [90].
The contribution of the inflammasome pathway, particularly in epithelial cells during IAV infection, has not been fully explored, but its importance is suggested by the presence of viral mechanisms that interfere with inflammasome activation. For example, the NS1 protein of the H1N1 IAV subtype (eg, A/PR/8/34) is capable of blocking caspase-1 activation, IL-1β maturation, and apoptosis [91]. The caspase-1 inhibitory effect of NS1 seems specific to certain strains, since NS1 from the highly pathogenic avian H5N1 appears not to activate caspases and induces apoptosis of epithelial cells instead [92].
IFN response and interferon stimulated genes in epithelial cells during influenza
Activation of type I interferons is the key consequence of intracellular recognition of IAV infection by TLRs and RLRs. These cytokines bind to the IFN-α/β receptor (IFNAR) on infected as well as neighbouring cells and induces the transcription of a large group of genes (interferon stimulated genes or ISG) whose main task is to limit spread of infection. Although plasmacytoid dendritic cells (DCs) are recognized as the cell type specialized for the production of large amounts of type I interferons [93] during IAV Infection, there is clear evidence that generation and detection of IFN signals also occur in airway epithelial cells. In epithelial cells, type I IFN has the additional task of acting as an early warning system, communicating viral threat between infected and uninfected cells. Another group of interferons, type III interferons, consisting (in humans) of four IFN-λ molecules called IFN-λ1 (IL-29), IFN-λ2 (IL-28A), IFN-λ3 (IL-28B) and IFN-λ4, have been recently identified [94], [95]. IFN-λs signal through a receptor heterodimer complex consisting of IL-10 receptor β and IFN-λR1 (also known as IL-28RA). Despite the distinct receptor complexes used by type I (ie, IFNAR-1 and IFNAR-2) and type III interferons, they trigger similar intracellular signaling pathways in a wide variety of target cells, resulting in many of the same biological activities. However, unlike type I interferon receptors, which are widely expressed on many cell types, including leukocytes, the receptors for IFN-λs are largely restricted to cells of epithelial origin. Moreover, although type I IFN responses are global and can be generated in almost all nucleated cell types, type III responses appear restricted to areas exposed to pathogens like the airway or gut epithelium [96], [97]. There is growing evidence that type III IFNs are the dominant IFN response in the airway epithelium [98], [99], [100], [101], [102], [103], [104], [105], [106] and one specialized for defence against infection at the mucosal interface [107]. Recent studies by Klinkhammer et al. demonstrated that IFN-λ was critical for control of influenza virus dissemination in the upper airways. Mice lacking functional IFN-λ receptors shed significantly more infectious virus particles and transmitted the virus much more efficiently to naïve contacts compared with wild-type mice or mice lacking functional type I IFN receptors [108].
While initiation of Type I IFNs responses can be accompanied by severe immunopathology [109], the generation of type III IFN responses at barrier surfaces generates an antiviral state with limited damage to the host [96]. In humans, mucosal epithelial cells both produce and respond to type III IFNs [61], [110], [111]. In vivo, type III IFNs, rather than type I, are the primary IFNs found in the airways after influenza A virus infection [112]. There appears to be a degree of functional redundancy between type I and III IFNs in the airway epithelium [113], [114]. However, only when both pathways were ablated did mice become highly susceptible to respiratory infections [75]. There is also evidence to suggest chronology in the induction of IFN responses in the lung with type III induced prior to type I [103].
Activation of both type I and III IFN results in induction of hundreds of ISGs. ISGs trigger apoptosis [115], shut down protein synthesis [47] and activate key components of the innate and adaptive immune systems, including antigen presentation and production of cytokines involved in activation of T, B, and natural killer (NK) cells [47]. The most prominent products of ISGs that have an impact on IAV are IFITM3, Mx, PKR and the 2′–5′-oligoadenylate synthetase (OAS)/RNase L system. IFITM3 blocks viral entry by impairing virus-host cell membrane fusion [116], [117]. IAV-infected mice lacking Ifitm3 had higher viral titres, morbidity and mortality compared to control mice [116], [117]. An association between an IFTM3 (rs12252) SNP which causes truncation of IFTM3 and severe influenza infection was also found in UK and Chinese patients [118]. The role of IFITM3 as a restriction factor in IAV infection seems clear though it is not known which IFITM3-expressing cells contribute most to curb IAV spread.
The anti-viral activity of Mx, a family of GTPases, has been recognised for several decades [119], [120], [121], [122]. Its expression is tightly regulated by IFNs although induced rapidly upon IFN pathway activation with kinetics similar to that of type I IFN mRNA expression [123]. The human MxA protein interacts with the IAV nucleocapsid, interfering with nuclear import of viral nucleocapsids as well as with genome transcription and replication, but can be counteracted by the viral NP [124], [125], [126]. Many common inbred laboratory mouse stains which are susceptible to influenza infection have an inactive Mx1 gene due to exon deletion or nonsense mutation which causes termination of translation to protein [120], [122], [127]. A mouse strain with functional Mx protein, the A2G mouse strain, has been shown to be highly resistant to IAV infection [120], [122], [128]. However, the Mx family may be less critical than other ISGs since mice with intact Mx1, but deficient Mavs and TLR7 were found to show increased mortality independent of viral load [63].
The dsRNA-binding kinase, PKR, promotes translational arrest, especially early in IAV infection [129]. PKR also activates NF-κB [130] and recruits RLRs and other antiviral ISG products to optimize antiviral signalling [131].
Another ISG worth mentioning in IAV infection is IRF7 (or interferon regulatory factor 7), an interferon regulatory transcription factor. Knockdown of IRF7 leads to greatly increased viral replication and, conversely, loss of IRF7 repressors substantially lowers influenza replication [132], [133]. Mice lacking IRF7 generated almost no IFNαβ response to IAV infection and exhibited significantly more weight loss and mortality than controls. IRF7 mutations have been linked to severe IAV disease in human, a heterozygous null IRF7 mutation with defects in type I and III IFN response was detected in an otherwise healthy child who suffered life-threatening influenza infection [134]. This study is of particular interest as the investigators generated pulmonary epithelial cells from this patient, and found that IFN responsiveness was impaired and levels of viral protein higher in the infected patient's cells compared to controls [134].
In summary, it seems clear that PRRs in epithelial cells are involved in host defence, but there is not enough evidence to confirm an absolute requirement of epithelial-expressed PRRs for successful protection. Neither the absence of TLR-3 [57], nor the RIG-I signaling adaptor MAVS/IPS-1 [135], diminishes viral clearance and adaptive immunity to IAV infection.
There is likely to be considerable redundancy among different PRRs in their ability to support the early antiviral sensor roles and their presence in epithelial cells may not add greatly to that offered by other sentinel cells in terms of further activity of the innate immune cells. However, conceptually, epithelial TLRs and RLRs and consequent ISG expression are likely to be particularly important in IAV, since sensing and then inducing an anti-viral state in the cells that IAV infects and hijack for propagation, would seem vital to halting infection before spread. There is also a possibility that these PRRs may contribute to immunopathology, be critical determinants of the balance between eradication and immunopathology, and therefore the outcome from IAV infection.
Epithelium-derived cytokines and chemokines during IAV infection
The airway and alveolar epithelium can secrete a variety of cytokines and chemokines that activate and attract other immune cells to the lung. This response is rapid – expression of IL-1β, IL-6, CXCL10, TGF-β, TNF-α, IL-8 and CCL2 are upregulated within 3–24hrs after IAV infection [136], [137], [138], [139], [140]. Expression is hierarchical and synergises with IL-1β and TNF-α, stimulating expression of other cytokines and chemokines [141]. However, many of these cytokines and chemokines are also secreted by resident and recruited immune cells in response to influenza infection and so delineating the specific contribution of epithelial cells to the prevailing cytokine milieu is difficult. Additionally, temporal effects compound the situation. Epithelial cells are likely to be the main early source of cytokines, but within 24–48hrs this balance shifts with recruited and resident immune cells becoming the dominant producers [140], [141], [142]. Here we discuss the roles of some key epithelial-derived cytokine and chemokines IL-1β, IL-6, CCL2 and TGF-β in shaping the immune responses to influenza.
As outlined above, viral triggering of the NLRP3 inflammasome leads to release by epithelial cells of IL-1β a key early pro-inflammatory cytokine [143], [144]. Together with IL-1β produced by resident immune cell populations, chiefly, alveolar macrophages [142], this pluripotent cytokine stimulates secretion of other cytokines and chemokines by the epithelium, and increases adhesion molecule expression, thus enhancing immune cell recruitment to the lungs [145], [146]. Upon infection with IAV, human airway epithelial cells secrete IL-1β from their apical surface, suggesting an autocrine signalling role [83]. In mice, IL-1 signalling is necessary for survival during IAV infection, but this beneficial effect is at the cost of IL-1β driven lung immunopathology [147], [148]. Mice lacking IL-1 receptors had reduced pulmonary inflammation due to impaired neutrophil recruitment, and diminished CD4 T cell activation and recruitment [147].
IL-6 can function as both an anti- and pro-inflammatory cytokine, with diverse roles in the innate and adaptive immune response during IAV infection [149], [150], [151], [152]. Both bronchial and type II alveolar epithelial cells secrete IL-6 after challenge with H1N1 and H5N1 viruses in vitro [137]. IL-6 was elevated in the serum of IAV- infected patients and was a hallmark of disease severity [153], [154]. A recent study by Yang using IL-6 knockout mice demonstrated a role for IL-6 in recovery from lung injury by controlling extracellular matrix turnover and suppressing TGF-β production to prevent fibrosis [155]. IL-6 also reduced virus-mediated apoptosis of airway epithelial cells and improved IAV clearance by macrophage phagocytosis [155].
CCL2 is a monocyte chemo-attractant, secreted by both airway epithelium and activated monocytes [156], [157]. Monocytes recruited to the lung are required for host defence as CCL2 deficient mice do not survive IAV infection. However, they are also a major determinant of immune driven pathology [158], [159]. Mice lacking CCL2 receptors have a better outcome from severe IAV infection [160], [161], [162]. Several studies have linked high levels of CCL2 and inflammatory monocytes in the lungs to severe disease in influenza infected patients [159], [163], [164], [165], [166].
TGF- β is a multifunctional cytokine with important immune regulatory roles. Although most cells can produce TGF- β, its expression is increased in the airway epithelium after IAV infection [140], [167], [168]. IAV has evolved to exploit the immune regulatory properties of TGF- β as the NA protein can cleave latent TGF-β into its active form [167], [169], which then regulates type 1 IFN production. A recent study showed that overexpression of TGF- β in the lungs resulted in reduced inflammatory responses, but impaired viral clearance [170]. When TGF- β was specifically deleted from the airway epithelium, there was a marked reduction in IAV-induced weight loss, airway inflammation and pathology, and viral titres [140]. This protection was not associated with a heightened lymphocytic immune response, but an enhanced early IFN- β release [140]. Therefore, IAV infection induced TGF- β expression in epithelial cells and its subsequent cleavage to the active form, which in turn restrains early IFN- β responses, and resulting in a viral permissive state.
These studies show that the four key cytokines and chemokines secreted by the airway epithelium during IAV infection act to recruit neutrophils, monocytes and CD4 T cells, impacting on repair and anti-inflammatory signals like TGF- β. Their actions are also strongly associated with lung immunopathology.
Epithelium-associated immune cells and IAV infection
The airway epithelium supports niches for several specialised tissue-resident immune cell populations including intra-epithelial conventional DCs (cDCs), innate lymphoid cells (ILCs), unconventional T cell populations (natural killer T cells, NKT; mucosal-associated invariant T cells, MAIT and γδ T cells) and intra-epithelial CD8 T cells. The cDCs and CD8+ T cell population occupy specialised surveillance positions within the epithelium which allow them to sample both the luminal environment and detect perturbations in the epithelial layer. Both cDCs and CD8+ T cells express CD103 (Integrin αE) which binds to integrin β7 expressed by epithelial cells, anchoring them between epithelial cells [2], [3]. There is substantial cross-talk between epithelial and immune cells sequestered in the epithelium [171]. CD103+cDCs continuously sample the airway via extended dendrites and respond to chemokines and cytokines (including type I and III IFNs) and DAMPs secreted by IAV- infected epithelial cells [171], [172], [173].
Intra-epithelial dendritic cells are essential to generate protective IAV-specific CD8+ T cells; mice lacking this DC subset succumb to severe disease and impaired viral clearance (reviewed in Ref. [174]).
Intra epithelial CD8+ T cells differ phenotypically from peripheral blood CD8+ T cells [175]. Piet et al. demonstrated that in human lung tissue, intra-epithelial resident CD8+ T cells function as a local memory population [3]. At homeostasis these resident CD8+ T cells are non-cytotoxic, protecting epithelial integrity [3]. However, recognition of their specific antigen or contact with epithelium derived type I IFNs rapidly increase both cytokine production and cytotoxicity [3].
In addition to the intra-epithelial niche, other immune cell populations are located in close proximity to the epithelium and able to respond to epithelial changes. These include invariant natural killer T (iNKT) cells, a CD1d-restricted lymphocyte with semi-invariant T cell receptors recognizing glycolipid antigens. iNKT cells have immunoregulatory roles [176] and have been shown to be ‘held’ within the intravascular space of the lungs, next to airway epithelium [177]. In this position iNKT cells can be activated rapidly and move into the lung interstitium and airway lumen where they may be involved in enhancing the immune response and regulating the size of myeloid responses [177], [178], [179]. There is evidence that human airway epithelium can express CD1d [179], [180], but it is unknown whether epithelial CD1d is involved in activating iNKT cells; and the cognate glycolipid which activates iNKT cells during IAV infection is also unknown However, IL-12 produced by intra-epithelial CD103 DC may activate iNKT cells in an indirect pathway [181], [182], [183] In the mouse, presence, and exogenous activation, of lung iNKT cells by α-GalCer, protects against lethal H1N1 and H3N2 influenza in prophylactic settings [158], [178] [224]. Recent work by Gaya et al. demonstrated that iNKT cells resident in the interfollicular areas of lung draining lymph nodes are the earliest source of IL-4, prior to activity of T follicular helper cells in this site. This IL-4 seems crucial to the formation of germinal centres and in the ability of antibody-secreting cells to class-switch to IgG1 during IAV infection [184]. It is thought that CD1d-bearing resident macrophages activate iNKT cells in this process, though it is possible that CD1d-expressing epithelial cells may also contribute.
Another group of innate immune cells, the MAIT cells are also associated physically with the airway epithelium and have recently been shown to be protective in influenza infection [185]. Classical activation of MAIT cells is via small molecule ligands, metabolic products generated by microbes, and presented by an MHC-I-like molecule, MR-1; however, during viral infection MAIT cells can be activated via epithelial cytokines [185]. ILC2 populations are enriched at airway epithelial surfaces and respond to epithelial derived cytokines including IL-33 secreted in response to damage or IAV infection [186]. ILC2 can also contribute to epithelial repair via secretion of amphiregulin, observed during IAV infection [187].
There is a group of resident natural killer (NK) cells, now classified as a group 1 ILC together with ILC1s [188], [189], which express a mature and quiescent phenotype with high levels of inhibitory receptors and low levels of activating receptors, migration/adhesion-associated molecules and co-stimulatory molecules [190]. Cells with this phenotype (like intra-epithelial CD8 cells) protect epithelial integrity at homeostasis, but are rapidly activated and express functional molecules to respond to infection [190]. NK cells are one of the first innate cells to be found during IAV infection, but it is unclear whether these are resident NK cells or those recruited from the circulation.
In the above capacity, the airway epithelium provides a platform for the interaction of several innate immune cells which act as early cellular responders to IAV infection.
The respiratory epithelium – injury and repair during IAV infection
The epithelial cells’ attempt to clear IAV results in inevitable tissue injury, in part because of collateral damage from the accompanying innate immune response and direct induction of apoptosis by IAV [92], [191], [192], but also because cytotoxic T cells will eventually kill cells with IAV peptides presented on their MHC class I molecules. If epithelial cells are not killed they undergo apoptosis or de-differentiation. If IAV reaches the alveolar epithelium, various injurious events can occur. The type 1 alveolar epithelial cells are closely apposed to the endothelium forming a barrier [11], and are bound to each other by tight junctions. The alveolar epithelium also keeps the alveolar lumen free of fluid through the action of ion channels and membrane proteins, which includes the epithelial sodium channels (ENaCs), the cystic fibrosis transmembrane conductance regulator (CFTR), and many different aquaporins. ENaC, is found on the apical surface of both type 1 and type 2 alveolar epithelial cells [193], [194]. Very early after infection, IAV causes fluid accumulation in the alveolar lumen by direct inhibition of ENaCs [195]. The M2 ion channel of the virus might also inhibit the activity of ENaCs on epithelial cells by triggering the production of reactive oxygen species and subsequently activating protein kinase C [196]. All these result in compromise of the endothelial-alveolar epithelial barrier, and oedema fluid fill the airspace, causing respiratory insufficiency.
Airway epithelial injury leaves immediate and longer term consequences. One major immediate sequel is co- or secondary bacterial infections. Most influenza deaths are associated with histologic changes or microbiological evidence of bacterial infections [197], [198], [199]. Viral denudation of the airway epithelium provides increased attachment sites for bacteria, resulting in invasive disease and greatly increased mortality. In mouse models, secondary bacterial infection correlates most closely to the point of greatest IAV-induced lung tissue damage [200]. The effect of IAV infection on susceptibility to bacterial infection extends to epithelium-associated innate cells. Alveolar macrophages, a major defence against bacterial infection have been shown to be completely depleted in severe infection [159] although, these were rapidly replaced by monocyte-derived macrophages, and there was no direct evidence that this contributed to secondary infection. However, defects in alveolar macrophage-mediated phagocytosis following IAV infection have been widely reported [201], [202], [203], possibly mediated by IFN-γ [204], [205]. More recent studies investigating the causes of enhanced secondary bacterial infection after IAV infection have implicated disruption of innate immune defences (NK cells, neutrophils) as equally important factors in secondary bacterial infections.
As for longer term sequelae of airway epithelial injury, there is evidence that the anti-inflammatory immune responses upregulated to restore airway epithelial integrity may also leave a mark on susceptibility to future infections, For example, Hussell and colleagues propose that IAV can cause long-term modification of the lung microenvironment through an increase in the myeloid cell negative regulator CD200R (CD200 receptor) and de-sensitization to bacterial products [206], [207]. This may limit subsequent inflammatory damage while the lung is healing, but is likely to predispose to bacterial colonization which worsens outcome.
Multiple mechanisms come into play in the early stages of infection to repair and regenerate after IAV infection. During infection, attempts are also made early on by lung epithelial cells to contain cell damage. This happens in several ways and simultaneously. For example, the airway epithelium expresses CD200, which can engage its receptor (CD200R) displayed by inflammatory macrophages during IAV infection, inhibiting the proinflammatory activity of this cell population [208]. The presence of DAMPs signals can also induce the production of the anti-inflammatory TGF-β and IL-10 from epithelial cells. An intriguing study showed that alveolar macrophages can also establish immunosuppressive communication links with each other through synchronized Ca2+ waves in the epithelium [209] and direct reduction of alveolar neutrophil recruitment and diminished secretion of proinflammatory cytokines in the lungs.
After IAV infection, the clearance of apoptotic cells and cellular debris from the airways is essential to regain tissue homeostasis [210], [211]. Although macrophages are the primary player in this clearance process, they can also enhance phagocytosis in other professional or nonprofessional phagocytes via the release of factors and extracellular vesicles [212]. For example, phagocyte-derived microvesicles and exosomes modulate phagocytic capacity in epithelial cells and insulin-like growth factor-1 (IGF-1) released from macrophages promotes the engulfment of macrophage-derived microvesicles by epithelial cells, leading to reduced inflammatory responses in epithelial cells [212]. Using an epithelial cell specific knockout mouse model, Juncadella at al demonstrated that intracellular signalling via the small GTPase, Rac1, is critical for epithelial cell engulfment [211]. Furthermore induction of this pathway and phagocytosis by epithelial cell resulted in an altered ‘wound healing’ epithelial cell phenotype with increased TGF-β and PGE2 production [211] as observed during house dust mite and ovalbumin administration. Although there is no evidence for this pathway in IAV infection, the study is one of few which were able to attribute a function specifically to airway epithelium. Potentially, the airway epithelium could also contribute similarly to repair after IAV infection.
Once virus and virus-infected cells are cleared and associated pulmonary inflammation is controlled, the repair process begins in earnest. The lung's ability to regenerate the airway epithelium (in contrast to alveolar epithelium) is rapid. Neighbouring epithelial cells spread into the denuded area and progenitor cells proliferate to reconstitute the epithelium, first as squamous epithelial cells, which then differentiate into their original epithelial type. In the alveolar epithelium, the initial repair process is re-epithelialization of the denuded basement membrane and reassembly of tight junctions. This is accomplished primarily by type II alveolar epithelial cells, which are relatively resistant to injury [213], [214], [215], [216], [217], [218]. Type II cells spread, proliferate, and transdifferentiate into type I cells to restore normal alveolar structure and barrier function [213]. Various signaling pathways have been identified that promote type II cell spreading [219], [220], proliferation [214], [221], and trans-differentiation [218], [222]. A recent study shows that a subset of type II alveolar epithelial that is specifically responsive to Wnt and Fgf signalling may be the dominant progenitor cells during injury in mouse and humans [223]. The human lung also contains stem cells capable of forming functional bronchioles, alveoli, and pulmonary vessels after tissue damage, although these cells are less likely than progenitor cells to be relevant to IAV injury [224].
At the same time as epithelial activity, phagocytes - monocyte-derived macrophages and the resident alveolar resident macrophages begin to ingest microparticles released from leukocytes and tissue cells, matrix materials, surfactant components, pathogens (either dead or alive), apoptotic bodies, and intact apoptotic cells [225], [226]. All these must be cleared in order to restore function of the alveoli. Overall, mouse studies depleting macrophages during different stages after tissue injury suggest that macrophages are important for the inflammatory, repair, as well as the resolution stages of lung injury [227], [228], but the relative importance of resident alveolar macrophages to monocyte-derived macrophages in repair is unclear. IAV infection depletes resident alveolar macrophages that are found scattered along the alveolar epithelium early in infection [159], which may impact on repair of the airway epithelium since these macrophages have pro-repair properties.
Conclusion
The respiratory epithelium, comprising the airways and alveolar epithelial cells and their associated resident immune cells and progenitors have evolved a multi-layered defence against influenza A virus. In most individuals the virus is contained within the upper respiratory tract by stringent barrier, sensing and specific anti-viral mechanisms, providing protection of the alveoli from damage. IAV strains have evolved mechanisms to counter these defence, but as long as the airway epithelium can prevent infection and spillover of the inflammatory process to the alveolar epithelium, the likelihood of a good outcome from IAV infection is high. The repair process for the upper airway is robust, enhancing protection of the lower airways (alveolar duct and alveoli) from infection and over exuberant immune responses, essential to survival of the host.
Funding
Laura Denney is supported by NIHR UK Oxford Biomedical Research Centre (BRC) and Ling-Pei Ho by MRC and Oxford University.
Conflicts of interest
The authors declare no conflicts of interest.
Footnotes
Peer review under responsibility of Chang Gung University.
How the respiratory epithelium responds to influenza A virus. The review will explore the role of the airway and alveolar epithelial cells in protection against, sensing and eradication of the virus, and briefly on how they regenerate after infection.
Supplementary data related to this article can be found at https://doi.org/10.1016/j.bj.2018.08.004.
Appendix A. Supplementary data
The following is the supplementary data related to this article:
References
- 1.Ganesan S., Comstock A.T., Sajjan U.S. Barrier function of airway tract epithelium. Tissue Barriers. 2013;1:e24997. doi: 10.4161/tisb.24997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.del Rio M.L., Bernhardt G., Rodriguez-Barbosa J.I., Forster R. Development and functional specialization of CD103+ dendritic cells. Immunol Rev. 2010;234:268–281. doi: 10.1111/j.0105-2896.2009.00874.x. [DOI] [PubMed] [Google Scholar]
- 3.Piet B., de Bree G.J., Smids-Dierdorp B.S., van der Loos C.M., Remmerswaal E.B., von der Thusen J.H. CD8(+) T cells with an intraepithelial phenotype upregulate cytotoxic function upon influenza infection in human lung. J Clin Invest. 2011;121:2254–2263. doi: 10.1172/JCI44675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Qin G., Liu Y., Zheng J., Xiang Z., Ng I.H., Malik Peiris J.S. Phenotypic and functional characterization of human gammadelta T-cell subsets in response to influenza A viruses. J Infect Dis. 2012;205:1646–1653. doi: 10.1093/infdis/jis253. [DOI] [PubMed] [Google Scholar]
- 5.Lambrecht B.N. Alveolar macrophage in the driver's seat. Immunity. 2006;24:366–368. doi: 10.1016/j.immuni.2006.03.008. [DOI] [PubMed] [Google Scholar]
- 6.Hussell T., Bell T.J. Alveolar macrophages: plasticity in a tissue-specific context. Nat Rev Immunol. 2014;14:81–93. doi: 10.1038/nri3600. [DOI] [PubMed] [Google Scholar]
- 7.Shinya K., Ebina M., Yamada S., Ono M., Kasai N., Kawaoka Y. Avian flu: influenza virus receptors in the human airway. Nature. 2006;440:435–436. doi: 10.1038/440435a. [DOI] [PubMed] [Google Scholar]
- 8.Cole S.L., Ho L.P. Contribution of innate immune cells to pathogenesis of severe influenza virus infection. Clin Sci (Lond) 2017;131:269–283. doi: 10.1042/CS20160484. [DOI] [PubMed] [Google Scholar]
- 9.Couceiro J.N., Paulson J.C., Baum L.G. Influenza virus strains selectively recognize sialyloligosaccharides on human respiratory epithelium; the role of the host cell in selection of hemagglutinin receptor specificity. Virus Res. 1993;29:155–165. doi: 10.1016/0168-1702(93)90056-s. [DOI] [PubMed] [Google Scholar]
- 10.Simonsen L., Clarke M.J., Schonberger L.B., Arden N.H., Cox N.J., Fukuda K. Pandemic versus epidemic influenza mortality: a pattern of changing age distribution. J Infect Dis. 1998;178:53–60. doi: 10.1086/515616. [DOI] [PubMed] [Google Scholar]
- 11.Short K.R., Kroeze E.J.B.V., Fouchier R.A.M., Kuiken T. Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect Dis. 2014;14:57–69. doi: 10.1016/S1473-3099(13)70286-X. [DOI] [PubMed] [Google Scholar]
- 12.Auewarakul P., Suptawiwat O., Kongchanagul A., Sangma C., Suzuki Y., Ungchusak K. An avian influenza H5N1 virus that binds to a human-type receptor. J Virol. 2007;81:9950–9955. doi: 10.1128/JVI.00468-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.van Riel D., den Bakker M.A., Leijten L.M., Chutinimitkul S., Munster V.J., de Wit E. Seasonal and pandemic human influenza viruses attach better to human upper respiratory tract epithelium than avian influenza viruses. Am J Pathol. 2010;176:1614–1618. doi: 10.2353/ajpath.2010.090949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Davies J.A., Garrod D.R. Molecular aspects of the epithelial phenotype. Bioessays. 1997;19:699–704. doi: 10.1002/bies.950190810. [DOI] [PubMed] [Google Scholar]
- 15.Roche W.R., Montefort S., Baker J., Holgate S.T. Cell adhesion molecules and the bronchial epithelium. Am Rev Respir Dis. 1993;148:S79–S82. doi: 10.1164/ajrccm/148.6_Pt_2.S79. [DOI] [PubMed] [Google Scholar]
- 16.Mukherjee A., Morosky S.A., Shen L., Weber C.R., Turner J.R., Kim K.S. Retinoic acid-induced gene-1 (RIG-I) associates with the actin cytoskeleton via caspase activation and recruitment domain-dependent interactions. J Biol Chem. 2009;284:6486–6494. doi: 10.1074/jbc.M807547200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wu N.H., Yang W., Beineke A., Dijkman R., Matrosovich M., Baumgartner W. The differentiated airway epithelium infected by influenza viruses maintains the barrier function despite a dramatic loss of ciliated cells. Sci Rep. 2016;6:39668. doi: 10.1038/srep39668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tecle T., Tripathi S., Hartshorn K.L. Review: defensins and cathelicidins in lung immunity. Innate Immun. 2010;16:151–159. doi: 10.1177/1753425910365734. [DOI] [PubMed] [Google Scholar]
- 19.McCray P.B., Jr., Bentley L. Human airway epithelia express a beta-defensin. Am J Respir Cell Mol Biol. 1997;16:343–349. doi: 10.1165/ajrcmb.16.3.9070620. [DOI] [PubMed] [Google Scholar]
- 20.Chong K.T., Thangavel R.R., Tang X. Enhanced expression of murine beta-defensins (MBD-1, -2,- 3, and -4) in upper and lower airway mucosa of influenza virus infected mice. Virology. 2008;380:136–143. doi: 10.1016/j.virol.2008.07.024. [DOI] [PubMed] [Google Scholar]
- 21.Ryan L.K., Dai J., Yin Z., Megjugorac N., Uhlhorn V., Yim S. Modulation of human beta-defensin-1 (hBD-1) in plasmacytoid dendritic cells (PDC), monocytes, and epithelial cells by influenza virus, Herpes simplex virus, and Sendai virus and its possible role in innate immunity. J Leukoc Biol. 2011;90:343–356. doi: 10.1189/jlb.0209079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Tripathi S., Tecle T., Verma A., Crouch E., White M., Hartshorn K.L. The human cathelicidin LL-37 inhibits influenza A viruses through a mechanism distinct from that of surfactant protein D or defensins. J Gen Virol. 2013;94:40–49. doi: 10.1099/vir.0.045013-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Barlow P.G., Svoboda P., Mackellar A., Nash A.A., York I.A., Pohl J. Antiviral activity and increased host defense against influenza infection elicited by the human cathelicidin LL-37. PLoS One. 2011;6:e25333. doi: 10.1371/journal.pone.0025333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Barbier D., Garcia-Verdugo I., Pothlichet J., Khazen R., Descamps D., Rousseau K. Influenza A induces the major secreted airway mucin MUC5AC in a protease-EGFR-extracellular regulated kinase-Sp1-dependent pathway. Am J Respir Cell Mol Biol. 2012;47:149–157. doi: 10.1165/rcmb.2011-0405OC. [DOI] [PubMed] [Google Scholar]
- 25.Buchweitz J.P., Harkema J.R., Kaminski N.E. Time-dependent airway epithelial and inflammatory cell responses induced by influenza virus A/PR/8/34 in C57BL/6 mice. Toxicol Pathol. 2007;35:424–435. doi: 10.1080/01926230701302558. [DOI] [PubMed] [Google Scholar]
- 26.Ehre C., Worthington E.N., Liesman R.M., Grubb B.R., Barbier D., O'Neal W.K. Overexpressing mouse model demonstrates the protective role of Muc5ac in the lungs. Proc Natl Acad Sci U S A. 2012;109:16528–16533. doi: 10.1073/pnas.1206552109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Whitsett J.A., Wert S.E., Weaver T.E. Alveolar surfactant homeostasis and the pathogenesis of pulmonary disease. Annu Rev Med. 2010;61:105–119. doi: 10.1146/annurev.med.60.041807.123500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hartshorn K.L. Role of surfactant protein A and D (SP-A and SP-D) in human antiviral host defense. Front Biosci (Schol Ed) 2010;2:527–546. doi: 10.2741/s83. [DOI] [PubMed] [Google Scholar]
- 29.Ariki S., Nishitani C., Kuroki Y. Diverse functions of pulmonary collectins in host defense of the lung. J Biomed Biotechnol. 2012;2012:532071. doi: 10.1155/2012/532071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Benne C.A., Kraaijeveld C.A., van Strijp J.A., Brouwer E., Harmsen M., Verhoef J. Interactions of surfactant protein A with influenza A viruses: binding and neutralization. J Infect Dis. 1995;171:335–341. doi: 10.1093/infdis/171.2.335. [DOI] [PubMed] [Google Scholar]
- 31.Hartshorn K.L., Crouch E.C., White M.R., Eggleton P., Tauber A.I., Chang D. Evidence for a protective role of pulmonary surfactant protein D (SP-D) against influenza A viruses. J Clin Invest. 1994;94:311–319. doi: 10.1172/JCI117323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mikerov A.N., White M., Hartshorn K., Wang G., Floros J. Inhibition of hemagglutination activity of influenza A viruses by SP-A1 and SP-A2 variants expressed in CHO cells. Med Microbiol Immunol. 2008;197:9–12. doi: 10.1007/s00430-007-0051-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hartshorn K.L., White M.R., Shepherd V., Reid K., Jensenius J.C., Crouch E.C. Mechanisms of anti-influenza activity of surfactant proteins A and D: comparison with serum collectins. Am J Physiol. 1997;273:L1156–L1166. doi: 10.1152/ajplung.1997.273.6.L1156. [DOI] [PubMed] [Google Scholar]
- 34.Hartshorn K.L., Reid K.B., White M.R., Jensenius J.C., Morris S.M., Tauber A.I. Neutrophil deactivation by influenza A viruses: mechanisms of protection after viral opsonization with collectins and hemagglutination-inhibiting antibodies. Blood. 1996;87:3450–3461. [PubMed] [Google Scholar]
- 35.LeVine A.M., Whitsett J.A., Hartshorn K.L., Crouch E.C., Korfhagen T.R. Surfactant protein D enhances clearance of influenza A virus from the lung in vivo. J Immunol. 2001;167:5868–5873. doi: 10.4049/jimmunol.167.10.5868. [DOI] [PubMed] [Google Scholar]
- 36.Kingma P.S., Zhang L., Ikegami M., Hartshorn K., McCormack F.X., Whitsett J.A. Correction of pulmonary abnormalities in Sftpd-/- mice requires the collagenous domain of surfactant protein D. J Biol Chem. 2006;281:24496–24505. doi: 10.1074/jbc.M600651200. [DOI] [PubMed] [Google Scholar]
- 37.Kongchanagul A., Suptawiwat O., Boonarkart C., Kitphati R., Puthavathana P., Uiprasertkul M. Decreased expression of surfactant protein D mRNA in human lungs in fatal cases of H5N1 avian influenza. J Med Virol. 2011;83:1410–1417. doi: 10.1002/jmv.22105. [DOI] [PubMed] [Google Scholar]
- 38.To K.K.W., Zhou J., Song Y.Q., Hung I.F.N., Ip W.C.T., Cheng Z.S. Surfactant protein B gene polymorphism is associated with severe influenza. Chest. 2014;145:1237–1243. doi: 10.1378/chest.13-1651. [DOI] [PubMed] [Google Scholar]
- 39.Herrera-Ramos E., Lopez-Rodriguez M., Ruiz-Hernandez J.J., Horcajada J.P., Borderias L., Lerma E. Surfactant protein A genetic variants associate with severe respiratory insufficiency in pandemic influenza A virus infection. Crit Care. 2014;18:R127. doi: 10.1186/cc13934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lakadamyali M., Rust M.J., Zhuang X. Endocytosis of influenza viruses. Microb Infect. 2004;6:929–936. doi: 10.1016/j.micinf.2004.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Samji T. Influenza A: understanding the viral life cycle. Yale J Biol Med. 2009;82:153–159. [PMC free article] [PubMed] [Google Scholar]
- 42.Leung Y.H., Nicholls J.M., Ho C.K., Sia S.F., Mok C.K., Valkenburg S.A. Highly pathogenic avian influenza A H5N1 and pandemic H1N1 virus infections have different phenotypes in Toll-like receptor 3 knockout mice. J Gen Virol. 2014;95:1870–1879. doi: 10.1099/vir.0.066258-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Diebold S.S., Kaisho T., Hemmi H., Akira S., Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004;303:1529–1531. doi: 10.1126/science.1093616. [DOI] [PubMed] [Google Scholar]
- 44.Heil F., Hemmi H., Hochrein H., Ampenberger F., Kirschning C., Akira S. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004;303:1526–1529. doi: 10.1126/science.1093620. [DOI] [PubMed] [Google Scholar]
- 45.Kawai T., Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–384. doi: 10.1038/ni.1863. [DOI] [PubMed] [Google Scholar]
- 46.Seo S.U., Kwon H.J., Song J.H., Byun Y.H., Seong B.L., Kawai T. MyD88 signaling is indispensable for primary influenza A virus infection but dispensable for secondary infection. J Virol. 2010;84:12713–12722. doi: 10.1128/JVI.01675-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Iwasaki A., Pillai P.S. Innate immunity to influenza virus infection. Nat Rev Immunol. 2014;14:315–328. doi: 10.1038/nri3665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tsai S.Y., Segovia J.A., Chang T.H., Morris I.R., Berton M.T., Tessier P.A. DAMP molecule S100A9 acts as a molecular pattern to enhance inflammation during influenza A virus infection: role of DDX21-TRIF-TLR4-MyD88 pathway. PLoS Pathog. 2014;10:e1003848. doi: 10.1371/journal.ppat.1003848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Teijaro J.R., Walsh K.B., Rice S., Rosen H., Oldstone M.B. Mapping the innate signaling cascade essential for cytokine storm during influenza virus infection. Proc Natl Acad Sci U S A. 2014;111:3799–3804. doi: 10.1073/pnas.1400593111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang S.Y., Jouanguy E., Ugolini S., Smahi A., Elain G., Romero P. TLR3 deficiency in patients with herpes simplex encephalitis. Science. 2007;317:1522–1527. doi: 10.1126/science.1139522. [DOI] [PubMed] [Google Scholar]
- 51.Bernard J.J., Cowing-Zitron C., Nakatsuji T., Muehleisen B., Muto J., Borkowski A.W. Ultraviolet radiation damages self noncoding RNA and is detected by TLR3. Nat Med. 2012;18:1286–1290. doi: 10.1038/nm.2861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Takemura N., Kawasaki T., Kunisawa J., Sato S., Lamichhane A., Kobiyama K. Blockade of TLR3 protects mice from lethal radiation-induced gastrointestinal syndrome. Nat Commun. 2014;5:3492. doi: 10.1038/ncomms4492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Le Goffic R., Pothlichet J., Vitour D., Fujita T., Meurs E., Chignard M. Cutting Edge: influenza A virus activates TLR3-dependent inflammatory and RIG-I-dependent antiviral responses in human lung epithelial cells. J Immunol. 2007;178:3368–3372. doi: 10.4049/jimmunol.178.6.3368. [DOI] [PubMed] [Google Scholar]
- 54.Yoneyama M., Fujita T. Function of RIG-I-like receptors in antiviral innate immunity. J Biol Chem. 2007;282:15315–15318. doi: 10.1074/jbc.R700007200. [DOI] [PubMed] [Google Scholar]
- 55.Teng F., Slavik V., Duffy K.E., San Mateo L., Goldschmidt R. Toll-like receptor 3 is involved in airway epithelial cell response to nontypeable Haemophilus influenzae. Cell Immunol. 2010;260:98–104. doi: 10.1016/j.cellimm.2009.09.008. [DOI] [PubMed] [Google Scholar]
- 56.Guillot L., Le Goffic R., Bloch S., Escriou N., Akira S., Chignard M. Involvement of toll-like receptor 3 in the immune response of lung epithelial cells to double-stranded RNA and influenza A virus. J Biol Chem. 2005;280:5571–5580. doi: 10.1074/jbc.M410592200. [DOI] [PubMed] [Google Scholar]
- 57.Le Goffic R., Balloy V., Lagranderie M., Alexopoulou L., Escriou N., Flavell R. Detrimental contribution of the Toll-like receptor (TLR)3 to influenza A virus-induced acute pneumonia. PLoS Pathog. 2006;2:e53. doi: 10.1371/journal.ppat.0020053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Heer A.K., Shamshiev A., Donda A., Uematsu S., Akira S., Kopf M. TLR signaling fine-tunes anti-influenza B cell responses without regulating effector T cell responses. J Immunol. 2007;178:2182–2191. doi: 10.4049/jimmunol.178.4.2182. [DOI] [PubMed] [Google Scholar]
- 59.Pang I.K., Pillai P.S., Iwasaki A. Efficient influenza A virus replication in the respiratory tract requires signals from TLR7 and RIG-I. Proc Natl Acad Sci U S A. 2013;110:13910–13915. doi: 10.1073/pnas.1303275110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Sykes A., Edwards M.R., Macintyre J., Del Rosario A., Gielen V., Haas J. TLR3, TLR4 and TLRs7-9 induced interferons are not impaired in airway and blood cells in well controlled asthma. PLoS One. 2013;8:e65921. doi: 10.1371/journal.pone.0065921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ioannidis I., Ye F., McNally B., Willette M., Flano E. Toll-like receptor expression and induction of type I and type III interferons in primary airway epithelial cells. J Virol. 2013;87:3261–3270. doi: 10.1128/JVI.01956-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Jeisy-Scott V., Kim J.H., Davis W.G., Cao W., Katz J.M., Sambhara S. TLR7 recognition is dispensable for influenza virus A infection but important for the induction of hemagglutinin-specific antibodies in response to the 2009 pandemic split vaccine in mice. J Virol. 2012;86:10988–10998. doi: 10.1128/JVI.01064-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pillai P.S., Molony R.D., Martinod K., Dong H., Pang I.K., Tal M.C. Mx1 reveals innate pathways to antiviral resistance and lethal influenza disease. Science. 2016;352:463–466. doi: 10.1126/science.aaf3926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yoneyama M., Kikuchi M., Natsukawa T., Shinobu N., Imaizumi T., Miyagishi M. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004;5:730–737. doi: 10.1038/ni1087. [DOI] [PubMed] [Google Scholar]
- 65.Liu G., Park H.S., Pyo H.M., Liu Q., Zhou Y. Influenza a virus panhandle structure is directly involved in RIG-I activation and interferon induction. J Virol. 2015;89:6067–6079. doi: 10.1128/JVI.00232-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Meylan E., Tschopp J., Karin M. Intracellular pattern recognition receptors in the host response. Nature. 2006;442:39–44. doi: 10.1038/nature04946. [DOI] [PubMed] [Google Scholar]
- 67.Slater L., Bartlett N.W., Haas J.J., Zhu J., Message S.D., Walton R.P. Co-ordinated role of TLR3, RIG-I and MDA5 in the innate response to rhinovirus in bronchial epithelium. PLoS Pathog. 2010;6:e1001178. doi: 10.1371/journal.ppat.1001178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yoneyama M., Fujita T. RNA recognition and signal transduction by RIG-I-like receptors. Immunol Rev. 2009;227:54–65. doi: 10.1111/j.1600-065X.2008.00727.x. [DOI] [PubMed] [Google Scholar]
- 69.Poeck H., Bscheider M., Gross O., Finger K., Roth S., Rebsamen M. Recognition of RNA virus by RIG-I results in activation of CARD9 and inflammasome signaling for interleukin 1 beta production. Nat Immunol. 2010;11:63–69. doi: 10.1038/ni.1824. [DOI] [PubMed] [Google Scholar]
- 70.Rintahaka J., Wiik D., Kovanen P.E., Alenius H., Matikainen S. Cytosolic antiviral RNA recognition pathway activates caspases 1 and 3. J Immunol. 2008;180:1749–1757. doi: 10.4049/jimmunol.180.3.1749. [DOI] [PubMed] [Google Scholar]
- 71.Opitz B., Rejaibi A., Dauber B., Eckhard J., Vinzing M., Schmeck B. IFNbeta induction by influenza A virus is mediated by RIG-I which is regulated by the viral NS1 protein. Cell Microbiol. 2007;9:930–938. doi: 10.1111/j.1462-5822.2006.00841.x. [DOI] [PubMed] [Google Scholar]
- 72.Kato H., Takeuchi O., Sato S., Yoneyama M., Yamamoto M., Matsui K. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441:101–105. doi: 10.1038/nature04734. [DOI] [PubMed] [Google Scholar]
- 73.Kandasamy M., Suryawanshi A., Tundup S., Perez J.T., Schmolke M., Manicassamy S. RIG-I signaling is critical for efficient polyfunctional T cell responses during influenza virus infection. PLoS Pathog. 2016;12:e1005754. doi: 10.1371/journal.ppat.1005754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Benitez A.A., Panis M., Xue J., Varble A., Shim J.V., Frick A.L. In vivo RNAi screening identifies MDA5 as a significant contributor to the cellular defense against influenza a virus. Cell Rep. 2015;11:1714–1726. doi: 10.1016/j.celrep.2015.05.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Crotta S., Davidson S., Mahlakoiv T., Desmet C.J., Buckwalter M.R., Albert M.L. Type I and type III interferons drive redundant amplification loops to induce a transcriptional signature in influenza-infected airway epithelia. PLoS Pathog. 2013;9:e1003773. doi: 10.1371/journal.ppat.1003773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pothlichet J., Meunier I., Davis B.K., Ting J.P., Skamene E., von Messling V. Type I IFN triggers RIG-I/TLR3/NLRP3-dependent inflammasome activation in influenza A virus infected cells. PLoS Pathog. 2013;9:e1003256. doi: 10.1371/journal.ppat.1003256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ichinohe T., Lee H.K., Ogura Y., Flavell R., Iwasaki A. Inflammasome recognition of influenza virus is essential for adaptive immune responses. J Exp Med. 2009;206:79–87. doi: 10.1084/jem.20081667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Subramanian N., Natarajan K., Clatworthy M.R., Wang Z., Germain R.N. The adaptor MAVS promotes NLRP3 mitochondrial localization and inflammasome activation. Cell. 2013;153:348–361. doi: 10.1016/j.cell.2013.02.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chen G., Shaw M.H., Kim Y.G., Nunez G. NOD-like receptors: role in innate immunity and inflammatory disease. Annu Rev Pathol. 2009;4:365–398. doi: 10.1146/annurev.pathol.4.110807.092239. [DOI] [PubMed] [Google Scholar]
- 80.Kanneganti T.D., Body-Malapel M., Amer A., Park J.H., Whitfield J., Franchi L. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem. 2006;281:36560–36568. doi: 10.1074/jbc.M607594200. [DOI] [PubMed] [Google Scholar]
- 81.Sutterwala F.S., Ogura Y., Szczepanik M., Lara-Tejero M., Lichtenberger G.S., Grant E.P. Critical role for NALP3/CIAS1/Cryopyrin in innate and adaptive immunity through its regulation of caspase-1. Immunity. 2006;24:317–327. doi: 10.1016/j.immuni.2006.02.004. [DOI] [PubMed] [Google Scholar]
- 82.Mariathasan S., Weiss D.S., Newton K., McBride J., O'Rourke K., Roose-Girma M. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–232. doi: 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
- 83.Allen I.C., Scull M.A., Moore C.B., Holl E.K., McElvania-TeKippe E., Taxman D.J. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 2009;30:556–565. doi: 10.1016/j.immuni.2009.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Tran H.B., Lewis M.D., Tan L.W., Lester S.E., Baker L.M., Ng J. Immunolocalization of NLRP3 inflammasome in normal murine airway epithelium and changes following induction of ovalbumin-induced airway inflammation. J Allergy. 2012;2012:819176. doi: 10.1155/2012/819176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kostadinova E., Chaput C., Gutbier B., Lippmann J., Sander L.E., Mitchell T.J. NLRP3 protects alveolar barrier integrity by an inflammasome-independent increase of epithelial cell adherence. Sci Rep. 2016;6:30943. doi: 10.1038/srep30943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Fink S.L., Cookson B.T. Apoptosis, pyroptosis, and necrosis: mechanistic description of dead and dying eukaryotic cells. Infect Immun. 2005;73:1907–1916. doi: 10.1128/IAI.73.4.1907-1916.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gallucci S., Matzinger P. Danger signals: SOS to the immune system. Curr Opin Immunol. 2001;13:114–119. doi: 10.1016/s0952-7915(00)00191-6. [DOI] [PubMed] [Google Scholar]
- 88.Trautmann A. Extracellular ATP in the immune system: more than just a "danger signal". Sci Signal. 2009;2:pe6. doi: 10.1126/scisignal.256pe6. [DOI] [PubMed] [Google Scholar]
- 89.Bianchi M.E. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81:1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
- 90.van Zoelen M.A., van der Sluijs K.F., Achouiti A., Florquin S., Braun-Pater J.M., Yang H. Receptor for advanced glycation end products is detrimental during influenza A virus pneumonia. Virology. 2009;391:265–273. doi: 10.1016/j.virol.2009.05.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Stasakova J., Ferko B., Kittel C., Sereinig S., Romanova J., Katinger H. Influenza A mutant viruses with altered NS1 protein function provoke caspase-1 activation in primary human macrophages, resulting in fast apoptosis and release of high levels of interleukins 1beta and 18. J Gen Virol. 2005;86:185–195. doi: 10.1099/vir.0.80422-0. [DOI] [PubMed] [Google Scholar]
- 92.Lam W.Y., Tang J.W., Yeung A.C., Chiu L.C., Sung J.J., Chan P.K. Avian influenza virus A/HK/483/97(H5N1) NS1 protein induces apoptosis in human airway epithelial cells. J Virol. 2008;82:2741–2751. doi: 10.1128/JVI.01712-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Asselin-Paturel C., Boonstra A., Dalod M., Durand I., Yessaad N., Dezutter-Dambuyant C. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat Immunol. 2001;2:1144–1150. doi: 10.1038/ni736. [DOI] [PubMed] [Google Scholar]
- 94.Donnelly R.P., Kotenko S.V. Interferon-lambda: a new addition to an old family. J Interferon Cytokine Res. 2010;30:555–564. doi: 10.1089/jir.2010.0078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Lazear H.M., Nice T.J., Diamond M.S. Interferon-lambda: immune functions at barrier surfaces and beyond. Immunity. 2015;43:15–28. doi: 10.1016/j.immuni.2015.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wack A., Terczynska-Dyla E., Hartmann R. Guarding the frontiers: the biology of type III interferons. Nat Immunol. 2015;16:802–809. doi: 10.1038/ni.3212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.McNab F., Mayer-Barber K., Sher A., Wack A., O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol. 2015;15:87–103. doi: 10.1038/nri3787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Garcia-Sastre A., Biron C.A. Type 1 interferons and the virus-host relationship: a lesson in detente. Science. 2006;312:879–882. doi: 10.1126/science.1125676. [DOI] [PubMed] [Google Scholar]
- 99.Cheon H., Holvey-Bates E.G., Schoggins J.W., Forster S., Hertzog P., Imanaka N. IFNbeta-dependent increases in STAT1, STAT2, and IRF9 mediate resistance to viruses and DNA damage. EMBO J. 2013;32:2751–2763. doi: 10.1038/emboj.2013.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kim H.J., Kim C.H., Ryu J.H., Kim M.J., Park C.Y., Lee J.M. Reactive oxygen species induce antiviral innate immune response through IFN-lambda regulation in human nasal epithelial cells. Am J Respir Cell Mol Biol. 2013;49:855–865. doi: 10.1165/rcmb.2013-0003OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sheppard P., Kindsvogel W., Xu W., Henderson K., Schlutsmeyer S., Whitmore T.E. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol. 2003;4:63–68. doi: 10.1038/ni873. [DOI] [PubMed] [Google Scholar]
- 102.Yao Q., Fischer K.P., Arnesen K., Tyrrell D.L., Gutfreund K.S. Molecular cloning, expression and characterization of Pekin duck interferon-lambda. Gene. 2014;548:29–38. doi: 10.1016/j.gene.2014.06.066. [DOI] [PubMed] [Google Scholar]
- 103.Galani I.E., Triantafyllia V., Eleminiadou E.E., Koltsida O., Stavropoulos A., Manioudaki M. Interferon-lambda mediates non-redundant front-line antiviral protection against influenza virus infection without compromising host fitness. Immunity. 2017;46 doi: 10.1016/j.immuni.2017.04.025. 875–90.e6. [DOI] [PubMed] [Google Scholar]
- 104.Berry C.M. Understanding interferon subtype therapy for viral infections: harnessing the power of the innate immune system. Cytokine Growth Factor Rev. 2016;31:83–90. doi: 10.1016/j.cytogfr.2016.08.001. [DOI] [PubMed] [Google Scholar]
- 105.Sheahan T., Imanaka N., Marukian S., Dorner M., Liu P., Ploss A. Interferon lambda alleles predict innate antiviral immune responses and hepatitis C virus permissiveness. Cell Host Microbe. 2014;15:190–202. doi: 10.1016/j.chom.2014.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Nice T.J., Baldridge M.T., McCune B.T., Norman J.M., Lazear H.M., Artyomov M. Interferon-lambda cures persistent murine norovirus infection in the absence of adaptive immunity. Science. 2015;347:269–273. doi: 10.1126/science.1258100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wack A. Interfering with transmission. Elife. 2018;7 doi: 10.7554/eLife.37552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Klinkhammer J., Schnepf D., Ye L., Schwaderlapp M., Gad H.H., Hartmann R. IFN-lambda prevents influenza virus spread from the upper airways to the lungs and limits virus transmission. Elife. 2018;7 doi: 10.7554/eLife.33354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Davidson S., Crotta S., McCabe T.M., Wack A. Pathogenic potential of interferon alphabeta in acute influenza infection. Nat Commun. 2014;5:3864. doi: 10.1038/ncomms4864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Khaitov M.R., Laza-Stanca V., Edwards M.R., Walton R.P., Rohde G., Contoli M. Respiratory virus induction of alpha-, beta- and lambda-interferons in bronchial epithelial cells and peripheral blood mononuclear cells. Allergy. 2009;64:375–386. doi: 10.1111/j.1398-9995.2008.01826.x. [DOI] [PubMed] [Google Scholar]
- 111.Wang J., Oberley-Deegan R., Wang S., Nikrad M., Funk C.J., Hartshorn K.L. Differentiated human alveolar type II cells secrete antiviral IL-29 (IFN-lambda 1) in response to influenza A infection. J Immunol. 2009;182:1296–1304. doi: 10.4049/jimmunol.182.3.1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Jewell N.A., Cline T., Mertz S.E., Smirnov S.V., Flano E., Schindler C. Lambda interferon is the predominant interferon induced by influenza A virus infection in vivo. J Virol. 2010;84:11515–11522. doi: 10.1128/JVI.01703-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Dumoutier L., Tounsi A., Michiels T., Sommereyns C., Kotenko S.V., Renauld J.C. Role of the interleukin (IL)-28 receptor tyrosine residues for antiviral and antiproliferative activity of IL-29/interferon-lambda 1 – similarities with type I interferon signaling. J Biol Chem. 2004;279:32269–32274. doi: 10.1074/jbc.M404789200. [DOI] [PubMed] [Google Scholar]
- 114.Mordstein M., Neugebauer E., Ditt V., Jessen B., Rieger T., Falcone V. Lambda interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections. J Virol. 2010;84:5670–5677. doi: 10.1128/JVI.00272-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Platanias L.C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol. 2005;5:375–386. doi: 10.1038/nri1604. [DOI] [PubMed] [Google Scholar]
- 116.Bailey C.C., Huang I.C., Kam C., Farzan M. Ifitm3 limits the severity of acute influenza in mice. PLoS Pathog. 2012;8:e1002909. doi: 10.1371/journal.ppat.1002909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Everitt A.R., Clare S., Pertel T., John S.P., Wash R.S., Smith S.E. IFITM3 restricts the morbidity and mortality associated with influenza. Nature. 2012;484:519–523. doi: 10.1038/nature10921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zhang Y.H., Zhao Y., Li N., Peng Y.C., Giannoulatou E., Jin R.H. Interferon-induced transmembrane protein-3 genetic variant rs12252-C is associated with severe influenza in Chinese individuals. Nat Commun. 2013;4:1418. doi: 10.1038/ncomms2433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Krug R.M., Shaw M., Broni B., Shapiro G., Haller O. Inhibition of influenza viral mRNA synthesis in cells expressing the interferon-induced Mx gene product. J Virol. 1985;56:201–206. doi: 10.1128/jvi.56.1.201-206.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lindenmann J., Lane C.A., Hobson D. The resistance of A2g mice to myxoviruses. J Immunol. 1963;90:942–951. [PubMed] [Google Scholar]
- 121.Staeheli P., Haller O., Boll W., Lindenmann J., Weissmann C. Mx protein: constitutive expression in 3T3 cells transformed with cloned Mx cDNA confers selective resistance to influenza virus. Cell. 1986;44:147–158. doi: 10.1016/0092-8674(86)90493-9. [DOI] [PubMed] [Google Scholar]
- 122.Lindenmann J. Resistance of mice to mouse-adapted influenza A virus. Virology. 1962;16:203–204. doi: 10.1016/0042-6822(62)90297-0. [DOI] [PubMed] [Google Scholar]
- 123.Ronni T., Sareneva T., Pirhonen J., Julkunen I. Activation of IFN-alpha, IFN-gamma, MxA, and IFN regulatory factor 1 genes in influenza A virus-infected human peripheral blood mononuclear cells. J Immunol. 1995;154:2764–2774. [PubMed] [Google Scholar]
- 124.Manz B., Dornfeld D., Gotz V., Zell R., Zimmermann P., Haller O. Pandemic influenza A viruses escape from restriction by human MxA through adaptive mutations in the nucleoprotein. PLoS Pathog. 2013;9:e1003279. doi: 10.1371/journal.ppat.1003279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Xiao H., Killip M.J., Staeheli P., Randall R.E., Jackson D. The human interferon-induced MxA protein inhibits early stages of influenza A virus infection by retaining the incoming viral genome in the cytoplasm. J Virol. 2013;87:13053–13058. doi: 10.1128/JVI.02220-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zimmermann P., Manz B., Haller O., Schwemmle M., Kochs G. The viral nucleoprotein determines Mx sensitivity of influenza A viruses. J Virol. 2011;85:8133–8140. doi: 10.1128/JVI.00712-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Staeheli P., Grob R., Meier E., Sutcliffe J.G., Haller O. Influenza virus-susceptible mice carry Mx genes with a large deletion or a nonsense mutation. Mol Cell Biol. 1988;8:4518–4523. doi: 10.1128/mcb.8.10.4518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Staeheli P., Pravtcheva D., Lundin L.G., Acklin M., Ruddle F., Lindenmann J. Interferon-regulated influenza virus resistance gene Mx is localized on mouse chromosome 16. J Virol. 1986;58:967–969. doi: 10.1128/jvi.58.3.967-969.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Khaperskyy D.A., Emara M.M., Johnston B.P., Anderson P., Hatchette T.F., McCormick C. Influenza a virus host shutoff disables antiviral stress-induced translation arrest. PLoS Pathog. 2014;10:e1004217. doi: 10.1371/journal.ppat.1004217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Gil J., Garcia M.A., Gomez-Puertas P., Guerra S., Rullas J., Nakano H. TRAF family proteins link PKR with NF-kappa B activation. Mol Cell Biol. 2004;24:4502–4512. doi: 10.1128/MCB.24.10.4502-4512.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Onomoto K., Jogi M., Yoo J.S., Narita R., Morimoto S., Takemura A. Critical role of an antiviral stress granule containing RIG-I and PKR in viral detection and innate immunity. PLoS One. 2012;7:e43031. doi: 10.1371/journal.pone.0043031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Hamamoto I., Takaku H., Tashiro M., Yamamoto N. High yield production of influenza virus in Madin Darby canine kidney (MDCK) cells with stable knockdown of IRF7. PLoS One. 2013;8:e59892. doi: 10.1371/journal.pone.0059892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Colina R., Costa-Mattioli M., Dowling R.J., Jaramillo M., Tai L.H., Breitbach C.J. Translational control of the innate immune response through IRF-7. Nature. 2008;452:323–328. doi: 10.1038/nature06730. [DOI] [PubMed] [Google Scholar]
- 134.Ciancanelli M.J., Huang S.X., Luthra P., Garner H., Itan Y., Volpi S. Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science. 2015;348:448–453. doi: 10.1126/science.aaa1578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Koyama S., Ishii K.J., Kumar H., Tanimoto T., Coban C., Uematsu S. Differential role of TLR- and RLR-signaling in the immune responses to influenza A virus infection and vaccination. J Immunol. 2007;179:4711–4720. doi: 10.4049/jimmunol.179.7.4711. [DOI] [PubMed] [Google Scholar]
- 136.Xing Z., Harper R., Anunciacion J., Yang Z., Gao W., Qu B. Host immune and apoptotic responses to avian influenza virus H9N2 in human tracheobronchial epithelial cells. Am J Respir Cell Mol Biol. 2011;44:24–33. doi: 10.1165/rcmb.2009-0120OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Chan M.C., Cheung C.Y., Chui W.H., Tsao S.W., Nicholls J.M., Chan Y.O. Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respir Res. 2005;6:135. doi: 10.1186/1465-9921-6-135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chan M.C., Chan R.W., Yu W.C., Ho C.C., Yuen K.M., Fong J.H. Tropism and innate host responses of the 2009 pandemic H1N1 influenza virus in ex vivo and in vitro cultures of human conjunctiva and respiratory tract. Am J Pathol. 2010;176:1828–1840. doi: 10.2353/ajpath.2010.091087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Lam W.Y., Yeung A.C., Chu I.M., Chan P.K. Profiles of cytokine and chemokine gene expression in human pulmonary epithelial cells induced by human and avian influenza viruses. Virol J. 2010;7:344. doi: 10.1186/1743-422X-7-344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Denney L., Branchett W., Gregory L.G., Oliver R.A., Lloyd C.M. Epithelial-derived TGF-beta1 acts as a pro-viral factor in the lung during influenza A infection. Mucosal Immunol. 2018;11:523–535. doi: 10.1038/mi.2017.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Sanders C.J., Doherty P.C., Thomas P.G. Respiratory epithelial cells in innate immunity to influenza virus infection. Cell Tissue Res. 2011;343:13–21. doi: 10.1007/s00441-010-1043-z. [DOI] [PubMed] [Google Scholar]
- 142.Peiro T., Patel D.F., Akthar S., Gregory L.G., Pyle C.J., Harker J.A. Neutrophils drive alveolar macrophage IL-1beta release during respiratory viral infection. Thorax. 2018;73:546–556. doi: 10.1136/thoraxjnl-2017-210010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kanneganti T.D. Central roles of NLRs and inflammasomes in viral infection. Nat Rev Immunol. 2010;10:688–698. doi: 10.1038/nri2851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Thomas P.G., Dash P., Aldridge J.R., Jr., Ellebedy A.H., Reynolds C., Funk A.J. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 2009;30:566–575. doi: 10.1016/j.immuni.2009.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Tamaru M., Tomura K., Sakamoto S., Tezuka K., Tamatani T., Narumi S. Interleukin-1beta induces tissue- and cell type-specific expression of adhesion molecules in vivo. Arterioscler Thromb Vasc Biol. 1998;18:1292–1303. doi: 10.1161/01.atv.18.8.1292. [DOI] [PubMed] [Google Scholar]
- 146.Dinarello C.A. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–550. doi: 10.1146/annurev.immunol.021908.132612. [DOI] [PubMed] [Google Scholar]
- 147.Schmitz N., Kurrer M., Bachmann M.F., Kopf M. Interleukin-1 is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. J Virol. 2005;79:6441–6448. doi: 10.1128/JVI.79.10.6441-6448.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hennet T., Ziltener H.J., Frei K., Peterhans E. A kinetic study of immune mediators in the lungs of mice infected with influenza A virus. J Immunol. 1992;149:932–939. [PubMed] [Google Scholar]
- 149.Longhi M.P., Wright K., Lauder S.N., Nowell M.A., Jones G.W., Godkin A.J. Interleukin-6 is crucial for recall of influenza-specific memory CD4 T cells. PLoS Pathog. 2008;4:e1000006. doi: 10.1371/journal.ppat.1000006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Dienz O., Rud J.G., Eaton S.M., Lanthier P.A., Burg E., Drew A. Essential role of IL-6 in protection against H1N1 influenza virus by promoting neutrophil survival in the lung. Mucosal Immunol. 2012;5:258–266. doi: 10.1038/mi.2012.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lee S.W., Youn J.W., Seong B.L., Sung Y.C. IL-6 induces long-term protective immunity against a lethal challenge of influenza virus. Vaccine. 1999;17:490–496. doi: 10.1016/s0264-410x(98)00223-0. [DOI] [PubMed] [Google Scholar]
- 152.Dienz O., Eaton S.M., Bond J.P., Neveu W., Moquin D., Noubade R. The induction of antibody production by IL-6 is indirectly mediated by IL-21 produced by CD4+ T cells. J Exp Med. 2009;206:69–78. doi: 10.1084/jem.20081571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kaiser L., Fritz R.S., Straus S.E., Gubareva L., Hayden F.G. Symptom pathogenesis during acute influenza: interleukin-6 and other cytokine responses. J Med Virol. 2001;64:262–268. doi: 10.1002/jmv.1045. [DOI] [PubMed] [Google Scholar]
- 154.Hagau N., Slavcovici A., Gonganau D.N., Oltean S., Dirzu D.S., Brezoszki E.S. Clinical aspects and cytokine response in severe H1N1 influenza A virus infection. Crit Care. 2010;14:R203. doi: 10.1186/cc9324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Yang M.L., Wang C.T., Yang S.J., Leu C.H., Chen S.H., Wu C.L. IL-6 ameliorates acute lung injury in influenza virus infection. Sci Rep. 2017;7:43829. doi: 10.1038/srep43829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Herold S., von Wulffen W., Steinmueller M., Pleschka S., Kuziel W.A., Mack M. Alveolar epithelial cells direct monocyte transepithelial migration upon influenza virus infection: impact of chemokines and adhesion molecules. J Immunol. 2006;177:1817–1824. doi: 10.4049/jimmunol.177.3.1817. [DOI] [PubMed] [Google Scholar]
- 157.Perrone L.A., Plowden J.K., Garcia-Sastre A., Katz J.M., Tumpey T.M. H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice. PLoS Pathog. 2008;4:e1000115. doi: 10.1371/journal.ppat.1000115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Kok W.L., Denney L., Benam K., Cole S., Clelland C., McMichael A.J. Pivotal Advance: invariant NKT cells reduce accumulation of inflammatory monocytes in the lungs and decrease immune-pathology during severe influenza A virus infection. J Leukoc Biol. 2012;91:357–368. doi: 10.1189/jlb.0411184. [DOI] [PubMed] [Google Scholar]
- 159.Cole S.L., Dunning J., Kok W.L., Benam K.H., Benlahrech A., Repapi E. M1-like monocytes are a major immunological determinant of severity in previously healthy adults with life-threatening influenza. JCI Insight. 2017;2:e91868. doi: 10.1172/jci.insight.91868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Dawson T.C., Beck M.A., Kuziel W.A., Henderson F., Maeda N. Contrasting effects of CCR5 and CCR2 deficiency in the pulmonary inflammatory response to influenza A virus. Am J Pathol. 2000;156:1951–1959. doi: 10.1016/S0002-9440(10)65068-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lin K.L., Suzuki Y., Nakano H., Ramsburg E., Gunn M.D. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J Immunol. 2008;180:2562–2572. doi: 10.4049/jimmunol.180.4.2562. [DOI] [PubMed] [Google Scholar]
- 162.Lin S.J., Lo M., Kuo R.L., Shih S.R., Ojcius D.M., Lu J. The pathological effects of CCR2+ inflammatory monocytes are amplified by an IFNAR1-triggered chemokine feedback loop in highly pathogenic influenza infection. J Biomed Sci. 2014;21:99. doi: 10.1186/s12929-014-0099-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Voudoukis E., Pappas A., Panoutsopoulos A., Xynos K., Rozi F., Giannakopoulou K. Novel influenza A (H1N1) infection vs. common influenza-like illness: a prospective study. Med Sci Monit. 2011;17:CR185–CR188. doi: 10.12659/MSM.881713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Giamarellos-Bourboulis E.J., Raftogiannis M., Antonopoulou A., Baziaka F., Koutoukas P., Savva A. Effect of the novel influenza A (H1N1) virus in the human immune system. PLoS One. 2009;4:e8393. doi: 10.1371/journal.pone.0008393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Gill M.A., Long K., Kwon T., Muniz L., Mejias A., Connolly J. Differential recruitment of dendritic cells and monocytes to respiratory mucosal sites in children with influenza virus or respiratory syncytial virus infection. J Infect Dis. 2008;198:1667–1676. doi: 10.1086/593018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Oshansky C.M., Gartland A.J., Wong S.S., Jeevan T., Wang D., Roddam P.L. Mucosal immune responses predict clinical outcomes during influenza infection independently of age and viral load. Am J Respir Crit Care Med. 2014;189:449–462. doi: 10.1164/rccm.201309-1616OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Schultz-Cherry S., Hinshaw V.S. Influenza virus neuraminidase activates latent transforming growth factor beta. J Virol. 1996;70:8624–8629. doi: 10.1128/jvi.70.12.8624-8629.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Gibbs J.D., Ornoff D.M., Igo H.A., Zeng J.Y., Imani F. Cell cycle arrest by transforming growth factor beta1 enhances replication of respiratory syncytial virus in lung epithelial cells. J Virol. 2009;83:12424–12431. doi: 10.1128/JVI.00806-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Carlson C.M., Turpin E.A., Moser L.A., O'Brien K.B., Cline T.D., Jones J.C. Transforming growth factor-beta: activation by neuraminidase and role in highly pathogenic H5N1 influenza pathogenesis. PLoS Pathog. 2010;6:e1001136. doi: 10.1371/journal.ppat.1001136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Williams A.E., Humphreys I.R., Cornere M., Edwards L., Rae A., Hussell T. TGF-beta prevents eosinophilic lung disease but impairs pathogen clearance. Microb Infect. 2005;7:365–374. doi: 10.1016/j.micinf.2004.11.012. [DOI] [PubMed] [Google Scholar]
- 171.Lambrecht B.N., Hammad H. Biology of lung dendritic cells at the origin of asthma. Immunity. 2009;31:412–424. doi: 10.1016/j.immuni.2009.08.008. [DOI] [PubMed] [Google Scholar]
- 172.Kotenko S.V., Durbin J.E. Contribution of type III interferons to antiviral immunity: location, location, location. J Biol Chem. 2017;292:7295–7303. doi: 10.1074/jbc.R117.777102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Condon T.V., Sawyer R.T., Fenton M.J., Riches D.W. Lung dendritic cells at the innate-adaptive immune interface. J Leukoc Biol. 2011;90:883–895. doi: 10.1189/jlb.0311134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Kim T.H., Lee H.K. Differential roles of lung dendritic cell subsets against respiratory virus infection. Immune Network. 2014;14:128–137. doi: 10.4110/in.2014.14.3.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.de Bree G.J., van Leeuwen E.M., Out T.A., Jansen H.M., Jonkers R.E., van Lier R.A. Selective accumulation of differentiated CD8+ T cells specific for respiratory viruses in the human lung. J Exp Med. 2005;202:1433–1442. doi: 10.1084/jem.20051365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Godfrey D.I., Stankovic S., Baxter A.G. Raising the NKT cell family. Nat Immunol. 2010;11:197–206. doi: 10.1038/ni.1841. [DOI] [PubMed] [Google Scholar]
- 177.Scanlon S.T., Thomas S.Y., Ferreira C.M., Bai L., Krausz T., Savage P.B. Airborne lipid antigens mobilize resident intravascular NKT cells to induce allergic airway inflammation. J Exp Med. 2011;208:2113–2124. doi: 10.1084/jem.20110522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Ho L.P., Denney L., Luhn K., Teoh D., Clelland C., McMichael A.J. Activation of invariant NKT cells enhances the innate immune response and improves the disease course in influenza A virus infection. Eur J Immunol. 2008;38:1913–1922. doi: 10.1002/eji.200738017. [DOI] [PubMed] [Google Scholar]
- 179.Benam K.H., Kok W.L., McMichael A.J., Ho L.P. Alternative spliced CD1d transcripts in human bronchial epithelial cells. PLoS One. 2011;6:e22726. doi: 10.1371/journal.pone.0022726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Rizvi Z.A., Puri N., Saxena R.K. Lipid antigen presentation through CD1d pathway in mouse lung epithelial cells, macrophages and dendritic cells and its suppression by poly-dispersed single-walled carbon nanotubes. Toxicol In Vitro. 2015;29:1275–1282. doi: 10.1016/j.tiv.2014.10.022. [DOI] [PubMed] [Google Scholar]
- 181.Martinez-Lopez M., Iborra S., Conde-Garrosa R., Sancho D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur J Immunol. 2015;45:119–129. doi: 10.1002/eji.201444651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Tyznik A.J., Tupin E., Nagarajan N.A., Her M.J., Benedict C.A., Kronenberg M. Cutting edge: the mechanism of invariant NKT cell responses to viral danger signals. J Immunol. 2008;181:4452–4456. doi: 10.4049/jimmunol.181.7.4452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Reilly E.C., Wands J.R., Brossay L. Cytokine dependent and independent iNKT cell activation. Cytokine. 2010;51:227–231. doi: 10.1016/j.cyto.2010.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Gaya M., Barral P., Burbage M., Aggarwal S., Montaner B., Warren Navia A. Initiation of antiviral B cell immunity relies on innate signals from spatially positioned NKT cells. Cell. 2018;172:517–533. doi: 10.1016/j.cell.2017.11.036. e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.van Wilgenburg B., Scherwitzl I., Hutchinson E.C., Leng T., Kurioka A., Kulicke C. MAIT cells are activated during human viral infections. Nat Commun. 2016;7:11653. doi: 10.1038/ncomms11653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Le Goffic R., Arshad M.I., Rauch M., L'Helgoualc'h A., Delmas B., Piquet-Pellorce C. Infection with influenza virus induces IL-33 in murine lungs. Am J Respir Cell Mol Biol. 2011;45:1125–1132. doi: 10.1165/rcmb.2010-0516OC. [DOI] [PubMed] [Google Scholar]
- 187.Monticelli L.A., Sonnenberg G.F., Abt M.C., Alenghat T., Ziegler C.G., Doering T.A. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat Immunol. 2011;12:1045–1054. doi: 10.1031/ni.2131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Fuchs A. ILC1s in tissue inflammation and infection. Front Immunol. 2016;7:104. doi: 10.3389/fimmu.2016.00104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Leavy O. Innate-like lymphocytes: will the real ILC1 please stand up? Nat Rev Immunol. 2013;13:67. doi: 10.1038/nri3397. [DOI] [PubMed] [Google Scholar]
- 190.Wang J., Li F., Zheng M., Sun R., Wei H., Tian Z. Lung natural killer cells in mice: phenotype and response to respiratory infection. Immunology. 2012;137:37–47. doi: 10.1111/j.1365-2567.2012.03607.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Brydon E.W., Smith H., Sweet C. Influenza A virus-induced apoptosis in bronchiolar epithelial (NCI-H292) cells limits pro-inflammatory cytokine release. J Gen Virol. 2003;84:2389–2400. doi: 10.1099/vir.0.18913-0. [DOI] [PubMed] [Google Scholar]
- 192.Arndt U., Wennemuth G., Barth P., Nain M., Al-Abed Y., Meinhardt A. Release of macrophage migration inhibitory factor and CXCL8/interleukin-8 from lung epithelial cells rendered necrotic by influenza A virus infection. J Virol. 2002;76:9298–9306. doi: 10.1128/JVI.76.18.9298-9306.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Folkesson H.G., Matthay M.A. Alveolar epithelial ion and fluid transport: recent progress. Am J Respir Cell Mol Biol. 2006;35:10–19. doi: 10.1165/rcmb.2006-0080SF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Berthiaume Y., Matthay M.A. Alveolar edema fluid clearance and acute lung injury. Respir Physiol Neurobiol. 2007;159:350–359. doi: 10.1016/j.resp.2007.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Chen X.J., Seth S., Yue G., Kamat P., Compans R.W., Guidot D. Influenza virus inhibits ENaC and lung fluid clearance. Am J Physiol Lung Cell Mol Physiol. 2004;287:L366–L373. doi: 10.1152/ajplung.00011.2004. [DOI] [PubMed] [Google Scholar]
- 196.Lazrak A., Iles K.E., Liu G., Noah D.L., Noah J.W., Matalon S. Influenza virus M2 protein inhibits epithelial sodium channels by increasing reactive oxygen species. FASEB J. 2009;23:3829–3842. doi: 10.1096/fj.09-135590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Louria D.B., Blumenfeld H.L., Ellis J.T., Kilbourne E.D., Rogers D.E. Studies on influenza in the pandemic of 1957-1958. II. Pulmonary complications of influenza. J Clin Invest. 1959;38:213–265. doi: 10.1172/JCI103791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Hers J.F., Masurel N., Mulder J. Bacteriology and histopathology of the respiratory tract and lungs in fatal Asian influenza. Lancet. 1958;2:1141–1143. doi: 10.1016/s0140-6736(58)92404-8. [DOI] [PubMed] [Google Scholar]
- 199.Gill J.R., Sheng Z.M., Ely S.F., Guinee D.G., Beasley M.B., Suh J. Pulmonary pathologic findings of fatal 2009 pandemic influenza A/H1N1 viral infections. Arch Pathol Lab Med. 2010;134:235–243. doi: 10.5858/134.2.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Alymova I.V., Portner A., Takimoto T., Boyd K.L., Babu Y.S., McCullers J.A. The novel parainfluenza virus hemagglutinin-neuraminidase inhibitor BCX 2798 prevents lethal synergism between a paramyxovirus and Streptococcus pneumoniae. Antimicrob Agents Chemother. 2005;49:398–405. doi: 10.1128/AAC.49.1.398-405.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Jakab G.J. Mechanisms of bacterial superinfections in viral pneumonias. Schweiz Med Wochenschr. 1985;115:75–86. [PubMed] [Google Scholar]
- 202.Nickerson C.L., Jakab G.J. Pulmonary antibacterial defenses during mild and severe influenza virus infection. Infect Immun. 1990;58:2809–2814. doi: 10.1128/iai.58.9.2809-2814.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Warshauer D., Goldstein E., Akers T., Lippert W., Kim M. Effect of influenza viral infection on the ingestion and killing of bacteria by alveolar macrophages. Am Rev Respir Dis. 1977;115:269–277. doi: 10.1164/arrd.1977.115.2.269. [DOI] [PubMed] [Google Scholar]
- 204.Sun K., Metzger D.W. Inhibition of pulmonary antibacterial defense by interferon-gamma during recovery from influenza infection. Nat Med. 2008;14:558–564. doi: 10.1038/nm1765. [DOI] [PubMed] [Google Scholar]
- 205.Hang do TT., Choi E.J., Song J.Y., Kim S.E., Kwak J., Shin Y.K. Differential effect of prior influenza infection on alveolar macrophage phagocytosis of Staphylococcus aureus and Escherichia coli: involvement of interferon-gamma production. Microbiol Immunol. 2011;55:751–759. doi: 10.1111/j.1348-0421.2011.00383.x. [DOI] [PubMed] [Google Scholar]
- 206.Hussell T., Cavanagh M.M. The innate immune rheostat: influence on lung inflammatory disease and secondary bacterial pneumonia. Biochem Soc Trans. 2009;37:811–813. doi: 10.1042/BST0370811. [DOI] [PubMed] [Google Scholar]
- 207.Didierlaurent A., Goulding J., Patel S., Snelgrove R., Low L., Bebien M. Sustained desensitization to bacterial Toll-like receptor ligands after resolution of respiratory influenza infection. J Exp Med. 2008;205:323–329. doi: 10.1084/jem.20070891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Snelgrove R.J., Goulding J., Didierlaurent A.M., Lyonga D., Vekaria S., Edwards L. A critical function for CD200 in lung immune homeostasis and the severity of influenza infection. Nat Immunol. 2008;9:1074–1083. doi: 10.1038/ni.1637. [DOI] [PubMed] [Google Scholar]
- 209.Westphalen K., Gusarova G.A., Islam M.N., Subramanian M., Cohen T.S., Prince A.S. Sessile alveolar macrophages communicate with alveolar epithelium to modulate immunity. Nature. 2014;506:503–506. doi: 10.1038/nature12902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Park S.Y., Kim I.S. Engulfment signals and the phagocytic machinery for apoptotic cell clearance. Exp Mol Med. 2017;49:e331. doi: 10.1038/emm.2017.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Juncadella I.J., Kadl A., Sharma A.K., Shim Y.M., Hochreiter-Hufford A., Borish L. Apoptotic cell clearance by bronchial epithelial cells critically influences airway inflammation. Nature. 2013;493:547–551. doi: 10.1038/nature11714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Han C.Z., Juncadella I.J., Kinchen J.M., Buckley M.W., Klibanov A.L., Dryden K. Macrophages redirect phagocytosis by non-professional phagocytes and influence inflammation. Nature. 2016;539:570–574. doi: 10.1038/nature20141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Barkauskas C.E., Cronce M.J., Rackley C.R., Bowie E.J., Keene D.R., Stripp B.R. Type 2 alveolar cells are stem cells in adult lung. J Clin Invest. 2013;123:3025–3036. doi: 10.1172/JCI68782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Zemans R.L., Briones N., Campbell M., McClendon J., Young S.K., Suzuki T. Neutrophil transmigration triggers repair of the lung epithelium via beta-catenin signaling. Proc Natl Acad Sci U S A. 2011;108:15990–15995. doi: 10.1073/pnas.1110144108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Chapman H.A., Li X., Alexander J.P., Brumwell A., Lorizio W., Tan K. Integrin alpha6beta4 identifies an adult distal lung epithelial population with regenerative potential in mice. J Clin Invest. 2011;121:2855–2862. doi: 10.1172/JCI57673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Rock J.R., Barkauskas C.E., Cronce M.J., Xue Y., Harris J.R., Liang J. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc Natl Acad Sci U S A. 2011;108:E1475–E1483. doi: 10.1073/pnas.1117988108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Liu Y., Sadikot R.T., Adami G.R., Kalinichenko V.V., Pendyala S., Natarajan V. FoxM1 mediates the progenitor function of type II epithelial cells in repairing alveolar injury induced by Pseudomonas aeruginosa. J Exp Med. 2011;208:1473–1484. doi: 10.1084/jem.20102041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Zhou Y., Liu Q., Dai X., Yan Y., Yang Y., Li H. Transdifferentiation of type II alveolar epithelial cells induces reactivation of dormant tumor cells by enhancing TGF-beta1/SNAI2 signaling. Oncol Rep. 2018;39:1874–1882. doi: 10.3892/or.2018.6247. [DOI] [PubMed] [Google Scholar]
- 219.Atabai K., Ishigaki M., Geiser T., Ueki I., Matthay M.A., Ware L.B. Keratinocyte growth factor can enhance alveolar epithelial repair by nonmitogenic mechanisms. Am J Physiol Lung Cell Mol Physiol. 2002;283:L163–L169. doi: 10.1152/ajplung.00396.2001. [DOI] [PubMed] [Google Scholar]
- 220.Zemans R.L., McClendon J., Aschner Y., Briones N., Young S.K., Lau L.F. Role of beta-catenin-regulated CCN matricellular proteins in epithelial repair after inflammatory lung injury. Am J Physiol Lung Cell Mol Physiol. 2013;304:L415–L427. doi: 10.1152/ajplung.00180.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Cakarova L., Marsh L.M., Wilhelm J., Mayer K., Grimminger F., Seeger W. Macrophage tumor necrosis factor-alpha induces epithelial expression of granulocyte-macrophage colony-stimulating factor: impact on alveolar epithelial repair. Am J Respir Crit Care Med. 2009;180:521–532. doi: 10.1164/rccm.200812-1837OC. [DOI] [PubMed] [Google Scholar]
- 222.Bhaskaran M., Kolliputi N., Wang Y., Gou D., Chintagari N.R., Liu L. Trans-differentiation of alveolar epithelial type II cells to type I cells involves autocrine signaling by transforming growth factor beta 1 through the Smad pathway. J Biol Chem. 2007;282:3968–3976. doi: 10.1074/jbc.M609060200. [DOI] [PubMed] [Google Scholar]
- 223.Zacharias W.J., Frank D.B., Zepp J.A., Morley M.P., Alkhaleel F.A., Kong J. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature. 2018;555:251–255. doi: 10.1038/nature25786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Kajstura J., Rota M., Hall S.R., Hosoda T., D'Amario D., Sanada F. Evidence for human lung stem cells. N Engl J Med. 2011;364:1795–1806. doi: 10.1056/NEJMoa1101324. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 225.Hoeve M.A., Nash A.A., Jackson D., Randall R.E., Dransfield I. Influenza virus A infection of human monocyte and macrophage subpopulations reveals increased susceptibility associated with cell differentiation. PLoS One. 2012;7:e29443. doi: 10.1371/journal.pone.0029443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Zemans R.L., Henson P.M., Henson J.E., Janssen W.J. Conceptual approaches to lung injury and repair. Ann Am Thorac Soc. 2015;12:S9–S15. doi: 10.1513/AnnalsATS.201408-402MG. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Duffield J.S., Forbes S.J., Constandinou C.M., Clay S., Partolina M., Vuthoori S. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest. 2005;115:56–65. doi: 10.1172/JCI22675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Gibbons M.A., MacKinnon A.C., Ramachandran P., Dhaliwal K., Duffin R., Phythian-Adams A.T. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med. 2011;184:569–581. doi: 10.1164/rccm.201010-1719OC. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.


