Abstract
The human lung is constantly exposed to the environment and potential pathogens. As the interface between host and environment, the respiratory epithelium has evolved sophisticated sensing mechanisms as part of its defense against pathogens. In this review, we examine how the respiratory epithelium senses and responds to influenza A virus, the biggest cause of respiratory viral deaths worldwide.
Keywords: respiratory epithelium, influenza A virus, viral sensing
Influenza viruses belong to the Orthomyxoviridae family. They are enveloped viruses that contain eight negative sense single-stranded RNA (ssRNA) segments encoding 11 proteins, including the hemagglutinin, neuraminidase, and nucleoprotein (1). Influenza A virus (IAV), which causes pandemic and seasonal influenza infections, is one of the four genera of the Orthomyxoviridae family (the other three are influenza B and C viruses and Thogotovirus) and a major public health burden worldwide (1–3). IAV is responsible for an estimated 645,000 deaths worldwide annually (4, 5). Mutations in the surface proteins (hemagglutinin and neuraminidase) result in antigenic drifts and seasonal epidemics, whereas reassortment of genetic material of two or more strains in a common host forms novel IAV strains and pandemics. The World Health Organization provides annual recommendations for the composition of influenza virus vaccines before upcoming influenza seasons, but mismatches between the predicted and actual circulating seasonal variants occur as frequently as every 3–4 years (6, 7). Therefore, IAV infection remains a threat to humans, and there is a continued need to understand how the lung protects itself from IAV infection to harness this understanding for new strategies against IAV infections.
IAV preferentially binds α2,6-linked sialic acid (α2,6-SA) receptors expressed by airway epithelium from the nose to respiratory bronchioles (8–10). The first epithelial defense takes the form of cell–cell junctions, including tight junctions (located in the apical portion of the lateral epithelial membrane), adherens junctions, desmosomes and gap junctions (11, 12), that are linked to the cellular cytoskeleton via numerous adaptor proteins. These proteins form an impermeable mechanical barrier that is highly resistant to damage during IAV infections (13). The pseudostratified columnar epithelium in the trachea, bronchi, and bronchioles is interspersed by a further diverse group of cells: serous, club, neuroendocrine, and goblet cells that secrete mucin (not club cells) and a variety of enzymes, protease inhibitors, oxidants, and antimicrobial peptides (14). Among these, mucin, β-defensins, and LL37 have been shown to have specific anti-IAV roles (15–22).
The respiratory epithelium is therefore the first point of defense in the lung’s response to IAV. It works closely with the innate immune cells, including monocytes, neutrophils, and natural killer (NK) cells, to deliver an optimal first-line protection system. It also provides a bridge between the innate and adaptive immune responses by producing cytokines and chemokines that attract, mature, and differentiate T and B cells. It communicates with the principal antigen-presenting cells (dendritic cells [DCs]) via secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF), thymic stromal lymphopoietin (TSLP), IL-25, and IL-33 and provides a focal point for lymphoid aggregates such as inducible bronchus-associated lymphoid tissue (iBALT) that surround the airways after infection (23). In addition to the infiltrating innate immune cells, there are also lung-resident immune cells—intraepithelial conventional dendritic cells (cDCs), intraepithelial CD8 T cells, innate lymphoid cells (ILCs), and unconventional T-cell populations such as the invariant natural killer T (iNKT) cells, mucosal-associated invariant T cells (MAIT), and γδ T cells that may play important roles in the “hyper-early” immune activities during influenza. Together with the infiltrating innate immune cells, the respiratory epithelium and resident immune cells form the first innate immune response to IAV infection, and they reduce the viral burden (often clearing the virus altogether) and determine the shape of subsequent T- and B-cell responses, the health of the tissue, and the ensuing immunological memory. In this paper, we examine the first point of this process: how the respiratory epithelium senses and reacts to IAV infection.
Sensing the Influenza Virus
Mucins in the mucus layer are widely regarded as the first defense against IAV. Not only do they form a physical barrier against the virus, but one isoform, Muc5ac, contains α2,3-linked sialic acid motifs that can act as decoys for the sialic acid receptors on the respiratory epithelium. Thus, the IAV is kept in the mucinous phase and prevented from accessing the airway’s surface, a necessary process to internalize the virus (18). More recently, a study suggested that Muc5ac can also relay the presence of viruses (24). Iversen and colleagues showed that airway epithelial cells can sense herpes simplex virus via the mucin layer, triggering the earliest release of CXCL10 from epithelial cells and recruitment of neutrophils independently and even before the early type I IFN responses (24). However, direct studies with IAV have not been performed.
Once the IAV gets past these early barriers, it attaches its surface glycoprotein, hemagglutinin (HA), to sialic acid expressed on sialic acid receptor found on the epithelial cell surface and is endocytosed. In the endosome, the viral HA is acidified and undergoes a conformational rearrangement, enabling viral and endosomal membranes to fuse (reviewed by Reference 25). The H+ ions in the acidic environment of the endosome then permeates the virus via its M2 ion channel, disrupting the viral envelope (25). The viral RNA is then released into the cytoplasm of the host cell and imported into the nucleus to facilitate transcription of the viral genome and translation of viral proteins (25). The host cell senses IAV as soon as it is internalized, using pathogen recognition receptors (PRRs). In respiratory epithelium, these are primarily the Toll-like receptors (TLRs), retinoic acid-inducible gene 1–like receptors (RLRs), and NOD (nucleotide-binding oligomerization domain-containing protein)-like receptors (NLRs).
TLRs
TLRs are the first PRRs to confront IAV in the respiratory epithelium. Three TLRs are implicated in IAV recognition by human respiratory epithelial cells: TLR3, TLR7, and TLR8 (2, 26–32). Upon recognition of the relevant protein-associated molecular patterns (PAMPs), these TLRs recruit Toll/IL-1 receptor domain–containing adaptor proteins such as MyD88 (myeloid differentiation primary response 88) and TRIF (Toll/IL-1 receptor domain-containing adapter-inducing IFN-β), which then initiate signal transduction pathways that culminate in the expression of types I and III IFN via the transcription factors NF-κB and IRFs (IFN-regulatory factors) (Figure 1) (2). IAV-infected MyD88−/− and MyD88/TRIF double-deficient mice display markedly reduced cytokine production in the lung compared with wild-type mice, indicating the importance of this pathway during IAV infection (26, 31, 32). TLR3 is constitutively expressed by both bronchial and alveolar epithelial cells and recognizes viral double-stranded RNA (dsRNA) (intermediate RNA species produced during replication of IAV), siRNAs, and self-RNAs derived from damaged cells generated during infection (33–36). Unlike retinoic acid-inducible gene 1 (RIG-1), melanoma differentiation-associated protein 5 (MDA5), and NLRP3 (NACHT, LRR and PYD domains-containing protein 3), TLR3 is expressed on both the cell surface and the endosome, allowing the epithelial cells to sense IAV PAMPs both externally and internally (in the endosome) (27, 36–39). A distinctive feature of TLR3 in IAV infection is its link to secretion of proinflammatory cytokines, including IL-6, IL-8, and RANTES (regulated upon activation, normal T cell expressed and secreted) (36), and to immunopathology (36, 39, 40). Tlr3−/− mice showed better survival after lethal IAV infection, despite higher viral loads in the lungs (40). They also have lower chemokine and CD8+ T-cell concentrations after IAV infection, but antibody and CD4+ and CD8+ T-cell responses to sublethal doses of IAV infection are unaffected (41). Tlr3−/− mice also showed greater lung pathology after H5N1 infection, but not after infection with pandemic H1N1 infection, possibly because of the greater innate immune response generated by the H5N1 virus (26, 40). In all these studies, no differentiation was made between TLR3 expressed by respiratory epithelial cells and that by myeloid cells; thus, the actual significance of epithelial TLR3 in vivo is still unknown.
Figure 1.
Overview of pattern recognition receptors and inflammasome sensing mechanism during influenza A virus (IAV) infection. IAV infects cells by binding of the virion surface glycoprotein hemagglutinin to sialic acid that is expressed by a cell surface receptor and then is endocytosed. The viral hemagglutinin fuses with the endosomal membrane; the virion particle unravels; and the viral RNA is released into the cytoplasm. TLR3 and TLR7 sense viral double-stranded RNA and single-stranded RNA (ssRNA), respectively, within the endosome, whereas retinoic acid–inducible gene 1–like receptors (RIG-I and MDA5) recognize cytosolic ssRNA or viral RNA containing 5′-triphosphate. Via their C-terminal caspase-recruitment domains, RIG-I and MDA5 attach to the signaling adaptor protein (MAVS, found in the mitochondria), causing activation of IRFs and NF-κB and transcription of proforms of IL-1β, IL-18, caspase 1, and the types I and III IFNs. As a result of IRF and NF-κB activation (also acting as signal 1 of the inflammasome pathway), other components essential to formation of the inflammasome complex, such as ASC and NLRP3, are upregulated. NLRP3 forms one of the inflammasome second signal receptors, and when activated (e.g., when sensing ssRNA from IAV and IAV-associated DAMPs), it nucleates ASC, which bridges the inflammasome sensor to oligomerize pro–caspase 1, causing caspase 1 activation. Active caspase 1 cleaves the pro–IL-1β and pro–IL-18 proteins, generating the biologically active cytokines IL-1β and IL-18. ASC = apoptotic speck protein containing a C-terminal caspase recruitment domain; DAMPs = damage-associated molecular patterns; IFNAR = IFN-αβ receptor; IRF3/7 = IFN-regulatory factor 3/7; ISGs = IFN-stimulated genes; MAVS = mitochondrial antiviral signaling protein; MDA5 = melanoma differentiation-associated protein 5; MyD88 = myeloid differentiation primary response 88; NLR = nucleotide-binding oligomerization domain-like receptor; NLRP3 = NACHT, LRR and PYD domains-containing protein 3; RIG-I = retinoic acid-inducible gene 1; STAT1/2 = signal transducer and activator of transcription 1/2; TLR = Toll-like receptor; TRIF = Toll/IL-1 receptor domain-containing adapter-inducing IFN-β. Adapted by permission from Reference 22.
TLR7 and TLR8 are expressed in endosomes and bind ssRNA ligands, leading to recruitment of the adaptor molecule MyD88 and subsequent activation of NF-κB and IRF7. TLR7 is highly expressed in certain immune cell types, such as plasmacytoid dendritic cells, which secrete type I IFNs upon infection with IAV (42, 43), and is also found in human bronchial epithelial cells (44). Sykes and colleagues reported that stimulation of healthy primary human bronchial epithelial cells in ex vivo undifferentiated culture conditions with R848, a TLR7/8 ligand, did not induce IFN-β secretion (45); rather, it induced secretion of IL-6, IL-8, and epithelium-specific IFN-λ proteins. In a separate study by Ioannidis and colleagues, TLR7 activation by a thiazoloquinolone derivative (CL075) failed to induce secretion of IFN-α, IFN-β, IFN-λ, and IL-6 from polarized, well-differentiated human alveolar epithelial cells in vitro and only slightly increased IL-8 secretion (46). These somewhat contradictory findings suggest that epithelial cell differentiation could influence antiviral and proinflammatory responses to TLR7 activation. In an in vivo study, TLR7−/− mice infected with IAV were found to have poor formation of germinal center B cells in lungs and spleen and subsequent poorer B-cell response upon rechallenge, but the viral titers in TLR7−/− mice during the acute period were not different from those of control animals (47).
RLRs
The RLR family of innate receptors are the main cytoplasmic PRR and comprises three cytosolic virus recognition receptors: RIG-I, MDA5, and Laboratory of Genetics and Physiology 2 (LGP-2) (48). All three are RNA helicases and act as viral dsRNA sensors (49). RIG-I and MDA5 also possess C-terminal caspase recruitment domains (CARDs), allowing them to engage with the CARD-containing adapter mitochondrial antiviral signaling protein (MAVS), which then allows activation of IRF and NF-κB (Figure 1). LGP-2, which is devoid of CARDs, functions as a negative regulator of RIG-I–induced IFN signaling (50) by sequestering dsRNA (49–51). IAV RNA species found in the cytoplasm are rapidly detected by RIG-I and MDA5 (51, 52), aided by an increase in RIG-I and MDA5 levels in IAV-infected epithelial (including alveolar epithelial) cells (36, 53, 54). The primary consequence of RLR signaling is activation of IFN genes. This occurs via interaction with MAVS, which in turn engages the IRF3/7 transcription factor signaling pathway (55) and NF-κB, leading to type I IFN induction (36, 49).
RIG-I, at least in murine bone marrow–derived dendritic cells (BM-DCs) and human HEK293 cells, has also been reported to detect IAV ssRNA that carries phosphorylated 5′-ends (56). Unlike mammalian mRNA, the IAV viral genome comprises uncapped RNA segments with phosphate groups at their 5′-termini (57, 58). The ability of RIG-I to recognize these 5′-phosphate groups on ssRNA and the dsRNA domains makes it a superior sensor. There are also several other distinctive features of RIG-I induction of an antiviral state. First, engagement with the virus occurs almost immediately. It was observed that RIG-I, IAV polymerases, and MAVS associate with each other and with the mitochondria as early as 3 hours after infection (59). In response, IAVs have established built-in methods to attenuate this pathway. The most prominent is the virus’s nonstructural protein 1 (NS1), which suppresses the type I IFN response by interacting with RIG-I (60, 61) and TRIM25 (62). IAV’s polymerases (PB1 and PA) also associate very early with MAV in the RIG-I signaling pathway and the mitochondria to inhibit type I IFN expression (59).
The second noteworthy feature of RIG-I signaling is that it induces an antiviral state in several ways. It not only causes IFN induction but also directly inhibits virus replication by binding to the 5′-triphosphate dsRNA end and promoting the disassembly of the viral polymerase complex (63). It also promotes the assembly of an inflammasome complex. RIG-I stimulates NF-κB–mediated expression of pro–IL-1β expression via MAVS and CARD9 (2). In parallel, RIG-I can also directly activate the inflammasome complex by binding to the inflamasome’s adaptor, ASC (apoptotic speck protein containing a C-terminal caspase-recruitment domain) (64, 65). IFN responses are reduced in RIG-I or MDA5 deficient alveolar epithelial cell lines (A549) (54).
In vivo studies on RLRs have been limited because RIG-I–null mice are not viable on common laboratory background stains (66). However, recent studies have shown that RIG-I–knockout mice on an ICR background (which are viable) demonstrated delayed IAV clearing and display higher viral loads (67). Within RIG-I–like receptor family, RIG-I may be the primary and MDA5 a secondary contributor to viral sensing (68). Between cell types, there may also be a differential contribution of these sensing pathways to the generation of an IFN response. For example, Crotta and colleagues found that in alveolar epithelial cell lines, knocking out TLR7 or MyD88 had minimal effect on type I or III IFN production, but ablation of MAVS greatly reduced IFN levels (69). However, there are currently no in vivo studies that localize the effect of RIG-I knockout to respiratory epithelial cells.
NLRs
NLRs are cytoplasmic PRRs that contain central nucleotide and oligomerization (NACHT) domains; these domains determine the families of NLRs. NLRs contribute to the inflammasomes, which are multiprotein complexes that assemble in the cytosol after exposure to PAMPs or damage-associated molecular patterns (DAMPs), with resultant activation of caspase 1 and subsequent release of the proinflammatory cytokines IL-1β and IL-18 (70) (Figure 1). These inflammasome complexes usually consist of a cytosolic pattern PRR, pro–caspase 1, and an adaptor protein (ASC) (71). A number of distinct inflammasome complexes have been identified, each with unique PRR and activation triggers (72). The most widely studied is the NLRP3 complex, which contains the NLR, NLRP3, ASC, pro–caspase 1, and the NEK7, a serine-threonine kinase (71, 72). The complete activation of the inflammasome pathway requires two separate signals (73). Signal 1 involves the priming of cells by detection of PAMPs by cytoplasmic PRRs (e.g., TLRs 3, 7, and 8; and RLRs described earlier). This results in increased expression of caspase 1, pro–IL-1β, and pro–IL-18 (priming step, signal 1) (71–73). Subsequently, indirect activation of NLRP3 occurs through a number of diverse signals (whole pathogens, PAMPs/DAMPs, potassium efflux, lysosomal damaging factors [e.g., silica, uric acid, and alum] and endogenous factors [amyloid-β and cholesterol crystals], as well as by mitochondrial damage), leading to complex assembly and activation of caspase 1 (signal 2) (71–73). Active caspase 1 then cleaves the precursor cytokines pro–IL-1β and pro–IL-18, leading to generation of the biologically active cytokines IL-1β and IL-18, respectively, which are then secreted from the cell (71–73). Secretion of the mature IL-1β and IL-18 inflammatory cytokines then leads to recruitment of neutrophils, monocytes, and macrophages to the site of infection (74, 75). Other inflammasomes can be activated by more directly; double-stranded DNA activates the absent in melanoma 2 (AIM2) complex; anthrax toxin activates NLRP1, and bacterial flagellin activates NLR family CARD-containing protein 4 (NLRC4) (76).
NLRP3 is the most studied NLR family member in IAV infection. Unlike the TLR and RLR, NLRP3 is activated by both PAMPs (ssRNA) and DAMPS generated during an IAV infection (77–81). Intracellular molecules (i.e., ATP and high-mobility group box 1 protein [HMGB1]) released during infection-induced apoptosis, necrosis, or pyroptosis (82), can serve as DAMPs during IAV infection. They accumulate in the extracellular space at a high concentration and act as signal 1 for inflammasome activation (83–85). However, recognition of DAMPs does not always result in an enhanced immune response and viral clearance. For example, recognition of HMGB1 through the DAMP receptor, receptor for advanced glycation end products (RAGE) appears detrimental during IAV infection, as both ligand and receptor become highly expressed in endothelium and bronchial epithelium (86). NLRP3 is required for a successful defence against IAV-human airway epithelial cells express NLRP3 and secrete IL-1β when challenged with IAV (87, 88) and mice without NLRP3 (or inflammasome pathway components) showed reduced survival and impaired innate immune responses (87). However, the majority of the NLRP3 response is linked to myeloid cells rather than from respiratory epithelial cells in this murine model (87); hence, there is still little to indicate the importance of epithelium-expressed NLRP3. Interestingly, a recent study in bacterial infection suggested that NLRP3 expressed by epithelial cells has a role during infection but via the maintenance of alveolar barrier integrity, independent of involvement of the inflammasome, IL-1β, and IL-18 (89). Nonetheless, the importance of the inflammasome pathway during IAV infection is suggested by presence of viral mechanisms that interfere with inflammasome activation. For example, the viral NS1 protein is can block caspase 1 activation, IL-1β maturation, and apoptosis (90). The caspase 1–inhibitory effect of NS1 demonstrates strain-specificity H5N1 NS1 does not appear to activate caspases, but instead induces apoptosis of the epithelial cells (91).
Consequence of Viral Sensing
IFN Response
The central consequence of PRR activation during IAV infection is the induction of the IFN response. There are three classes of IFN: type I (IFN-α and IFN-β), type II (IFN-γ), and type III (IFN-λ), each with distinct expression patterns and roles in the antiviral response. In epithelial cells, the key consequence of intracellular recognition of IAV infection by TLRs and RLRs is the activation of type I and type III IFN pathway. Type I IFN responses are ubiquitous; almost all nucleated cell types are able to secrete type I IFNs and express the IFN-αβ receptor (92). Type II IFN (IFN-γ) is secreted by specific immune cell populations including CD8+ T cells, CD4+ T-helper type 1 cells (93), invariant natural killer T cells (94), type 1 ILCs (95), and NK cells (96). Type III IFNs are primarily secreted by epithelial cells, and type III IFN responses are restricted to barrier sites linked to high exposure to pathogens (e.g., airway or intestinal epithelium) (97, 98). The expression patterns and the levels of types I and III IFNs are dependent both on the site of infection and on the pathogen (99, 100).
Although distinct receptor complexes bind types I and III IFNs, many of the intracellular signaling molecules and pathways are shared between type I and type III IFNs. Activation of these pathways by either IFN class results in broadly similar biological activities. Types I and III IFNs rapidly induce an antiviral state by inducing the expression of hundreds of genes, grouped as IFN-stimulated genes (ISGs). These genes carry IFN-responsive elements in their promoters and suppress transcription of other genes, including viral genes. These ISGs both limit viral replication and spread, and they induce immune responses in neighboring cells. In contrast, type II IFNs have limited direct antiviral effects but promote innate and adaptive immune responses (92).
Recent studies have highlighted type III IFNs (rather than type I IFNs) as the dominant IFN response in the airway epithelium (100) and are potentially the superior of the two IFNs in host defense. Robust type I IFN responses are associated with effective viral clearance, but at the cost of bystander immunopathology (101). Generation of type III IFN responses at barrier surfaces, on the other hand, induces an antiviral state with limited damage to the host (97). The bias toward type III IFN responses is restricted to tissues continually exposed to pathogens, such as the lungs and gut. There is also evidence to suggest that type III IFN is generated as a first line of defense before potentially more damaging type I responses (100) and is critical for control of influenza virus dissemination in the upper airways (102). IFN-λ receptors deficient mice release significantly more infectious virus particles and transmit the virus much more efficiently to naive contacts than did wild-type mice or mice lacking functional type I IFN receptors (102).
Downstream from IFNs, ISG-derived proteins perform two key functions. First, they directly limit viral replication by shutting down protein synthesis (2) and triggering apoptosis (103). Second, ISGs activate key components of the innate and adaptive immune systems, including antigen presentation and production of cytokines involved in activation of T cells, B cells, and NK cells (2). Thus far, several hundred ISGs have been identified, and three key genes— myxovirus resistance protein (Mx), IFN-induced transmembrane protein 3 (IFITM3), and IRF7—have been shown to have important roles in IAV infections. Mx proteins are a family of GTPases with broad antiviral activities against RNA viruses (104–107). They interact with the IAV nucleocapsid and its entry into the nucleus, thereby inhibiting viral genome transcription and replication (108–110). IFITM3, a small, conserved transmembrane protein, was among the first ISGs identified and restricts the proliferation of a broad range of viruses (111). By modulating endosomal cholesterol, IFITM3 blocks pH-dependent fusion in the late endosome and so impairs viral entry to the cytoplasm (112–114). Mice lacking Ifitm3 had higher viral titers, morbidity, and mortality than wild-type mice when infected with IAV (112, 113). A functional IFITM3 SNP (rs12252) has been linked to severe influenza infection in UK and Chinese patients (115). Although, the antiviral role of IFITM3 is well established, the contribution of epithelial IFITM3 to viral control has been little studied. However, IFITM3 is constitutively expressed by the respiratory epithelium and type II alveolar cells, which suggests that this is a potential mechanism by which respiratory epithelium might reduce viral dissemination (116). The transcription factor IRF7 is a master regulator of type I IFN induction and ISG expression (117). Although specific immune cell populations, including plasmacytoid dendritic cells and macrophages, constitutively express IRF7 (118–120), levels in respiratory epithelial cells are low but induced in response to IFN signaling (121). The critical role of the positive feedback loop generated by IFN and IRF7 is demonstrated by the near ablation of IFN production and increased mortality and morbidity observed in influenza-infected IRF7-knockout mice (122, 123). Furthermore, life-threatening influenza infection in an otherwise healthy child was linked to IRF7-null mutation and failure to generate an IFN response in respiratory epithelial cells isolated from the child (124).
Overall, it seems highly likely that PRRs in epithelial cells, including alveolar epithelial cells (125), are involved in host defense. However, there is not enough evidence to place their importance in the context of the overall host defense against IAV, because PRRs are also expressed on and in myeloid cells. Indeed, loss of TLR3 (40) and the RIG-I signaling adaptor MAVS and IPS-1 does not affect viral clearance and the adaptive immunity to IAV infection (126).
Cytokine and Chemokine Production
In addition to the initial IFN-mediated antiviral response, epithelial cells secrete various cytokines and chemokines, such as IL-6, TNF-α, IL-8/CXCL8, CXCL10, CCL2, and CCL5 (127–131). Activation of these pathways is rapid, and increased concentrations of cytokines and chemokines can be detected within a few hours of IAV infection (127, 132–136). These soluble mediators further activate and attract other immune cell populations into the lung (127, 132–135). However, both lung-resident and recruited immune cell populations are also able to secrete many of these cytokines. In this review, we highlight work that has specifically established the role of the epithelium-derived cytokines and chemokines during IAV infection.
CCL2 is secreted by both airway epithelium and activated monocytes and acts as a monocyte chemoattractant (137, 138). The CCL2–monocyte recruitment pathway has both beneficial and detrimental roles during IAV infection. The induction of a robust monocyte response is associated with immunopathology (139, 140). Several studies have demonstrated that mice lacking CCL2 receptor (CCR2) expression had improved outcomes in severe influenza infection (141–143), although these mice also showed delayed viral clearance (144). One study was able to show that CCL2 produced by epithelial cells rather than myeloid cells is linked to immunopathology. Maelfait and colleagues showed that deletion of the immune regulatory factor A20 (or TNF-α–induced protein 3 [TNFAIP3]) specifically in respiratory epithelial cells resulted in a better outcome from IAV infection, and this correlated with a strongly suppressed expression of the chemokine CCL2 during later stages of infection (145).
IL-1β, an early proinflammatory cytokine, is released in response to viral triggering of the NLRP3 inflammasome (146, 147). IL-1β both stimulates release of other cytokines and chemokines by the epithelium and increases adhesion molecule expression, enhancing immune cell recruitment to the lungs (148, 149). In mice, IL-1 signaling is necessary for survival during IAV infection, but conversely, IL-1β also drives lung immunopathology (150, 151). Ablating IL-1 signaling in IAV-infected mice impaired neutrophil recruitment and CD4 T-cell activation and recruitment with subsequent reduction in pulmonary inflammation (150). Respiratory epithelial cells have a key role in both the generation and regulation of the IL-1β response. At homeostasis, they secrete IL-1β inhibitors (IL-1RA and soluble IL-1RII) that prevent spontaneous IL-1β signaling. Therefore, by controlling IL-1β signaling at steady state, the respiratory epithelium promotes an antiinflammatory environment, preventing IL-1β–driven pathology; however, this respiratory epithelial cell tolerogenic state can rapidly be broken by proinflammatory cytokines, including TNF-α (152).
Transforming growth factor (TGF)-β is a pleiotropic cytokine with an integral role in regulation of immune responses. Many immune and nonimmune cells can produced TGF-β, but secretion is specifically increased in the airway epithelium early after IAV infection (135, 153, 154). Overexpression of TGF-β in the lungs reduced inflammatory responses but also viral clearance (155). The regulatory properties of TGF-β have been exploited by IAV to control the early immune responses in the lung. Neuraminidase viral protein can cleave and activate latent TGF-β (153, 156), regulating type I IFN production and so generating a degree of viral tolerance in the host. When TGF-β is ablated specifically in the airway epithelial cells, weight loss, airway inflammation, immunopathology, and viral titers are all reduced (135). This protective phenotype was due to an enhanced early IFN-β release from the respiratory epithelium and was amplified by alveolar macrophages rather than an altered lymphocytic immune response (135). Therefore, IAV infection generates a viral permissive state via neuraminidase cleavage and activation of epithelium-derived TGF-β, which then suppresses early protective IFN responses. The different roles of epithelial and myeloid cells in the generation of the TGF-β responses during IAV infection are further highlighted by Meliopoulos and colleagues. Mice that lacked the β6-integrin on the respiratory epithelium and so had impaired activation of TGF-β also showed improved survival after IAV infection due to constitutively activated airway macrophages with increased IFN responsiveness (157).
A key issue with the role of sensors on epithelial cells is the use of whole-organism responses as an endpoint, so that the segregation of epithelial versus hematopoietic cell contributions is not possible (exceptions being specific studies highlighted in the paragraphs above). In addition, experiments were often based on simple in vitro cell culture systems that utilize undifferentiated, nonpolarized respiratory epithelial cells. Moreover, there are significant interspecies differences between rodents and humans for certain aspects of IAV-triggered pathogenesis (e.g., induction of Mx proteins). Different responses of undifferentiated versus well-differentiated mucociliary epithelial in vitro cultures have been reported (e.g., TLR7-induced IFN production). Complementary approaches that combine better in vitro models (158), such as three-dimensionally reconstituted pseudostratified human epithelial innate responses or lung organ-on-chip approaches, will significantly enhance the studies. Animal models in which counterparts of human genes exist and can be selectively manipulated in the respiratory epithelium are needed in this area. However, defining the exact contribution of epithelial cells to the generation of the influenza immune responses in vivo is challenging. Several genetically modified models have been developed that allow epithelial cell targeted gene overexpression or deletion, although important caveats must be acknowledged (159, 160). Epithelial cell promoters in common use are Scgb1a1 (club cell secretory protein [CCSP]) and SFTPC (surfactant protein C or SPC) targeting the proximal and distal airway epithelial cells, respectively, in mice (160). These promoters can be used to drive gene overexpression or deletion of genes flanked by LoxP sites (floxed) via constitutive Cre recombinase (CRE) expression or an inducible expression system (i.e., tetracycline [doxycycline]-responsive element). However, constitutive CRE expression models can result in CRE toxicity (161) and may have limited utility when genes have key roles in development. Using an inducible tetracycline (doxycycline)-responsive element allows excision of a gene at a specific time point (i.e., shortly before .infection). However, deletion occurs only at the level of genomic DNA, and pools of both protein and RNA from the targeted gene can remain in the cell for some time (162, 163). It is important to note that neither system offers a complete deletion of the gene of interest in the target population (i.e., some cells will be unaffected), and CRE expression and gene deletion can occur in off-target cell types (164). Despite limitations with genetically modified models, specific targeting of genes in the epithelium provides important insight into the contribution of the epithelium to host defense during influenza infection.
Conclusions
This review is focused on how epithelial cells sense and respond to IAV during the first critical hours of infection. If successful, this first response triggers a process that can eradicate the virus and prevent infection. If not, IAV takes hold and replicates, and the host engages bigger waves of innate immune cells and subsequently the adaptive immune response. The comparative lack of studies localizing the specific role of the respiratory epithelium during IAV infection means that this large defense mechanism has been relatively untapped in its ability to prevent infection and to regulate immunopathology. Understanding the specific part that epithelial PRRs (e.g., the NLRP3 inflammasomes, which sense more triggers than TLR and RLR) play in immunopathology could be particularly useful, because modulating this pathway directly could potentially reduce inflammation in severe disease.
Supplementary Material
Footnotes
Supported by a Career Enhancement Award from Colorado SPORE in Lung Cancer (P50 CA058187), a Lush Young Researcher Award, and the University of Colorado Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine (K.H.B.); by the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC) (L.D.); and by the Medical Research Council UK (L.-P.H.).
Author Contributions: K.H.B. wrote the initial draft; K.H.B., L.D., and L.-P.H. further added, revised, and updated the manuscript.
Originally Published in Press as DOI: 10.1165/rcmb.2018-0247TR on October 29, 2018
Author disclosures are available with the text of this article at www.atsjournals.org.
References
- 1.Neumann G, Noda T, Kawaoka Y. Emergence and pandemic potential of swine-origin H1N1 influenza virus. Nature. 2009;459:931–939. doi: 10.1038/nature08157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Iwasaki A, Pillai PS. Innate immunity to influenza virus infection. Nat Rev Immunol. 2014;14:315–328. doi: 10.1038/nri3665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Leahy MB, Dessens JT, Weber F, Kochs G, Nuttall PA. The fourth genus in the Orthomyxoviridae: sequence analyses of two Thogoto virus polymerase proteins and comparison with influenza viruses. Virus Res. 1997;50:215–224. doi: 10.1016/s0168-1702(97)00072-5. [DOI] [PubMed] [Google Scholar]
- 4.Iuliano AD, Roguski KM, Chang HH, Muscatello DJ, Palekar R, Tempia S, et al. Global Seasonal Influenza-associated Mortality Collaborator Network. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018;391:1285–1300. doi: 10.1016/S0140-6736(17)33293-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Molinari NA, Ortega-Sanchez IR, Messonnier ML, Thompson WW, Wortley PM, Weintraub E, et al. The annual impact of seasonal influenza in the US: measuring disease burden and costs. Vaccine. 2007;25:5086–5096. doi: 10.1016/j.vaccine.2007.03.046. [DOI] [PubMed] [Google Scholar]
- 6.Monto AS, Ansaldi F, Aspinall R, McElhaney JE, Montaño LF, Nichol KL, et al. Influenza control in the 21st century: optimizing protection of older adults. Vaccine. 2009;27:5043–5053. doi: 10.1016/j.vaccine.2009.06.032. [DOI] [PubMed] [Google Scholar]
- 7.Carrat F, Flahault A. Influenza vaccine: the challenge of antigenic drift. Vaccine. 2007;25:6852–6862. doi: 10.1016/j.vaccine.2007.07.027. [DOI] [PubMed] [Google Scholar]
- 8.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]
- 9.Cole SL, Ho LP. 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]
- 10.Couceiro JN, Paulson JC, Baum LG. 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]
- 11.Kowalczyk AP, Green KJ. Structure, function, and regulation of desmosomes. Prog Mol Biol Transl Sci. 2013;116:95–118. doi: 10.1016/B978-0-12-394311-8.00005-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Whitsett JA, Alenghat T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat Immunol. 2015;16:27–35. doi: 10.1038/ni.3045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wu NH, Yang W, Beineke A, Dijkman R, Matrosovich M, Baumgärtner W, et al. 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]
- 14.Ganesan S, Comstock AT, Sajjan US. Barrier function of airway tract epithelium. Tissue Barriers. 2013;1:e24997. doi: 10.4161/tisb.24997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Barbier D, Garcia-Verdugo I, Pothlichet J, Khazen R, Descamps D, Rousseau K, et al. 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]
- 16.Buchweitz JP, Harkema JR, Kaminski NE. 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]
- 17.Chong KT, Thangavel RR, Tang X. Enhanced expression of murine β-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]
- 18.Ehre C, Worthington EN, Liesman RM, Grubb BR, Barbier D, O’Neal WK, et al. Overexpressing mouse model demonstrates the protective role of Muc5ac in the lungs. Proc Natl Acad Sci USA. 2012;109:16528–16533. doi: 10.1073/pnas.1206552109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ryan LK, Dai J, Yin Z, Megjugorac N, Uhlhorn V, Yim S, et al. Modulation of human β-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]
- 20.Barlow PG, Svoboda P, Mackellar A, Nash AA, York IA, Pohl J, et al. 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]
- 21.Tripathi S, Tecle T, Verma A, Crouch E, White M, Hartshorn KL. 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]
- 22.Denney L, Ho LP. The role of respiratory epithelium in host defence against influenza virus infection. Biomed J. 2018;41:218–233. doi: 10.1016/j.bj.2018.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Moyron-Quiroz JE, Rangel-Moreno J, Kusser K, Hartson L, Sprague F, Goodrich S, et al. Role of inducible bronchus associated lymphoid tissue (iBALT) in respiratory immunity. Nat Med. 2004;10:927–934. doi: 10.1038/nm1091. [DOI] [PubMed] [Google Scholar]
- 24.Iversen MB, Reinert LS, Thomsen MK, Bagdonaite I, Nandakumar R, Cheshenko N, et al. An innate antiviral pathway acting before interferons at epithelial surfaces. Nat Immunol. 2016;17:150–158. doi: 10.1038/ni.3319. [DOI] [PubMed] [Google Scholar]
- 25.Samji T. Influenza A: understanding the viral life cycle. Yale J Biol Med. 2009;82:153–159. [PMC free article] [PubMed] [Google Scholar]
- 26.Leung YH, Nicholls JM, Ho CK, Sia SF, Mok CK, Valkenburg SA, et al. 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]
- 27.Diebold SS, 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]
- 28.Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, et al. 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]
- 29.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]
- 30.Seo SU, Kwon HJ, Song JH, Byun YH, Seong BL, Kawai T, et al. 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]
- 31.Tsai SY, Segovia JA, Chang TH, Morris IR, Berton MT, Tessier PA, et al. 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]
- 32.Teijaro JR, Walsh KB, Rice S, Rosen H, Oldstone MB. Mapping the innate signaling cascade essential for cytokine storm during influenza virus infection. Proc Natl Acad Sci USA. 2014;111:3799–3804. doi: 10.1073/pnas.1400593111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhang SY, Jouanguy E, Ugolini S, Smahi A, Elain G, Romero P, et al. TLR3 deficiency in patients with herpes simplex encephalitis. Science. 2007;317:1522–1527. doi: 10.1126/science.1139522. [DOI] [PubMed] [Google Scholar]
- 34.Bernard JJ, Cowing-Zitron C, Nakatsuji T, Muehleisen B, Muto J, Borkowski AW, et al. 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]
- 35.Takemura N, Kawasaki T, Kunisawa J, Sato S, Lamichhane A, Kobiyama K, et al. 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]
- 36.Le Goffic R, Pothlichet J, Vitour D, Fujita T, Meurs E, Chignard M, et al. 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]
- 37.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]
- 38.Teng F, Slavik V, Duffy KE, 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]
- 39.Guillot L, Le Goffic R, Bloch S, Escriou N, Akira S, Chignard M, et al. 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]
- 40.Le Goffic R, Balloy V, Lagranderie M, Alexopoulou L, Escriou N, Flavell R, et al. 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]
- 41.Heer AK, Shamshiev A, Donda A, Uematsu S, Akira S, Kopf M, et al. 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]
- 42.Chaperot L, Blum A, Manches O, Lui G, Angel J, Molens JP, et al. Virus or TLR agonists induce TRAIL-mediated cytotoxic activity of plasmacytoid dendritic cells. J Immunol. 2006;176:248–255. doi: 10.4049/jimmunol.176.1.248. [DOI] [PubMed] [Google Scholar]
- 43.Kadowaki N, Ho S, Antonenko S, Malefyt RW, Kastelein RA, Bazan F, et al. Subsets of human dendritic cell precursors express different Toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–869. doi: 10.1084/jem.194.6.863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Sha Q, Truong-Tran AQ, Plitt JR, Beck LA, Schleimer RP. Activation of airway epithelial cells by Toll-like receptor agonists. Am J Respir Cell Mol Biol. 2004;31:358–364. doi: 10.1165/rcmb.2003-0388OC. [DOI] [PubMed] [Google Scholar]
- 45.Sykes A, Edwards MR, Macintyre J, Del Rosario A, Gielen V, Haas J, et al. TLR3, TLR4 and TLRs7–9 induced interferons are not impaired in airway and blood cells in well controlled asthma. PLoS One. 2013;8:e65921. doi: 10.1371/journal.pone.0065921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ioannidis I, Ye F, McNally B, Willette M, Flaño 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]
- 47.Jeisy-Scott V, Kim JH, Davis WG, Cao W, Katz JM, 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]
- 48.Ryu JH, Kim CH, Yoon JH. Innate immune responses of the airway epithelium. Mol Cells. 2010;30:173–183. doi: 10.1007/s10059-010-0146-4. [DOI] [PubMed] [Google Scholar]
- 49.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]
- 50.Rothenfusser S, Goutagny N, DiPerna G, Gong M, Monks BG, Schoenemeyer A, et al. The RNA helicase Lgp2 inhibits TLR-independent sensing of viral replication by retinoic acid-inducible gene-I. J Immunol. 2005;175:5260–5268. doi: 10.4049/jimmunol.175.8.5260. [DOI] [PubMed] [Google Scholar]
- 51.Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, et al. 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]
- 52.Liu G, Park HS, Pyo HM, 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]
- 53.Slater L, Bartlett NW, Haas JJ, Zhu J, Message SD, Walton RP, et al. 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]
- 54.Opitz B, Rejaibi A, Dauber B, Eckhard J, Vinzing M, Schmeck B, et al. IFNβ 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]
- 55.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]
- 56.Pichlmair A, Schulz O, Tan CP, Näslund TI, Liljeström P, Weber F, et al. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5′-phosphates. Science. 2006;314:997–1001. doi: 10.1126/science.1132998. [DOI] [PubMed] [Google Scholar]
- 57.Honda A, Mizumoto K, Ishihama A. Identification of the 5′ terminal structure of influenza virus genome RNA by a newly developed enzymatic method. Virus Res. 1998;55:199–206. doi: 10.1016/s0168-1702(98)00048-3. [DOI] [PubMed] [Google Scholar]
- 58.Plotch SJ, Tomasz J, Krug RM. Absence of detectable capping and methylating enzymes in influenza virions. J Virol. 1978;28:75–83. doi: 10.1128/jvi.28.1.75-83.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Liedmann S, Hrincius ER, Guy C, Anhlan D, Dierkes R, Carter R, et al. Viral suppressors of the RIG-I-mediated interferon response are pre-packaged in influenza virions. Nat Commun. 2014;5:5645. doi: 10.1038/ncomms6645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Guo Z, Chen LM, Zeng H, Gomez JA, Plowden J, Fujita T, et al. NS1 protein of influenza A virus inhibits the function of intracytoplasmic pathogen sensor, RIG-I. Am J Respir Cell Mol Biol. 2007;36:263–269. doi: 10.1165/rcmb.2006-0283RC. [DOI] [PubMed] [Google Scholar]
- 61.Mibayashi M, Martínez-Sobrido L, Loo YM, Cárdenas WB, Gale M, Jr, García-Sastre A. Inhibition of retinoic acid-inducible gene I-mediated induction of β interferon by the NS1 protein of influenza A virus. J Virol. 2007;81:514–524. doi: 10.1128/JVI.01265-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gack MU, Albrecht RA, Urano T, Inn KS, Huang IC, Carnero E, et al. Influenza A virus NS1 targets the ubiquitin ligase TRIM25 to evade recognition by the host viral RNA sensor RIG-I. Cell Host Microbe. 2009;5:439–449. doi: 10.1016/j.chom.2009.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Weber M, Sediri H, Felgenhauer U, Binzen I, Bänfer S, Jacob R, et al. Influenza virus adaptation PB2-627K modulates nucleocapsid inhibition by the pathogen sensor RIG-I. Cell Host Microbe. 2015;17:309–319. doi: 10.1016/j.chom.2015.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Poeck H, Bscheider M, Gross O, Finger K, Roth S, Rebsamen M, et al. Recognition of RNA virus by RIG-I results in activation of CARD9 and inflammasome signaling for interleukin 1β production. Nat Immunol. 2010;11:63–69. doi: 10.1038/ni.1824. [DOI] [PubMed] [Google Scholar]
- 65.Rintahaka J, Wiik D, Kovanen PE, 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]
- 66.Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. 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]
- 67.Kandasamy M, Suryawanshi A, Tundup S, Perez JT, Schmolke M, Manicassamy S, et al. 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]
- 68.Benitez AA, Panis M, Xue J, Varble A, Shim JV, Frick AL, et al. 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]
- 69.Crotta S, Davidson S, Mahlakoiv T, Desmet CJ, Buckwalter MR, Albert ML, et al. 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]
- 70.Chen IY, Ichinohe T. Response of host inflammasomes to viral infection. Trends Microbiol. 2015;23:55–63. doi: 10.1016/j.tim.2014.09.007. [DOI] [PubMed] [Google Scholar]
- 71.Rathinam VA, Fitzgerald KA. Inflammasome complexes: emerging mechanisms and effector functions. Cell. 2016;165:792–800. doi: 10.1016/j.cell.2016.03.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Malik A, Kanneganti TD. Inflammasome activation and assembly at a glance. J Cell Sci. 2017;130:3955–3963. doi: 10.1242/jcs.207365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41:1012–1021. doi: 10.1016/j.tibs.2016.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Netea MG, Simon A, van de Veerdonk F, Kullberg BJ, Van der Meer JW, Joosten LA. IL-1β processing in host defense: beyond the inflammasomes. PLoS Pathog. 2010;6:e1000661. doi: 10.1371/journal.ppat.1000661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kaplanski G. Interleukin-18: biological properties and role in disease pathogenesis. Immunol Rev. 2018;281:138–153. doi: 10.1111/imr.12616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Duncan JA, Canna SW. The NLRC4 inflammasome. Immunol Rev. 2018;281:115–123. doi: 10.1111/imr.12607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Subramanian N, Natarajan K, Clatworthy MR, Wang Z, Germain RN. 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]
- 78.Chen G, Shaw MH, Kim YG, Nuñez 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]
- 79.Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, Franchi L, et al. 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]
- 80.Sutterwala FS, Ogura Y, Szczepanik M, Lara-Tejero M, Lichtenberger GS, Grant EP, et al. 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]
- 81.Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–232. doi: 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
- 82.Fink SL, Cookson BT. 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]
- 83.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]
- 84.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]
- 85.Bianchi ME. 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]
- 86.van Zoelen MA, van der Sluijs KF, Achouiti A, Florquin S, Braun-Pater JM, Yang H, et al. 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]
- 87.Allen IC, Scull MA, Moore CB, Holl EK, McElvania-TeKippe E, Taxman DJ, et al. 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]
- 88.Tran HB, Lewis MD, Tan LW, Lester SE, Baker LM, Ng J, et al. Immunolocalization of NLRP3 inflammasome in normal murine airway epithelium and changes following induction of ovalbumin-induced airway inflammation. J Allergy (Cairo) 2012;2012:819176. doi: 10.1155/2012/819176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kostadinova E, Chaput C, Gutbier B, Lippmann J, Sander LE, Mitchell TJ, et al. 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]
- 90.Stasakova J, Ferko B, Kittel C, Sereinig S, Romanova J, Katinger H, et al. 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 1β and 18. J Gen Virol. 2005;86:185–195. doi: 10.1099/vir.0.80422-0. [DOI] [PubMed] [Google Scholar]
- 91.Lam WY, Tang JW, Yeung AC, Chiu LC, Sung JJ, Chan PK. 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]
- 92.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]
- 93.Katze MG, He Y, Gale M., Jr Viruses and interferon: a fight for supremacy. Nat Rev Immunol. 2002;2:675–687. doi: 10.1038/nri888. [DOI] [PubMed] [Google Scholar]
- 94.Moreno M, Molling JW, von Mensdorff-Pouilly S, Verheijen RH, Hooijberg E, Kramer D, et al. IFN-γ-producing human invariant NKT cells promote tumor-associated antigen-specific cytotoxic T cell responses. J Immunol. 2008;181:2446–2454. doi: 10.4049/jimmunol.181.4.2446. [DOI] [PubMed] [Google Scholar]
- 95.Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P. Influenza-activated ILC1s contribute to antiviral immunity partially influenced by differential GITR expression. Front Immunol. 2018;9:505. doi: 10.3389/fimmu.2018.00505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Matikainen S, Paananen A, Miettinen M, Kurimoto M, Timonen T, Julkunen I, et al. IFN-α and IL-18 synergistically enhance IFN-γ production in human NK cells: differential regulation of Stat4 activation and IFN-γ gene expression by IFN-α and IL-12. Eur J Immunol. 2001;31:2236–2245. [PubMed] [Google Scholar]
- 97.Wack A, Terczyńska-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]
- 98.Donnelly RP, Kotenko SV. Interferon-λ: 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]
- 99.Kotenko SV, Durbin JE. 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]
- 100.Galani IE, Triantafyllia V, Eleminiadou EE, Koltsida O, Stavropoulos A, Manioudaki M, et al. Interferon-γ mediates non-redundant front-line antiviral protection against influenza virus infection without compromising host fitness. Immunity. 2017;46:875–890.e6. doi: 10.1016/j.immuni.2017.04.025. [DOI] [PubMed] [Google Scholar]
- 101.Davidson S, Crotta S, McCabe TM, Wack A. Pathogenic potential of interferon αβ in acute influenza infection. Nat Commun. 2014;5:3864. doi: 10.1038/ncomms4864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Klinkhammer J, Schnepf D, Ye L, Schwaderlapp M, Gad HH, Hartmann R, et al. IFN-λ prevents influenza virus spread from the upper airways to the lungs and limits virus transmission. Elife. 2018;7:e33354. doi: 10.7554/eLife.33354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Platanias LC. 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]
- 104.Krug RM, 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]
- 105.Lindenmann J, Lane CA, Hobson D. The resistance of A2G mice to myxoviruses. J Immunol. 1963;90:942–951. [PubMed] [Google Scholar]
- 106.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]
- 107.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]
- 108.Mänz B, Dornfeld D, Götz V, Zell R, Zimmermann P, Haller O, et al. 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]
- 109.Xiao H, Killip MJ, Staeheli P, Randall RE, 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]
- 110.Zimmermann P, Mänz 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]
- 111.Friedman RL, Manly SP, McMahon M, Kerr IM, Stark GR. Transcriptional and posttranscriptional regulation of interferon-induced gene expression in human cells. Cell. 1984;38:745–755. doi: 10.1016/0092-8674(84)90270-8. [DOI] [PubMed] [Google Scholar]
- 112.Bailey CC, Huang IC, 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]
- 113.Everitt AR, Clare S, Pertel T, John SP, Wash RS, Smith SE, et al. GenISIS Investigators; MOSAIC Investigators. 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]
- 114.Amini-Bavil-Olyaee S, Choi YJ, Lee JH, Shi M, Huang IC, Farzan M, et al. The antiviral effector IFITM3 disrupts intracellular cholesterol homeostasis to block viral entry. Cell Host Microbe. 2013;13:452–464. doi: 10.1016/j.chom.2013.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhang YH, Zhao Y, Li N, Peng YC, Giannoulatou E, Jin RH, et al. 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]
- 116.Bailey CC, Zhong G, Huang IC, Farzan M. IFITM-family proteins: the cell’s first line of antiviral defense. Annu Rev Virol. 2014;1:261–283. doi: 10.1146/annurev-virology-031413-085537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Lazear HM, Lancaster A, Wilkins C, Suthar MS, Huang A, Vick SC, et al. IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling. PLoS Pathog. 2013;9:e1003118. doi: 10.1371/journal.ppat.1003118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Prakash A, Smith E, Lee CK, Levy DE. Tissue-specific positive feedback requirements for production of type I interferon following virus infection. J Biol Chem. 2005;280:18651–18657. doi: 10.1074/jbc.M501289200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Izaguirre A, Barnes BJ, Amrute S, Yeow WS, Megjugorac N, Dai J, et al. Comparative analysis of IRF and IFN-α expression in human plasmacytoid and monocyte-derived dendritic cells. J Leukoc Biol. 2003;74:1125–1138. doi: 10.1189/jlb.0603255. [DOI] [PubMed] [Google Scholar]
- 120.Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature. 2005;434:772–777. doi: 10.1038/nature03464. [DOI] [PubMed] [Google Scholar]
- 121.Bosco A, Wiehler S, Proud D. Interferon regulatory factor 7 regulates airway epithelial cell responses to human rhinovirus infection. BMC Genomics. 2016;17:76. doi: 10.1186/s12864-016-2405-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.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]
- 123.Colina R, Costa-Mattioli M, Dowling RJ, Jaramillo M, Tai LH, Breitbach CJ, et al. Translational control of the innate immune response through IRF-7. Nature. 2008;452:323–328. doi: 10.1038/nature06730. [DOI] [PubMed] [Google Scholar]
- 124.Ciancanelli MJ, Huang SX, Luthra P, Garner H, Itan Y, Volpi S, et al. 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]
- 125.Stegemann-Koniszewski S, Jeron A, Gereke M, Geffers R, Kröger A, Gunzer M, et al. Alveolar type II epithelial cells contribute to the anti-influenza A virus response in the lung by integrating pathogen- and microenvironment-derived signals. MBio. 2016;7:e00276-16. doi: 10.1128/mBio.00276-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Koyama S, Ishii KJ, Kumar H, Tanimoto T, Coban C, Uematsu S, et al. 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]
- 127.Chan MC, Cheung CY, Chui WH, Tsao SW, Nicholls JM, Chan YO, et al. 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]
- 128.Chan MC, Chan RW, Yu WC, Ho CC, Chui WH, Lo CK, et al. Influenza H5N1 virus infection of polarized human alveolar epithelial cells and lung microvascular endothelial cells. Respir Res. 2009;10:102. doi: 10.1186/1465-9921-10-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Vareille M, Kieninger E, Edwards MR, Regamey N. The airway epithelium: soldier in the fight against respiratory viruses. Clin Microbiol Rev. 2011;24:210–229. doi: 10.1128/CMR.00014-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Wang J, Nikrad MP, Phang T, Gao B, Alford T, Ito Y, et al. Innate immune response to influenza A virus in differentiated human alveolar type II cells. Am J Respir Cell Mol Biol. 2011;45:582–591. doi: 10.1165/rcmb.2010-0108OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yu WC, Chan RW, Wang J, Travanty EA, Nicholls JM, Peiris JS, et al. Viral replication and innate host responses in primary human alveolar epithelial cells and alveolar macrophages infected with influenza H5N1 and H1N1 viruses. J Virol. 2011;85:6844–6855. doi: 10.1128/JVI.02200-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Xing Z, Harper R, Anunciacion J, Yang Z, Gao W, Qu B, et al. 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]
- 133.Chan MC, Chan RW, Yu WC, Ho CC, Yuen KM, Fong JH, et al. 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]
- 134.Lam WY, Yeung AC, Chu IM, Chan PK. 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]
- 135.Denney L, Branchett W, Gregory LG, Oliver RA, Lloyd CM. Epithelial-derived TGF-β1 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]
- 136.Sanders CJ, Doherty PC, Thomas PG. 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]
- 137.Herold S, von Wulffen W, Steinmueller M, Pleschka S, Kuziel WA, Mack M, et al. 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]
- 138.Perrone LA, Plowden JK, García-Sastre A, Katz JM, Tumpey TM. 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]
- 139.Kok WL, Denney L, Benam K, Cole S, Clelland C, McMichael AJ, et al. 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]
- 140.Cole SL, Dunning J, Kok WL, Benam KH, Benlahrech A, Repapi E, et al. MOSAIC investigators. 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]
- 141.Dawson TC, Beck MA, Kuziel WA, 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]
- 142.Lin KL, Suzuki Y, Nakano H, Ramsburg E, Gunn MD. 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]
- 143.Lin SJ, Lo M, Kuo RL, Shih SR, Ojcius DM, Lu J, et al. 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]
- 144.Dessing MC, van der Sluijs KF, Florquin S, van der Poll T. Monocyte chemoattractant protein 1 contributes to an adequate immune response in influenza pneumonia. Clin Immunol. 2007;125:328–336. doi: 10.1016/j.clim.2007.08.001. [DOI] [PubMed] [Google Scholar]
- 145.Maelfait J, Roose K, Vereecke L, Mc Guire C, Sze M, Schuijs MJ, et al. A20 deficiency in lung epithelial cells protects against influenza A virus infection. PLoS Pathog. 2016;12:e1005410. doi: 10.1371/journal.ppat.1005410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kanneganti TD. 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]
- 147.Thomas PG, Dash P, Aldridge JR, Jr, Ellebedy AH, Reynolds C, Funk AJ, et al. 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]
- 148.Dinarello CA. 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]
- 149.Tamaru M, Tomura K, Sakamoto S, Tezuka K, Tamatani T, Narumi S. Interleukin-1β 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]
- 150.Schmitz N, Kurrer M, Bachmann MF, 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]
- 151.Hennet T, Ziltener HJ, 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]
- 152.Yang Y, Bin W, Aksoy MO, Kelsen SG. Regulation of interleukin-1β and interleukin-1β inhibitor release by human airway epithelial cells. Eur Respir J. 2004;24:360–366. doi: 10.1183/09031936.04.00089703. [DOI] [PubMed] [Google Scholar]
- 153.Schultz-Cherry S, Hinshaw VS. Influenza virus neuraminidase activates latent transforming growth factor β. J Virol. 1996;70:8624–8629. doi: 10.1128/jvi.70.12.8624-8629.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Gibbs JD, Ornoff DM, Igo HA, Zeng JY, Imani F. Cell cycle arrest by transforming growth factor β1 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]
- 155.Williams AE, Humphreys IR, Cornere M, Edwards L, Rae A, Hussell T. TGF-β prevents eosinophilic lung disease but impairs pathogen clearance. Microbes Infect. 2005;7:365–374. doi: 10.1016/j.micinf.2004.11.012. [DOI] [PubMed] [Google Scholar]
- 156.Carlson CM, Turpin EA, Moser LA, O’Brien KB, Cline TD, Jones JC, et al. Transforming growth factor-β: 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]
- 157.Meliopoulos VA, Van de Velde LA, Van de Velde NC, Karlsson EA, Neale G, Vogel P, et al. An epithelial integrin regulates the amplitude of protective lung interferon responses against multiple respiratory pathogens. PLoS Pathog. 2016;12:e1005804. doi: 10.1371/journal.ppat.1005804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Benam KH, Königshoff M, Eickelberg O. Breaking the in vitro barrier in respiratory medicine: engineered microphysiological systems for chronic obstructive pulmonary disease and beyond. Am J Respir Crit Care Med. 2018;197:869–875. doi: 10.1164/rccm.201709-1795PP. [DOI] [PubMed] [Google Scholar]
- 159.Becher B, Waisman A, Lu LF. Conditional gene-targeting in mice: problems and solutions. Immunity. 2018;48:835–836. doi: 10.1016/j.immuni.2018.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Perl AK, Zhang L, Whitsett JA. Conditional expression of genes in the respiratory epithelium in transgenic mice: cautionary notes and toward building a better mouse trap. Am J Respir Cell Mol Biol. 2009;40:1–3. doi: 10.1165/rcmb.2008-0011ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Toxic alert [editorial] Nature. 2007;449:378. doi: 10.1038/449378a. [DOI] [PubMed] [Google Scholar]
- 162.Eden E, Geva-Zatorsky N, Issaeva I, Cohen A, Dekel E, Danon T, et al. Proteome half-life dynamics in living human cells. Science. 2011;331:764–768. doi: 10.1126/science.1199784. [DOI] [PubMed] [Google Scholar]
- 163.Chen CY, Ezzeddine N, Shyu AB. Messenger RNA half-life measurements in mammalian cells. Methods Enzymol. 2008;448:335–357. doi: 10.1016/S0076-6879(08)02617-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Baron RM, Choi AJ, Owen CA, Choi AM. Genetically manipulated mouse models of lung disease: potential and pitfalls. Am J Physiol Lung Cell Mol Physiol. 2012;302:L485–L497. doi: 10.1152/ajplung.00085.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

