Abstract
Influenza viruses cause mild to moderate respiratory illness in most people, and only rarely devastating or fatal infections. The virulence factors encoded by viral genes can explain seasonal or geographic differences at the population level but are unlikely to account for inter-individual clinical variability. Inherited or acquired immunodeficiencies may thus underlie severe cases of influenza. The critical role of host genes was first demonstrated by forward genetics in inbred mice, with the identification of interferon (IFN)-α/β-inducible Mx as a canonical influenza susceptibility gene. Reverse genetics has subsequently characterized the in vivo role of other mouse genes involved in IFN-α/β and –λ immunity. A series of in vitro studies with mouse and human cells have also refined the cell-intrinsic mechanisms of protection against influenza viruses. Population-based human genetic studies have not yet uncovered variants with a significant impact. Interestingly, human primary immunodeficiencies affecting T and B cells were also not found to predispose to severe influenza. Recently however, human IRF7 was shown to be essential for IFN-α/β- and IFN-λ-dependent protective immunity against primary influenza in vivo, as inferred patient with life-threatening influenza revealed to be IRF7-deficient by whole exome sequencing. Next generation sequencing of human exomes and genomes will facilitate the analysis of the human genetic determinism of severe influenza.
Introduction
The enormous human inter-individual variability of severity of infectious diseases is a century old medical enigma [1-3]. For the vast majority of microorganisms, only a small minority of infected individuals develops life-threatening infections. The colossal burden of infectious diseases mostly results from their enormous diversity. A good example is influenza, which typically causes a self-limiting infection that resolves within one week. In contrast, a small proportion of individuals develop life-threatening and occasionally fatal infections. Global influenza pandemics have tantalized researchers for a century, from the pandemics of 1918 through 2009 to the threats of pandemic from highly pathogenic avian influenza in the past decades. The many fascinating examples of virulence factors encoded by influenza A viruses (IAVs) [4,5] have been tested by viral genetics. Likewise, the emergence of any new IAV by reshuffling of RNA segments from two or more viruses typically accounts for each new pandemic. These viral genetic variations provide a compelling explanation for the emergence of new, more virulent viruses and therefore for inter-population variability of clinical outcomes in the course of influenza. They however can hardly account for inter-individual variability in a given human population, especially of small size.
For any given IAV, whether responsible for a devastating pandemic or a banal seasonal epidemic, only a minority of infected individuals develops severe sickness, including pneumonia and acute respiratory distress syndrome. The proportion of severe disease varies from about 0.04-0.4% of symptomatic cases during seasonal influenza to about 1-10% symptomatic cases being fatal during the worst known influenza pandemic in 1918-1919 [6,7]. A fundamental problem in the field of influenza is therefore that of inter-individual variability in the course of infection by any given influenza virus. Well-known risk factors have been documented epidemiologically since the identification of the first human influenza virus in 1933 by Wilson Smith [8], shortly after Richard Shope identified the first influenza virus in swine [9]. They principally include pre-existing chronic pulmonary disease, in children and adults, but apparently exclude HIV-positivity as a risk factor [10]. The vast majority of cases remain unexplained, whether during primary infection in early childhood or during re-infection later in life. Importantly, this notion applies to both epidemic and pandemic influenza. Although the proportion of severe cases can be 30-fold higher in the course of an emerging pandemic than during seasonal influenza, these cases remain the minority and unexplained in both settings [11]. We herein review the mounting evidence attributing severe influenza to inborn errors of immunity in both mice and humans, with an emphasis on discoveries made by forward genetics of primary infection.
The influenza virus
IAVs possess a genome consisting of eight ssRNA segments that encode 11-12 proteins [12]. On the virion surface are the glycoproteins, hemagglutinin (HA) and neuraminidase (NA), which mediate cellular entry and release from the infected cell, respectively, and the ion channel, M2. The matrix protein, M1, creates the structure of the virion under the lipid membrane. The ribonucleoprotein complex consists of the viral RNA (vRNA) surrounded by the nucleoprotein, NP, that interacts with the viral RNA-dependent RNA polymerase subunits: PA, PB1, and PB2. An additional open reading frame in the PB1 RNA encodes PB1-F2, a modulator of apoptosis, while the function of a truncated gene product of the PB1 gene, PB1 N40, is still unclear. NS1 is the antagonist of host antiviral immunity, capable of sequestering double-stranded RNA and thus preventing activation of multiple pathways, including RIG-I/MAVS signaling, that converge on the activation of the transcription factors, IRF3 and IRF7, and transcription of IFN-α, -β and -λ [13,14]. Lastly, NEP (also known as NS2) is the nuclear export protein. When considering the causes of severe influenza disease, most of the attention has been placed on the viral genes. For example, the 1918 pandemic strain was partially explained by contributions from the HA gene and the polymerase complex genes and the emergence of 2009 pandemic H1N1 was explained by the triple-reassortment of genes from human, swine, and avian-like swine viruses [15,16]. Hypercytokinemia induced by infiltrating macrophages and other innate immune cells in response to high viral load is a potential source of lung injury during viral clearance in humans, as suggested by mouse models [17,18]. However, depletion of murine alveolar macrophages or neutrophils in mice can exacerbate lung injury due to delayed viral clearance [19,20]. Nevertheless, work in recent decades has begun to demonstrate the role that host genes play in susceptibility to severe or fatal influenza [21].
In vivo mouse studies – Mx
Shortly after his 1957 discovery of IFN with Alick Isaacs [22], Jean Lindenmann made a chance discovery of the selective in vivo resistance of A2G mice to experimental infection by influenza virus and other myxoviruses [23-25]. The connection to IFN was proven using antiserum against mouse IFN that could ablate the natural protection from influenza enjoyed by the A2G mouse [26,27]. Mx, located on chromosome 16 and encoding a 72kDa IFN-inducible protein, Mx1, is the first mouse gene discovered by forward genetics of infectious diseases, following an expression cloning approach [28-30]. Mx1 deficiency, common in most inbred laboratory mouse strains, due to a nonsense mutation in CBA/J mice and a large deletion causing a frameshift and a premature stop codon in BALB/c mice, leads to profound susceptibility to influenza virus, although mice are not natural hosts for any known influenza virus [31]. The formal demonstration of Mx1 being the critical missing factor in these strains came from the transgenic expression of murine Mx1 or human MxA, which rescued the susceptibility phenotype [32,33]. Mx1 is a dynamin-like GTPase that, in the mouse, localizes to the nucleus. It prevents transcription by the viral RNA polymerase, effectively blocking both transcription and replication of the orthomyxviruses, which use a nuclear replication strategy [34-37]. The GTPase domain is critical for Mx1 binding to the viral ribonucleoprotein complex and the inhibition of transcription [38]. The IAV nucleoprotein is the main determinant of Mx1 sensitivity and various viruses, including avian strains like H7N9, have acquired mutations in NP that evade Mx1-mediated defense in mice [39,40]. Interestingly, DBA/2J mice (an Mx1-deficient mouse strain) crossed with A2G mice to express the wild-type Mx1 allele remain highly susceptible to H1N1 IAV [41]. Thus, in some cases the presence of Mx1 alone is insufficient for protection from IAV infection, indicating that regulation conferred by the genetic background and/or the influence of other alleles are at play and should be sought.
In vivo mouse studies – genome-wide screens
Large-scale genome-wide studies offer the ability to identify novel pathways, genes and genetic variants affecting host control of IAV replication. The Collaborative Cross is an effort to measure the impact of genotypic diversity in eight inbred and three outbred mouse strains on multiple disease phenotypes including immune responses to infectious disease. Quantitative trait locus (QTL) mapping in the 8 inbred founders infected with IAV found several new candidate genes (Grap2, Nox4, and Il16) and led to the identification of a novel Mx1 allele in CAST/EiJ with a reduced ability to inhibit IAV replication [42]. Additionally, QTL mapping identified sex-dependent effects in 29 closely-related mouse strains and novel candidates – Hc, Pla2g7 and Tnfrsf21 [43]. Several QTLs on chromosome 6, including Samd9l and Ica1, were found by linkage analysis on cytokine production following infection with highly pathogenic H5N1 IAV [44]. Recently, a novel in vivo screen consisting of a library of siRNA-encoding IAVs that each target one of 100 IAV-induced genes identified a previously unappreciated role for Ifih1 (encoding melanoma-differentiation-associated gene 5 or MDA5), previously known for its recognition of positive-strand RNA viruses [45,46], in the immune response against IAV. Those viruses that targeted a necessary component of the innate immune response became more fit in the context of an in vivo infection and were identified by RNA-sequencing of the infected mouse lung [47].
In vivo mouse studies of IFN-inducing genes - reverse genetics
The type I and III IFN signaling pathways are central to the immune response against influenza virus (reviewed by [48]). For some constituent parts of these pathways, their roles have been demonstrated in vivo by reverse genetics, i.e. by knockout (KO) studies (see Table 1). Ifnb1−/− knockout mice are substantially more sensitive to IAV even in an Mx1-positive mouse [49]. In vitro studies showed that IAV infection is primarily recognized in hematopoietic cells by TLR7-MyD88 signaling [50,51] and in non-hematopoietic cells by RIG-I-MAVS signaling [46,52,53]. Mice deficient for Unc93b1, the adapter for endosomal TLRs – TLR3, 7, 8, and 9, show increased tissue pathology due to delayed viral clearance of influenza virus A/PR/8/34 infection [54]. However, Tlr3−/− mice display decreased pathology and prolonged survival despite higher IAV lung titers pointing to TLR3 signaling in the promotion of inflammatory and adaptive responses that are ultimately damaging to the lung [55]. The Myd88/Mavs double knockout that cripples both signaling arms causes these mice to succumb to a lethal dose of IAV with more than ten-fold higher lung titers [56]. Mavs−/− mice cope with infection like wild-type mice [56], whereas studies have conflicted on the absolute requirement for TLR7/MyD88 signaling for control of viral replication and development of IAV-specific antibodies in KO mice [56-59]. It should be noted that the above studies are performed in Mx1-deficient mice. Another study showed the necessity of TLR7 and MyD88 for survival after H7N7 IAV infection in an Mx1-positive mouse model [60]. This reflects the critical role of Mx1 as an influenza restriction factor and the potential need to revisit many in vivo KO studies performed in Mx1-deficient mouse models [5,31]. Lastly, a recent study identified Irf7 among 25 differentially expressed genes (DEGs) between C57BL/6J and DBA/2J mice infected with IAV and that overlapped with DEGs from other published studies. Irf7−/− C57BL/6 mice (Mx1-negative) lost significantly more body weight with poor survival compared to wild-type control mice [61].
Table 1.
Reverse genetics in mice of IFN-inducing and IFN-inducible genes
| gene knockout | protein name | mouse strain | Mx-background | virus strain | virus subtype | viral phenotype | morbidity | mortality | reference | |
|---|---|---|---|---|---|---|---|---|---|---|
| IFN-inducing | Ifnb−/− | IFN-β | B6.A2G.Mx1 | +/+ | SC35M and A/PR/8/34 | H7N7; H1N1 | elevated lung titer, decreased LD50 | no data | increased | Koerner et al 2007 (49) |
| Ifnar1−/− | IFNAR1 | B6.A2G.Mx1 | +/+ | SC35M | H7N7 | dramatically decreased LD50 | no data | increased | ||
| Ifnar1−/− | IFNAR1 | 129 | −/− | A/WSN/33 and A/PR/8/34 | H1N1 | elevated lung titers | disseminated infection with hepatic and brain pathology | no data | Garcia-Sastre et al 1998 (63) | |
| Stat1−/− | STAT1 | C57BL/6 | −/− | no data | ||||||
| Ikbke−/− | IKK-ε | C57BL/6 | −/− | A/WSN/33 | H1N1 | no data | increased | increased | tenOever et al 2007 (64) | |
| Tlr7−/− | TLR7 | B6.A2G.Mx1 | +/+ | SC35M | H7N7 | elevated lung titers | no data | increased | Kaminski et al 2012 (60) | |
| Tlr7−/− | TLR7 | C57BL/6 | −/− | A/PR/8/34 | H1N1 | equal to wt mice | decreased cytokine production | survive sublethal infection | Pang et al 2013 (59) | |
| Mavs−/− | MAVS | −/− | equal to wt mice | decreased cytokine production | survive sublethal infection | |||||
| Tlr7−/− Mavs−/− | TLR7 and MAVS | −/− | reduced virus titer | no data | no data | |||||
| Tlr7−/− | TLR7 | C57BL/6 | −/− | A/PR/8/34 | H1N1 | low antibody titers | no data | decreased | Jeisy-Scott et al 2012 (57) | |
| Myd88−/− | MyD88 | −/− | low antibody titers | no data | decreased | |||||
| Myd88−/− Trif−/− | MyD88 and TRIF | C57BL/6 | −/− | A/PR/8/34 | H1N1 | elevated lung titers | no difference in weight loss | increased | Seo et al 2010 (58) | |
| Tlr3−/− Tlr7−/− | TLR3 and TLR7 | C57BL/6 | −/− | A/PR/8/34 | H1N1 | no data | increased | |||
| Myd88−/− | MyD88 | B6.A2G.Mx1 | +/+ | SC35M | H7N7 | elevated lung and brain titers | neurotropic infection | increased | Kaminski et al 2012 (60) | |
| Myd88−/− | MyD88 | C57BL/6 | −/− | A/New Caledonia/20/99 | H1N1 | normal lung titers | no data | no data | Koyama et al 2007 (56) | |
| Ips1−/− | IPS-1 | C57BL/6 | −/− | normal lung titers | no data | no data | ||||
| Myd88−/− Ips1−/− | MYD88/IPS-1 | C57BL/6 | −/− | elevated lung titers | no data | no data | ||||
| Unc93b1−/− | UNC93B | C57BL/6 | −/− | A/PR/8/34 | H1N1 | elevated lung titers | increase lung pathology | slightly increased | Lafferty et al 2014 (54) | |
| Tlr3−/− | TLR3 | C57BL/6 | −/− | A/Scotland/20/74 | H3N2 | elevated lung titers | decreased lung pathology | decreased | Le Goffic et al 2006 (55) | |
| Irf7−/− | IRF7 | C57BL/6 | −/− | A/PR/8/34 serially passaged in Mx1−/− mice |
H1N1 | no data | increased weight loss | increased | Wilk et al 2015 (61) | |
| IFN-inducible | Isg15−/− | ISG15 | C57BL/6 | −/− | A/WSN/33 and B/Lee/40 | H1N1; - | elevated lung titers | increased inflammatory cytokine production | increased | Lenschow et al 2007 (65) |
| Eif2ak2−/− | PKR | 129/Sv | −/− | A/WSN/33 | H1N1 | elevated lung titers | increased weight loss | increased | Balachandran et al 2000 (68) | |
| Eif2ak2−/− | PKR | 129/Sv(ev)xC57BL/6J | −/− | A/PR/8/34 | H1N1 | elevated lung titers | increased weight loss | increased | Bergmann et al 2000 (70) | |
| Serpine1−/− | PAI1 | C57BL/6 | −/− | A/PR/8/34 | H1N1 | elevated lung titers | increased lung pathology and weight loss | increased | Dittmann et al 2015 (84) |
In vivo mouse studies of IFN-inducible genes - reverse genetics
Downstream of IFN production is the potential expression of >400 IFN-stimulated genes (ISGs) that are the ultimate effectors of the innate antiviral response [62]. Ifnar1−/− and Stat1−/− mice do not respond to type I and III IFNs and allow overwhelming replication of IAV resulting in systemic influenza infection that is no longer restricted to the respiratory epithelium [63]. Influenza virus infection of Ikbke−/− mice causes the production of normal amounts of type I IFN but they ultimately lack the induction of a subset of ISGs, including Adar1, Ifit3, Ifi203, due to a lack of STAT1 serine phosphorylation at position 708 by IKK-ε [64]. This reinforces the individual impact that specific ISGs, beside Mx1, can have during IAV infection. Mice deficient for ISG15, a 15kDa diubiquitin-like protein that can be similarly conjugated to viral target proteins, are more susceptible to influenza and other viruses [65,66]. However, IAV lethality in these mice is not due to higher viral titers but instead, a lack of conjugation-dependent immunity resulting in lung epithelial cell damage [67]. Double-stranded RNA (dsRNA) activated protein kinase R (PKR) phosphorylates eukaryotic initiation factor 2 alpha in response to dsRNA and inhibits protein synthesis [68]. PKR also sustains the IFN response by promoting the integrity of newly synthesized IFN-α and IFN-β mRNAs [69]. Pkr−/−mice show 2-3 log higher IAV lung titers by day 2 of infection and show higher mortality compared to wild-type mice [68,70]. In vivo studies highlight the central role of IFNs and especially their induced genes in primary infections, whereas the control of secondary infections is less dependent on innate immunity and the IFN system.
Human MX1 in vitro
Human MX1 encodes a 78kDa protein called MxA [71,72] that localizes to the cytoplasm of the cell, which differs from the nuclear murine Mx1. Human MxA thus can inhibit viruses that replicate in the cytoplasm or in the nucleus such as VSV[73], hPIV3 [74], HBV [75,76], LACV [77], ASFV [78]. MxA, along with other ISG products, binds to NP of IAV [79,80] and retains the viral genomic RNA in the cytoplasm near late endosome [79]. It has since been shown that human MxA is induced predominantly by type I and III IFNs and that its non-constitutive expression is tightly regulated by these IFNs[81]. This tight regulation is perhaps due to the link of its expression to sensitivity to apoptotic stimuli [82,83]. Several high-throughput screens in humans have identified MxA as a restriction factor for replication of IAV, VSV and Yellow Fever virus [84,85]. Interestingly, inherited MxA deficiency has not currently yet been described in humans. Human MX1 does have many heterozygous single nucleotide polymorphisms (SNPs) in the Exome Aggregation Consortium (ExAC) database, with frequencies ranging from 0.0008-0.27%, resulting in changes to the coding region including 12 heterozygous nonsense alleles. Eight of the missense alleles are present as homozygotes with 4 predicted to be damaging. None have yet been shown to be deleterious in vitro and the clinical status for these individuals, influenza infections in particular, is not reported. These data may be interpreted as suggesting that haploinsufficiency does not occur at the MX1 locus, but they certainly do not exclude potential negative dominance for some heterozygous variations. Thus, the search for humans carrying deleterious mutations in MX1, including mono- and bi-allelic variations, should be continued especially among those who suffer from severe influenza.
Other in vitro human studies
Novel antiviral pathways have been uncovered using large-scale in vitro studies of human cell lines. Shapira et al [86] combined a yeast-two-hybrid analysis of IAV genes and 12,000 human genes with a transcriptome-wide array in IAV-infected human broncheoepithelial cells (HBECs) to create a network of genes that are positively and negatively regulated during infection. Among the most prominent hits, validated by small interfering RNA (siRNA)-mediated knockdown and measurement of viral titers in HBECs, were a group of inflammasome-related sensors and RNA-binding proteins. Konig et al [87] and Karlas et al [88] each also used RNA-interference (RNAi) screens in the context of influenza replication but identified those genes critical for the replication of influenza virus and thus identified potential therapeutic targets. Pulloor et al [89] focused their RNAi screen of 18,120 genes on regulators of the RIG-I pathway, which recognizes viral genomic RNA of IAV [90], using a GFP reporter driven by the IFN-β promoter in HEK293 cells transfected with synthetic double-stranded RNA. This approach identified the cellular pathway synthesizing inositol pyrophosphates as a critical regulator of IRF3 regulation and revealed the power of a screen coupled with a careful biochemical dissection to identify a previously unknown level of regulation. Winterling et al demonstrated the role of virus-inducible large intergenic non-coding RNAs (lincRNAs) in influenza virus replication, thus revealing a novel set of host factors affecting virus replication [91]. Dittmann et al screened a library of 401 ISGs overexpressed from a lentiviral library for the ability to reduce the spread of influenza virus in A549 lung adenocarcinoma cells. Extensive in vitro and in vivo functional assays validated SERPINE1, which encodes PAI-1, as a novel inhibitor of the host proteases that leave and mature the IAV attachment glycoprotein, HA[84]. Meta-analyses of large-scale screens [92-94] show little overlap between the hits identified within the various approaches, highlighting the importance of subtle differences in the libraries, cell systems, and time points examined. Thus, this emphasizes the necessity of validation experiments to confirm the involvement of these newly identified genes and/or pathways.
Human and mouse IFITM3
Using an RNA interference (RNAi) screen targeting host factors affecting early stages of influenza virus growth in human osteosarcoma cells, Brass et al identified the IFITM proteins, IFITM1, 2 and 3, as restriction factors for influenza virus, West Nile virus, and dengue virus [95]. Independently, Yount et al employed a proteomic approach, which purified fatty-acylated proteins in mouse dendritic cells, and also identified IFITM3 for its antiviral activity against influenza virus [96]. IFITM3 is an IFN stimulated gene and resides largely in the endoplasmic reticulum. Post-translational modifications, S-palmitoylation and ubiquitination [96,97], regulate its function by relocalizing IFITM3 into the endosome where it blocks the virus fusion pore and thus the delivery of viral RNA into the cytosol. IFITM3 expression is negatively regulated by the NEDD4 E3 ligase, which targets IFITM3 for lysosomal degradation and can thus impact influenza replication [98]. IAV lung titers were higher, as were mortality and morbidity, in Ifitm3 KO mice than in wild-type littermates [99,100]. Based on these results, Everitt et al also tested the association of a SNP in IFITM3, rs12252, with a C allele causing a truncation in IFITM3 (Δ21) that is relocalized throughout the cytosol [100]. Subjects homozygous for the C allele were significantly more frequent among 53 patients from the UK hospitalized with severe influenza and further, among 32 Chinese patients with severe pH1N1 2009 infection [101]. However, the frequency of the C allele among healthy populations is quite high, especially in Asian populations: 49.7% (with 25.4% CC homozygous) in Han Chinese and 63.5% (43.8% CC homozygous) in Japanese populations [102]. Moreover, the association of rs12252 failed to reach significance with larger sample sizes [103]. This indicates that this IFITM3 SNP could not be a major causal factor, but perhaps a modifier, of the outcome of influenza infection. Additionally, recent functional studies showed that expression of IFITM3Δ21 in A549 cells restricted influenza virus comparably to wild-type IFITM3 [104]. Thus, there is convincing experimental evidence for the role of wild-type IFITM3 but further work is required to define the role of common SNPs of IFITM3 as modifiers for severe influenza.
Human in vivo studies – testing a common variant hypothesis
Common variants or SNPs have been tested in population-based studies of influenza outcomes, in “genome-wide association study (GWAS)-like” studies (reviewed in detail [105]). A SNP in CD55, rs2564978, was chosen from 5,166 SNPs identified by a small-scale GWAS study in 25 Chinese patients with severe influenza and 26 controls [106]. This SNP caused reduced expression of CD55 (also known as decay accelerating factor, DAF), which can reduce epithelial cell damage during influenza infection [106]. A similar GWAS study of 42 Chinese patients with severe pH1N1 or H7N9 infection and 42 controls crossed with a quantitative cis-QTL study in the lung yielded 1,114 SNPs. From these, a SNP (rs383510) that may regulate TMPRSS2 expression was chosen based on mouse studies showing Tmprss2 expression levels modulate infection outcomes [107,108]. Targeted screens of several candidate genes have also investigated SNPs associated with severe influenza without confirmed associations [105,109,110]. A deletion in the chemokine receptor CCR5 was reported to be enriched in a Canadian cohort suffering from severe influenza [111]. However, another study failed to reach significance in a similar cohort of European ancestry [112]. More interestingly, regulatory SNPs affecting expression of surfactant proteins, SFPTA (rs1965708 and rs1059046) in a Spanish cohort and in SFPTB (rs1130866) in a Chinese cohort [113,114], were associated with severe pH1N1 2009 influenza. Independent studies of Japanese and Chinese children who developed severe and/or fatal influenza-associated encephalopathy found significant association with exonic variants in CPT2 that result in thermolabile proteins that are hypomorphic during fever [115,116]. However overall, population-based studies have been underpowered and unreplicated and have not yet revealed major genetic determinants of severe influenza [110]. The common variants detected by these means might play a role, at best that of modifiers given the rarity of the disease, which needs to be experimentally tested [117,118].
Human in vivo studies – primary immunodeficiencies
Surprisingly, previously defined human inborn errors of immunity, including severe primary immunodeficiencies, such as severe combined immunodeficiency (a lack of T cells) or agammaglobulinemia (a lack of B cells) do not predispose children to severe influenza [119,120]. Although T and B cells are needed for generating acquired immunity to influenza virus or vaccine, these cells do not seem to be critical for the control of primary or secondary infection. This contrasts with parainfluenza viruses, which often kill children with inborn errors of T cells [119]. Remarkably, several patients with MonoMAC syndrome caused by GATA-2 deficiency died from influenza and these patients are diminished in several leukocyte subsets, including plasmacytoid dendritic cells (pDCs) [121,122]. Known inborn errors of type I and III IFNs, including STAT1, IL10RB, TLR3 and MyD88 deficiencies, have so far not been shown to have severe influenza [123,124]. Moreover, humans with ISG15 deficiency do not develop severe infections with influenza but instead demonstrate a susceptibility to mycobacterial disease and a type I interferonopathy [125,126]. Infection with influenza A and B viruses can also trigger acute necrotizing encephalitis (ANE) [127]. This is distinct from severe pulmonary influenza virus infection because cerebral spinal fluid and brain tissue of these individuals is negative for influenza and parainfluenza viruses [127]. ANE is thus not commonly seen as a PID in the traditional sense of the term. Neilson et al identified a heterozygous missense mutation, p.Thr585Met, in RANBP2 in 11 patients suffering from influenza-induced ANE [128]. RANBP2, a SUMO1 E3 ligase, regulates protein transport across the nuclear pore [129]. RANBP2 deficiency is only found in familial cases of ANE and is inherited as an autosomal dominant incompletely penetrant trait with 40% of heterozygous carriers developing ANE [128]. The connection between RANBP2 mutations and ANE remains unclear but there is no indication that those who carry these mutations have remarkable pulmonary complications from influenza. A molecular characterization is needed to pin down this mechanism and to elucidate the effect RANBP2 mutations have on virus replication and/or the immune response meant to control it.
Human monogenic lesions in vivo - inherited IRF7 deficiency
We hypothesized that severe influenza may be caused by single-gene inborn errors of immunity, as previously documented for an increasing variety of infections [1]. We enrolled 22 patients suffering from severe influenza and requiring hospitalization, among which were 3 children less than five years old. Using whole exome sequencing, we discovered compound heterozygous mutations in IRF7, encoding interferon regulatory factor 7 (IRF7), in a girl who was severely ill with influenza at 2.5 years old [130]. Both of her IRF7 alleles fail to upregulate type I IFN resulting in elevated IAV replication in fibroblasts and abolished type I and III IFN production in pDCs, which constitutively express high levels of IRF7 and produce large amounts of type I and III IFNs [131,132]. Perhaps not coincidentally, pDCs can be diminished in patients with GATA-2 deficiency [121]. The generation of airway epithelial cells from patient-specific induced pluripotent stem cells (iPSCs) allowed the demonstration of increased influenza virus spread associated with diminished IFN production due to a lack of IRF7. This represents the first demonstration of a genetic cause for severe disease influenza in humans. It is intriguing that known patients with mutations in STAT1 or IL10RB, which impair type I and III IFN signaling, have not developed severe influenza. Of note, the Irf7 KO mouse is highly susceptible to many RNA and DNA viruses and displays higher mortality when infected by IAV, while the IRF7 patient exhibited a narrower susceptibility [61,133-138]. IFN-mediated control of influenza virus has been known since the initial discovery of IFN itself [22], and the myriad ways that IAV antagonizes the IFN response underscores the in vivo role IFN plays in controlling pathogenesis [13]. The critically important question of the relative contributions of intrinsic immunity in pulmonary cells and of innate immunity from pDCs is still incompletely understood.
IRF7 in cell-intrinsic immunity against influenza virus
The essential role of mouse IRF7 in the initial induction and subsequent amplification of type I and III IFN immunity was well established by reverse genetics [133,139-143]. In a study previously discussed, IRF7 was also among the genes identified by Dittmann et al in a lentivirus-driven gain-of-function screen that selected for genes that control the spread of influenza virus in vitro [84]. Other recent forward somatic genetic studies point to a key role of IRF7 in anti-influenza immunity. A genome-wide screen of canine kidney cells showed that IRF7 knockdown led to the highest replication of influenza vaccine strain [144], while a loss of the translational repressors of IRF7, 4E-BP1 and 4E-BP2, led to dramatically lower influenza virus replication in mouse embryonic fibroblasts [145]. Moreover, a SNP diminishing IRF7 expression was associated with impaired antiviral responses in influenza-infected human DCs [146]. Such findings are consistent with the in vitro phenotypes observed in pDCs and iPSC-derived airway epithelial cells from the IRF7-deficient patient described above [130]. Further, these data, which all converged independently on IRF7, suggest that other inborn errors underlying severe influenza may be IRF7-related. For other human infections, a core gene was found, such as IFN-γ for mycobacterial diseases, IL-17 for mucocutaneous candidiasis, TLR3 for herpes simplex encephalitis, and CARD9 for invasive fungal diseases [147].
Conclusions
There is theoretical, experimental, clinical, and epidemiological evidence to propose and test the hypothesis that severe influenza in humans can be determined by inborn errors of immunity. Preliminary findings suggest that single-gene errors of innate immunity, affecting IRF7-dependent IFN-α/β and IFN-λ immunity in particular, can cause severe and life-threatening influenza. The study of influenza adds weight to the idea that severe infectious disease, in the course of primary infection, can be caused by monogenic lesions not necessarily fully penetrant [1]. Mutations that are found in humans and mice to underlie severe influenza point out the non-redundant pathways and molecules involved in protective immunity during primary infection. The clearest example is that of a single-gene inborn error of immunity, while the search for a polygenic explanation for this devastating disease has lagged behind. Of particular interest, genetic susceptibility to severe influenza in humans is probably narrow as most patients have been healthy otherwise. An understanding of the diversity of human antiviral responses and the deficiencies that confer susceptibility to each severe and fatal viral disease will fundamentally change the way we approach viral disease from a medical standpoint, including influenza in particular. The findings that can be confirmed in vivo, especially in humans, offer a clear target for manipulation in the clinic. For example, it is tempting to speculate that recombinant IFN-α may be useful in children with life-threatening influenza, especially in children deficient in an IFN-inducing gene such as IRF7. The advent of patient-tailored therapeutics and targeted functional screening based on next-generation sequencing data may augment host immunity to more effectively manage the disease course of influenza.
Highlights.
Severe influenza can be caused by single-gene inborn errors of immunity in mice and humans
Mouse Mx1 was the first influenza susceptibility gene identified by forward genetics
Inherited IRF7 deficiency caused life-threatening influenza in an otherwise healthy child
Susceptibility genes are presumably influenza-specific as most patients are healthy prior to and after severe influenza
Acknowledgments
We wish to thank the members of the Laboratory of Human Genetics of Infectious Diseases, with particular gratitude for helpful discussions and edits from Stuart Tangye, Emmanuelle Jouanguy, Carolyn Jackson, Hye Kyung Lim, Sarah Jill de Jong, Yuval Itan, Bertrand Boisson, Lahouari Amar and Yelena Nemirovskaya. We are indebted to the patients and their families for their involvement in our studies. The work was funded by National Center for Research Resources and National Center for Advancing Translational Sciences grant 8UL1TR000043; NIH grants 5R01NS072381; the Rockefeller University, the St. Giles Foundation, the French National Research Agency (ANR) under the “Investments for the future” program (grant n°ANR-10-IAHU-01), the Laboratoire d'Excellence Integrative Biology of Emerging Infectious Diseases (ANR-10-LABX-62-IBEID), INSERM, and Paris Descartes University.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Casanova JL, Abel L. The genetic theory of infectious diseases: a brief history and selected illustrations. Annu Rev Genomics Hum Genet. 2013;14:215–243. doi: 10.1146/annurev-genom-091212-153448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Casanova JL. Severe infectious diseases of childhood as monogenic inborn errors of immunity. Proceedings of the National Academy of Sciences. 2015 doi: 10.1073/pnas.1521651112. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Casanova JL. The human genetic basis of inter-individual variability in the course of infection. Proceedings of the National Academy of Sciences. 2015 doi: 10.1073/pnas.1521644112. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kash JC, Taubenberger JK. The role of viral, host, and secondary bacterial factors in influenza pathogenesis. Am J Pathol. 2015;185:1528–1536. doi: 10.1016/j.ajpath.2014.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.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]
- 6.Taubenberger JK, Morens DM. 1918 Influenza: the mother of all pandemics. Emerg Infect Dis. 2006;12:15–22. doi: 10.3201/eid1201.050979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.World Health Organization Influenza (Seasonal). WHO report 2014. Fact sheet No211. [Google Scholar]
- 8.Smith W, Andrewes CH, Laidlaw PP. A VIRUS OBTAINED FROM INFLUENZA PATIENTS. The Lancet. 222:66–68. [Google Scholar]
- 9.Shope RE. SWINE INFLUENZA: III. FILTRATION EXPERIMENTS AND ETIOLOGY. The Journal of Experimental Medicine. 1931;54:373–385. doi: 10.1084/jem.54.3.373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Martinez E, Marcos MA, Hoyo-Ulloa I, Anton A, Sanchez M, Vilella A, Larrousse M, Perez I, Moreno A, Trilla A, et al. Influenza A H1N1 in HIV-infected adults. HIV Med. 2011;12:236–245. doi: 10.1111/j.1468-1293.2010.00905.x. [DOI] [PubMed] [Google Scholar]
- 11.Jeffery T, David MM. 1918 Influenza: the Mother of All Pandemics. Emerging Infectious Disease journal. 2006;12:15. doi: 10.3201/eid1201.050979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Shaw ML, Palese P. Orthomyxoviridae. In: Knipe D, Howley P, editors. Fields Virology. 6th ed. Lippincott Williams, Wilkins; 2013. p. 1151-. [Google Scholar]
- 13.Ayllon J, García-Sastre A. The NS1 Protein: A Multitasking Virulence Factor. In: Oldstone MBA, Compans RW, editors. Influenza Pathogenesis and Control - Volume II. Springer International Publishing; 2015. pp. 73–107. Current Topics in Microbiology and Immunology, vol 386. [DOI] [PubMed] [Google Scholar]
- 14.Liu S, Cai X, Wu J, Cong Q, Chen X, Li T, Du F, Ren J, Wu Y-T, Grishin NV, et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science. 2015:347. doi: 10.1126/science.aaa2630. [DOI] [PubMed] [Google Scholar]
- 15.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]
- 16.Watanabe T, Kawaoka Y. Pathogenesis of the 1918 Pandemic Influenza Virus. PLoS Pathog. 2011;7:e1001218. doi: 10.1371/journal.ppat.1001218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cheung CY, Poon LLM, Lau AS, Luk W, Lau YL, Shortridge KF, Gordon S, Guan Y, Peiris JSM. Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? The Lancet. 2002;360:1831–1837. doi: 10.1016/s0140-6736(02)11772-7. [DOI] [PubMed] [Google Scholar]
- 18.de Jong MD, Simmons CP, Thanh TT, Hien VM, Smith GJD, Chau TNB, Hoang DM, Van Vinh Chau N, Khanh TH, Dong VC, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med. 2006;12:1203–1207. doi: 10.1038/nm1477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ghoneim HE, Thomas PG, McCullers JA. Depletion of alveolar macrophages during influenza infection facilitates bacterial superinfections. J Immunol. 2013;191:1250–1259. doi: 10.4049/jimmunol.1300014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tate MD, Ioannidis LJ, Croker B, Brown LE, Brooks AG, Reading PC. The role of neutrophils during mild and severe influenza virus infections of mice. PLoS One. 2011;6:e17618. doi: 10.1371/journal.pone.0017618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Albright FS, Orlando P, Pavia AT, Jackson GG, Cannon Albright LA. Evidence for a heritable predisposition to death due to influenza. J Infect Dis. 2008;197:18–24. doi: 10.1086/524064. [DOI] [PubMed] [Google Scholar]
- 22.Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc. R. Soc. London Ser. B. 1957;147:258–267. [PubMed] [Google Scholar]
- 23.Lindenmann J, Lane CA, Hobson D. The Resistance of A2g Mice to Myxoviruses. J Immunol. 1963;90:942–951. [PubMed] [Google Scholar]
- 24.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]
- 25.Lindenmann J. Elsevier, editor. Of Mice and Men – The Mx Connection. Comprehensive Biochemistry. 2005:267–295. vol Volume 44. [Google Scholar]
- 26.Haller O, Arnheiter H, Gresser I, Lindenmann J. Genetically determined, interferon-dependent resistance to influenza virus in mice. The Journal of Experimental Medicine. 1979;149:601–612. doi: 10.1084/jem.149.3.601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Haller O, Arnheiter H, Lindenmann J, Gresser I. Host gene influences sensitivity to interferon action selectively for influenza virus. Nature. 1980;283:660–662. doi: 10.1038/283660a0. [DOI] [PubMed] [Google Scholar]
- 28.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]
- 29.Casanova JL, Schurr E, Abel L, Skamene E. Forward genetics of infectious diseases: immunological impact. Trends Immunol. 2002;23:469–472. doi: 10.1016/s1471-4906(02)02289-5. [DOI] [PubMed] [Google Scholar]
- 30.Lindenmann J. Inheritance of Resistance to Influenza Virus in Mice. Experimental Biology and Medicine. 1964;116:506–509. doi: 10.3181/00379727-116-29292. [DOI] [PubMed] [Google Scholar]
- 31.Staeheli P, Grob R, Meier E, Sutcliffe JG, 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]
- 32.Tumpey TM, Szretter KJ, Van Hoeven N, Katz JM, Kochs G, Haller O, Garcia-Sastre A, Staeheli P. The Mx1 gene protects mice against the pandemic 1918 and highly lethal human H5N1 influenza viruses. J Virol. 2007;81:10818–10821. doi: 10.1128/JVI.01116-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Deeg C, Mutz P, Rheinemann L, Kallfass C, Nuernberger C, Schilling M, Soubies S, Staeheli P. Transgenic mice expressing interferon-regulated human influenza virus restriction factor MxA. Cytokine. 2014;70:70. [Google Scholar]
- 34•.Haller O, Staeheli P, Schwemmle M, Kochs G. Mx GTPases: dynamin-like antiviral machines of innate immunity. Trends Microbiol. 2015;23:154–163. doi: 10.1016/j.tim.2014.12.003. [Comprehensive review of Mx and the antiviral function of this prototypic influenza resistance gene.] [DOI] [PubMed] [Google Scholar]
- 35.Pavlovic J, Haller O, Staeheli P. Human and mouse Mx proteins inhibit different steps of the influenza virus multiplication cycle. Journal of Virology. 1992;66:2564–2569. doi: 10.1128/jvi.66.4.2564-2569.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Broni B, Julkunen I, Condra JH, Davies ME, Berry MJ, Krug RM. Parental influenza virion nucleocapsids are efficiently transported into the nuclei of murine cells expressing the nuclear interferon-induced Mx protein. Journal of Virology. 1990;64:6335–6340. doi: 10.1128/jvi.64.12.6335-6340.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.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. Journal of Virology. 1985;56:201–206. doi: 10.1128/jvi.56.1.201-206.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pitossi F, Blank A, Schröder A, Schwarz A, Hüssi P, Schwemmle M, Pavlovic J, Staeheli P. A functional GTP-binding motif is necessary for antiviral activity of Mx proteins. Journal of Virology. 1993;67:6726–6732. doi: 10.1128/jvi.67.11.6726-6732.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39•.Riegger D, Hai R, Dornfeld D, Mänz B, Leyva-Grado V, Sánchez-Aparicio MT, Albrecht RA, Palese P, Haller O, Schwemmle M, et al. The Nucleoprotein of Newly Emerged H7N9 Influenza A Virus Harbors a Unique Motif Conferring Resistance to Antiviral Human MxA. Journal of Virology. 2015;89:2241–2252. doi: 10.1128/JVI.02406-14. [This study describes naturally occurring mutations in avian influenza strains affecting human MxA sensitivity.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zimmermann P, Mänz B, Haller O, Schwemmle M, Kochs G. The Viral Nucleoprotein Determines Mx Sensitivity of Influenza A Viruses. Journal of Virology. 2011;85:8133–8140. doi: 10.1128/JVI.00712-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41•.Shin D-L, Hatesuer B, Bergmann S, Nedelko T, Schughart K. Protection from Severe Influenza Virus Infections in Mice Carrying the Mx1 Influenza Virus Resistance Gene Strongly Depends on Genetic Background. Journal of Virology. 2015;89:9998–10009. doi: 10.1128/JVI.01305-15. [This interesting study shows the importance of the mouse strain background during influenza virus infection, independent of the Mx1 genotype. Other cellular factors can affect the response to influenza virus by regulating Mx1 expression or by Mx1-independent pathways.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42•.Ferris MT, Aylor DL, Bottomly D, Whitmore AC, Aicher LD, Bell TA, Bradel-Tretheway B, Bryan JT, Buus RJ, Gralinski LE, et al. Modeling host genetic regulation of influenza pathogenesis in the collaborative cross. PLoS Pathog. 2013;9:e1003196. doi: 10.1371/journal.ppat.1003196. [Influenza virus infection of the incipient lines of the Collaborative Cross identify several candidate genes by quantitative trait locus mapping including a novel Mx1 allele.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Boivin GA, Pothlichet J, Skamene E, Brown EG, Loredo-Osti JC, Sladek R, Vidal SM. Mapping of clinical and expression quantitative trait loci in a sex-dependent effect of host susceptibility to mouse-adapted influenza H3N2/HK/1/68. J Immunol. 2012;188:3949–3960. doi: 10.4049/jimmunol.1103320. [DOI] [PubMed] [Google Scholar]
- 44.Boon A, Williams R, Sinasac D, Webby R. A novel genetic locus linked to pro-inflammatory cytokines after virulent H5N1 virus infection in mice. BMC Genomics. 2014;15:1017. doi: 10.1186/1471-2164-15-1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Gitlin L, Barchet W, Gilfillan S, Cella M, Beutler B, Flavell RA, Diamond MS, Colonna M. Essential role of mda-5 in type I IFN responses to polyriboinosinic:polyribocytidylic acid and encephalomyocarditis picornavirus. Proc Natl Acad Sci U S A. 2006;103:8459–8464. doi: 10.1073/pnas.0603082103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, Uematsu S, Jung A, Kawai T, Ishii KJ, 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]
- 47•.Benitez Asiel A, Panis M, Xue J, Varble A, Shim Jaehee V, Frick Amy L, López Carolina B, Sachs D, tenOever Benjamin R. In Vivo RNAi Screening Identifies MDA5 as a Significant Contributor to the Cellular Defense against Influenza A Virus. Cell Reports. 11:1714–1726. doi: 10.1016/j.celrep.2015.05.032. [Influenza virus infection of the incipient lines of the Collaborative Cross identify several candidate genes by quantitative trait locus mapping including a novel Mx1 allele.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Pulendran B, Maddur MS. Innate Immune Sensing and Response to Influenza. Current topics in microbiology and immunology. 2015;386:23–71. doi: 10.1007/82_2014_405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Koerner I, Kochs G, Kalinke U, Weiss S, Staeheli P. Protective role of beta interferon in host defense against influenza A virus. J Virol. 2007;81:2025–2030. doi: 10.1128/JVI.01718-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.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]
- 51.Lund JM, Alexopoulou L, Sato A, Karow M, Adams NC, Gale NW, Iwasaki A, Flavell RA. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci U S A. 2004;101:5598–5603. doi: 10.1073/pnas.0400937101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Loo YM, Fornek J, Crochet N, Bajwa G, Perwitasari O, Martinez-Sobrido L, Akira S, Gill MA, Garcia-Sastre A, Katze MG, et al. Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity. J Virol. 2008;82:335–345. doi: 10.1128/JVI.01080-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pichlmair A, Schulz O, Tan CP, Naslund TI, Liljestrom P, Weber F, Reis e Sousa C. 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]
- 54.Lafferty EI, Flaczyk A, Angers I, Homer R, d'Hennezel E, Malo D, Piccirillo CA, Vidal SM, Qureshi ST. An ENU-induced splicing mutation reveals a role for Unc93b1 in early immune cell activation following influenza A H1N1 infection. Genes Immun. 2014;15:320–332. doi: 10.1038/gene.2014.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Le Goffic R, Balloy V, Lagranderie M, Alexopoulou L, Escriou N, Flavell R, Chignard M, Si-Tahar M. Detrimental contribution of the Toll-like receptor (TLR)3 to influenza A virus-induced acute pneumonia. PLoS pathogens. 2006;2:e53. doi: 10.1371/journal.ppat.0020053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Koyama S, Ishii KJ, Kumar H, Tanimoto T, Coban C, Uematsu S, Kawai T, Akira S. Differential Role of TLR- and RLR-Signaling in the Immune Responses to Influenza A Virus Infection and Vaccination. The Journal of Immunology. 2007;179:4711–4720. doi: 10.4049/jimmunol.179.7.4711. [DOI] [PubMed] [Google Scholar]
- 57.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. Journal of Virology. 2012;86:10988–10998. doi: 10.1128/JVI.01064-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Seo S-U, Kwon H-J, Song J-H, Byun Y-H, Seong BL, Kawai T, Akira S, Kweon M-N. MyD88 Signaling Is Indispensable for Primary Influenza A Virus Infection but Dispensable for Secondary Infection. Journal of Virology. 2010;84:12713–12722. doi: 10.1128/JVI.01675-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pang IK, Pillai PS, Iwasaki A. Efficient influenza A virus replication in the respiratory tract requires signals from TLR7 and RIG-I. Proceedings of the National Academy of Sciences. 2013;110:13910–13915. doi: 10.1073/pnas.1303275110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60••.Kaminski MM, Ohnemus A, Cornitescu M, Staeheli P. Plasmacytoid dendritic cells and Toll-like receptor 7-dependent signalling promote efficient protection of mice against highly virulent influenza A virus. The Journal of general virology. 2012;93:555–559. doi: 10.1099/vir.0.039065-0. [A clear demonstration of the importance of Mx deficiency in most inbred mouse strains, this article shows the susceptibility of TLR7 and MyD88 knockout mice to influenza in an Mx1-positive mouse strain.] [DOI] [PubMed] [Google Scholar]
- 61.Wilk E, Pandey AK, Leist SR, Hatesuer B, Preusse M, Pommerenke C, Wang J, Schughart K. RNAseq expression analysis of resistant and susceptible mice after influenza A virus infection identifies novel genes associated with virus replication and important for host resistance to infection. BMC Genomics. 2015;16:655. doi: 10.1186/s12864-015-1867-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62•.Schoggins JW, MacDuff DA, Imanaka N, Gainey MD, Shrestha B, Eitson JL, Mar KB, Richardson RB, Ratushny AV, Litvak V, et al. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature. 2014;505:691–695. doi: 10.1038/nature12862. [This interesting study shows the importance of the mouse strain background during influenza virus infection, independent of the Mx1 genotype. Other cellular factors can affect the response to influenza virus by regulating Mx1 expression or by Mx1-independent pathways.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Garcia-Sastre A, Durbin RK, Zheng H, Palese P, Gertner R, Levy DE, Durbin JE. The role of interferon in influenza virus tissue tropism. J Virol. 1998;72:8550–8558. doi: 10.1128/jvi.72.11.8550-8558.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Tenoever BR, Ng SL, Chua MA, McWhirter SM, Garcia-Sastre A, Maniatis T. Multiple functions of the IKK-related kinase IKKepsilon in interferon-mediated antiviral immunity. Science. 2007;315:1274–1278. doi: 10.1126/science.1136567. [DOI] [PubMed] [Google Scholar]
- 65.Lenschow DJ, Lai C, Frias-Staheli N, Giannakopoulos NV, Lutz A, Wolff T, Osiak A, Levine B, Schmidt RE, Garcia-Sastre A, et al. IFN-stimulated gene 15 functions as a critical antiviral molecule against influenza, herpes, and Sindbis viruses. Proc Natl Acad Sci U S A. 2007;104:1371–1376. doi: 10.1073/pnas.0607038104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Durfee LA, Lyon N, Seo K, Huibregtse JM. The ISG15 Conjugation System Broadly Targets Newly Synthesized Proteins: Implications for the Antiviral Function of ISG15. Molecular Cell. 2010;38:722–732. doi: 10.1016/j.molcel.2010.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Morales DJ, Monte K, Sun L, Struckhoff JJ, Agapov E, Holtzman MJ, Stappenbeck TS, Lenschow DJ. Novel Mode of ISG15-Mediated Protection against Influenza A Virus and Sendai Virus in Mice. Journal of Virology. 2015;89:337–349. doi: 10.1128/JVI.02110-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Balachandran S, Roberts PC, Brown LE, Truong H, Pattnaik AK, Archer DR, Barber GN. Essential Role for the dsRNA-Dependent Protein Kinase PKR in Innate Immunity to Viral Infection. Immunity. 2000;13:129–141. doi: 10.1016/s1074-7613(00)00014-5. [DOI] [PubMed] [Google Scholar]
- 69.Schulz O, Pichlmair A, Rehwinkel J, Rogers NC, Scheuner D, Kato H, Takeuchi O, Akira S, Kaufman RJ, Reis e Sousa C. Protein Kinase R Contributes to Immunity against Specific Viruses by Regulating Interferon mRNA Integrity. Cell Host & Microbe. 2010;7:354–361. doi: 10.1016/j.chom.2010.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bergmann M, Garcia-Sastre A, Carnero E, Pehamberger H, Wolff K, Palese P, Muster T. Influenza virus NS1 protein counteracts PKR-mediated inhibition of replication. J Virol. 2000;74:6203–6206. doi: 10.1128/jvi.74.13.6203-6206.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Aebi M, Fäh J, Hurt N, Samuel CE, Thomis D, Bazzigher L, Pavlovic J, Haller O, Staeheli P. cDNA structures and regulation of two interferon-induced human Mx proteins. Molecular and Cellular Biology. 1989;9:5062–5072. doi: 10.1128/mcb.9.11.5062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Horisberger MA, McMaster GK, Zeller H, Wathelet MG, Dellis J, Content J. Cloning and sequence analyses of cDNAs for interferon- and virus-induced human Mx proteins reveal that they contain putative guanine nucleotide-binding sites: functional study of the corresponding gene promoter. J Virol. 1990;64:1171–1181. doi: 10.1128/jvi.64.3.1171-1181.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Staeheli P, Pavlovic J. Inhibition of vesicular stomatitis virus mRNA synthesis by human MxA protein. Journal of Virology. 1991;65:4498–4501. doi: 10.1128/jvi.65.8.4498-4501.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhao H, De BP, Das T, Banerjee AK. Inhibition of Human Parainfluenza Virus-3 Replication by Interferon and Human MxA. Virology. 1996;220:330–338. doi: 10.1006/viro.1996.0321. [DOI] [PubMed] [Google Scholar]
- 75.Yu Z, Wang Z, Chen J, Li H, Lin Z, Zhang F, Zhou Y, Hou J. GTPase activity is not essential for the interferon-inducible MxA protein to inhibit the replication of hepatitis B virus. Archives of Virology. 2008;153:1677–1684. doi: 10.1007/s00705-008-0168-9. [DOI] [PubMed] [Google Scholar]
- 76.Li N, Zhang L, Chen L, Feng W, Xu Y, Chen F, Liu X, Chen Z, Liu W. MxA inhibits hepatitis B virus replication by interaction with hepatitis B core antigen. Hepatology. 2012;56:803–811. doi: 10.1002/hep.25608. [DOI] [PubMed] [Google Scholar]
- 77.Kochs G, Janzen C, Hohenberg H, Haller O. Antivirally active MxA protein sequesters La Crosse virus nucleocapsid protein into perinuclear complexes. Proceedings of the National Academy of Sciences. 2002;99:3153–3158. doi: 10.1073/pnas.052430399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Netherton CL, Simpson J, Haller O, Wileman TE, Takamatsu H-H, Monaghan P, Taylor G. Inhibition of a Large Double-Stranded DNA Virus by MxA Protein. Journal of Virology. 2009;83:2310–2320. doi: 10.1128/JVI.00781-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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. Journal of Virology. 2013;87:13053–13058. doi: 10.1128/JVI.02220-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Turan K, Mibayashi M, Sugiyama K, Saito S, Numajiri A, Nagata K. Nuclear MxA proteins form a complex with influenza virus NP and inhibit the transcription of the engineered influenza virus genome. Nucleic Acids Research. 2004;32:643–652. doi: 10.1093/nar/gkh192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Holzinger D, Jorns C, Stertz S, Boisson-Dupuis S, Thimme R, Weidmann M, Casanova J-L, Haller O, Kochs G. Induction of MxA Gene Expression by Influenza A Virus Requires Type I or Type III Interferon Signaling. Journal of Virology. 2007;81:7776–7785. doi: 10.1128/JVI.00546-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mibayashi M, Nakade K, Nagata K. Promoted Cell Death of Cells Expressing Human MxA by Influenza Virus Infection. Microbiology and Immunology. 2002;46:29–36. doi: 10.1111/j.1348-0421.2002.tb02673.x. [DOI] [PubMed] [Google Scholar]
- 83.Li Y, Youssoufian H. MxA overexpression reveals a common genetic link in four Fanconi anemia complementation groups. The Journal of Clinical Investigation. 1997;100:2873–2880. doi: 10.1172/JCI119836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84••.Dittmann M, Hoffmann HH, Scull MA, Gilmore RH, Bell KL, Ciancanelli M, Wilson SJ, Crotta S, Yu Y, Flatley B, et al. A serpin shapes the extracellular environment to prevent influenza a virus maturation. Cell. 2015;160:631–643. doi: 10.1016/j.cell.2015.01.040. [A lentiviral screen of ISGs identified SERPINE1 as a novel ISG capable of inhibiting a late stage in IAV replication in in vitro cellular models, in vivo mouse models and in human cells with diminished gene expression.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, Rice CM. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472:481–485. doi: 10.1038/nature09907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Shapira SD, Gat-Viks I, Shum BO, Dricot A, de Grace MM, Wu L, Gupta PB, Hao T, Silver SJ, Root DE, et al. A physical and regulatory map of host-influenza interactions reveals pathways in H1N1 infection. Cell. 2009;139:1255–1267. doi: 10.1016/j.cell.2009.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Konig R, Stertz S, Zhou Y, Inoue A, Hoffmann HH, Bhattacharyya S, Alamares JG, Tscherne DM, Ortigoza MB, Liang Y, et al. Human host factors required for influenza virus replication. Nature. 2010;463:813–817. doi: 10.1038/nature08699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Karlas A, Machuy N, Shin Y, Pleissner KP, Artarini A, Heuer D, Becker D, Khalil H, Ogilvie LA, Hess S, et al. Genome-wide RNAi screen identifies human host factors crucial for influenza virus replication. Nature. 2010;463:818–822. doi: 10.1038/nature08760. [DOI] [PubMed] [Google Scholar]
- 89•.Pulloor NK, Nair S, McCaffrey K, Kostic AD, Bist P, Weaver JD, Riley AM, Tyagi R, Uchil PD, York JD, et al. Human genome-wide RNAi screen identifies an essential role for inositol pyrophosphates in Type-I interferon response. PLoS Pathog. 2014;10:e1003981. doi: 10.1371/journal.ppat.1003981. [This study employed an RNAi screen to determine regulators of RIG-I induced IFN-β production and discovered that the cellular pathway synthesizing inositol pyrophosphates governs IRF3 activation downstream of RIG-I.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rehwinkel J, Tan CP, Goubau D, Schulz O, Pichlmair A, Bier K, Robb N, Vreede F, Barclay W, Fodor E, et al. RIG-I Detects Viral Genomic RNA during Negative-Strand RNA Virus Infection. Cell. 2010;140:397–408. doi: 10.1016/j.cell.2010.01.020. [DOI] [PubMed] [Google Scholar]
- 91.Winterling C, Koch M, Koeppel M, Garcia-Alcalde F, Karlas A, Meyer TF. Evidence for a crucial role of a host non-coding RNA in influenza A virus replication. RNA Biol. 2014;11:66–75. doi: 10.4161/rna.27504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Meliopoulos VA, Andersen LE, Birrer KF, Simpson KJ, Lowenthal JW, Bean AG, Stambas J, Stewart CR, Tompkins SM, van Beusechem VW, et al. Host gene targets for novel influenza therapies elucidated by high-throughput RNA interference screens. FASEB J. 2012;26:1372–1386. doi: 10.1096/fj.11-193466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.de Chassey B, Meyniel-Schicklin L, Aublin-Gex A, Andre P, Lotteau V. Genetic screens for the control of influenza virus replication: from meta-analysis to drug discovery. Molecular BioSystems. 2012;8:1297–1303. doi: 10.1039/c2mb05416g. [DOI] [PubMed] [Google Scholar]
- 94.Chou YC, Lai MM, Wu YC, Hsu NC, Jeng KS, Su WC. Variations in genome-wide RNAi screens: lessons from influenza research. J Clin Bioinforma. 2015;5:2. doi: 10.1186/s13336-015-0017-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, Feeley EM, Ryan BJ, Weyer JL, van der Weyden L, Fikrig E, et al. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell. 2009;139:1243–1254. doi: 10.1016/j.cell.2009.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yount JS, Moltedo B, Yang Y-Y, Charron G, Moran TM, López CB, Hang HC. Palmitoylome profiling reveals S-palmitoylation–dependent antiviral activity of IFITM3. Nat Chem Biol. 2010;6:610–614. doi: 10.1038/nchembio.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Yount JS, Karssemeijer RA, Hang HC. S-Palmitoylation and Ubiquitination Differentially Regulate Interferon-induced Transmembrane Protein 3 (IFITM3)-mediated Resistance to Influenza Virus. Journal of Biological Chemistry. 2012;287:19631–19641. doi: 10.1074/jbc.M112.362095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chesarino NM, McMichael TM, Yount JS. E3 Ubiquitin Ligase NEDD4 Promotes Influenza Virus Infection by Decreasing Levels of the Antiviral Protein IFITM3. PLoS Pathog. 2015;11:e1005095. doi: 10.1371/journal.ppat.1005095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.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]
- 100.Everitt AR, Clare S, Pertel T, John SP, Wash RS, Smith SE, Chin CR, Feeley EM, Sims JS, Adams DJ, et al. IFITM3 restricts the morbidity and mortality associated with influenza. Nature. 484:519–523. doi: 10.1038/nature10921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Zhang YH, Zhao Y, Li N, Peng YC, Giannoulatou E, Jin RH, Yan HP, Wu H, Liu JH, Liu N, 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]
- 102.An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491:56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mills TC, Rautanen A, Elliott KS, Parks T, Naranbhai V, Ieven MM, Butler CC, Little P, Verheij T, Garrard CS, et al. IFITM3 and susceptibility to respiratory viral infections in the community. J Infect Dis. 2014;209:1028–1031. doi: 10.1093/infdis/jit468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Williams DE, Wu WL, Grotefend CR, Radic V, Chung C, Chung YH, Farzan M, Huang IC. IFITM3 polymorphism rs12252-C restricts influenza A viruses. PLoS One. 2014;9:e110096. doi: 10.1371/journal.pone.0110096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105••.To KK-W, Zhou J, Chan JF-W, Yuen K-Y. Host genes and influenza pathogenesis in humans: an emerging paradigm. Current Opinion in Virology. 2015;14:7–15. doi: 10.1016/j.coviro.2015.04.010. [Excellent broad review of human host genetics in severe influenza with an emphasis on GWAS studies.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zhou J, To KK, Dong H, Cheng ZS, Lau CC, Poon VK, Fan YH, Song YQ, Tse H, Chan KH, et al. A functional variation in CD55 increases the severity of 2009 pandemic H1N1 influenza A virus infection. J Infect Dis. 2012;206:495–503. doi: 10.1093/infdis/jis378. [DOI] [PubMed] [Google Scholar]
- 107.Cheng Z, Zhou J, To KK, Chu H, Li C, Wang D, Yang D, Zheng S, Hao K, Bosse Y, et al. Identification of TMPRSS2 as a Susceptibility Gene for Severe 2009 Pandemic A(H1N1) Influenza and A(H7N9) Influenza. J Infect Dis. 2015 doi: 10.1093/infdis/jiv246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hatesuer B, Bertram S, Mehnert N, Bahgat MM, Nelson PS, Pohlmann S, Schughart K. Tmprss2 is essential for influenza H1N1 virus pathogenesis in mice. PLoS Pathog. 2013;9:e1003774. doi: 10.1371/journal.ppat.1003774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Horby P, Nguyen NY, Dunstan SJ, Baillie JK. The role of host genetics in susceptibility to influenza: a systematic review. PLoS One. 2012;7:e33180. doi: 10.1371/journal.pone.0033180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110•.Horby P, Nguyen NY, Dunstan SJ, Baillie JK. An updated systematic review of the role of host genetics in susceptibility to influenza. Influenza Other Respir Viruses. 2013;7(Suppl 2):37–41. doi: 10.1111/irv.12079. [Comprehensive review on recent advances in determining the genetic basic of susceptibility to influenza.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Yoav K, Jennifer J, Adrienne M, Ball TB, Anand K, Ethan R, Keith RF. Chemokine Receptor 5 Δ32 Allele in Patients with Severe Pandemic (H1N1) 2009. Emerging Infectious Disease journal. 2010;16:1621. doi: 10.3201/eid1610.100108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Sironi M, Cagliani R, Pontremoli C, Rossi M, Migliorino G, Clerici M, Gori A. The CCR5Delta32 allele is not a major predisposing factor for severe H1N1pdm09 infection. BMC Research Notes. 2014;7:504. doi: 10.1186/1756-0500-7-504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Herrera-Ramos E, Lopez-Rodriguez M, Ruiz-Hernandez JJ, Horcajada JP, Borderias L, Lerma E, Blanquer J, Perez-Gonzalez MC, Garcia-Laorden MI, Florido Y, et al. 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]
- 114.To KK, Zhou J, Song YQ, Hung IF, Ip WC, Cheng ZS, Chan AS, Kao RY, Wu AK, Chau S, et al. 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]
- 115.Mak CM, Lam C-w, Fong N-c, Siu W-k, Lee H-cH, Siu T-s, Lai C-k, Law C-y, Tong S-f, Poon W-t, et al. Fatal viral infection-associated encephalopathy in two Chinese boys: a genetically determined risk factor of thermolabile carnitine palmitoyltransferase II variants. J Hum Genet. 2011;56:617–621. doi: 10.1038/jhg.2011.63. [DOI] [PubMed] [Google Scholar]
- 116.Yao D, Mizuguchi H, Yamaguchi M, Yamada H, Chida J, Shikata K, Kido H. Thermal instability of compound variants of carnitine palmitoyltransferase II and impaired mitochondrial fuel utilization in influenza-associated encephalopathy. Human Mutation. 2008;29:718–727. doi: 10.1002/humu.20717. [DOI] [PubMed] [Google Scholar]
- 117.Casanova JL, Conley ME, Seligman SJ, Abel L, Notarangelo LD. Guidelines for genetic studies in single patients: lessons from primary immunodeficiencies. Journal of Experimental Medicine. 2014 doi: 10.1084/jem.20140520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Chakravarti A, Clark AG, Mootha VK. Distilling Pathophysiology from Complex Disease Genetics. Cell. 155:21–26. doi: 10.1016/j.cell.2013.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Gennery AR, Cant AJ. Diagnosis of severe combined immunodeficiency. Journal of Clinical Pathology. 2001;54:191–195. doi: 10.1136/jcp.54.3.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Al-Herz W, Bousfiha A, Casanova J-L, Chatila T, Conley ME, Cunningham-Rundles C, Etzioni A, Franco JL, Gaspar HB, Holland S, et al. Primary immunodeficiency diseases: an update on the classification from the International Union of Immunological Societies Expert Committee for Primary Immunodeficiency. Frontiers in Immunology. 2014;5 doi: 10.3389/fimmu.2014.00162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Bigley V, Haniffa M, Doulatov S, Wang XN, Dickinson R, McGovern N, Jardine L, Pagan S, Dimmick I, Chua I, et al. The human syndrome of dendritic cell, monocyte, B and NK lymphoid deficiency. J Exp Med. 2011;208:227–234. doi: 10.1084/jem.20101459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Pasquet M, Bellanne-Chantelot C, Tavitian S, Prade N, Beaupain B, Larochelle O, Petit A, Rohrlich P, Ferrand C, Van Den Neste E, et al. High frequency of GATA2 mutations in patients with mild chronic neutropenia evolving to MonoMac syndrome, myelodysplasia, and acute myeloid leukemia. Blood. 2013;121:822–829. doi: 10.1182/blood-2012-08-447367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Boisson-Dupuis S, El Baghdadi J, Parvaneh N, Bousfiha A, Bustamante J, Feinberg J, Samarina A, Grant AV, Janniere L, El Hafidi N, et al. IL-12Rbeta1 deficiency in two of fifty children with severe tuberculosis from Iran, Morocco, and Turkey. PloS one. 2011;6:e18524. doi: 10.1371/journal.pone.0018524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Casanova JL, Abel L, Quintana-Murci L. Human TLRs and IL-1Rs in host defense: natural insights from evolutionary, epidemiological, and clinical genetics. Annual review of immunology. 2011;29:447–491. doi: 10.1146/annurev-immunol-030409-101335. [DOI] [PubMed] [Google Scholar]
- 125.Bogunovic D, Byun M, Durfee LA, Abhyankar A, Sanal O, Mansouri D, Salem S, Radovanovic I, Grant AV, Adimi P, et al. Mycobacterial disease and impaired IFN-gamma immunity in humans with inherited ISG15 deficiency. Science. 2012;337:1684–1688. doi: 10.1126/science.1224026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zhang X, Bogunovic D, Payelle-Brogard B, Francois-Newton V, Speer SD, Yuan C, Volpi S, Li Z, Sanal O, Mansouri D, et al. Human intracellular ISG15 prevents interferon-[agr]/[bgr] over-amplification and auto-inflammation. Nature. 2015;517:89–93. doi: 10.1038/nature13801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Neilson DE. The interplay of infection and genetics in acute necrotizing encephalopathy. Curr Opin Pediatr. 2010;22:751–757. doi: 10.1097/MOP.0b013e3283402bfe. [DOI] [PubMed] [Google Scholar]
- 128.Neilson DE, Adams MD, Orr CM, Schelling DK, Eiben RM, Kerr DS, Anderson J, Bassuk AG, Bye AM, Childs AM, et al. Infection-triggered familial or recurrent cases of acute necrotizing encephalopathy caused by mutations in a component of the nuclear pore, RANBP2. Am J Hum Genet. 2009;84:44–51. doi: 10.1016/j.ajhg.2008.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Pichler A, Gast A, Seeler JS, Dejean A, Melchior F. The nucleoporin RanBP2 has SUMO1 E3 ligase activity. Cell. 2002;108:109–120. doi: 10.1016/s0092-8674(01)00633-x. [DOI] [PubMed] [Google Scholar]
- 130••.Ciancanelli MJ, Huang SX, Luthra P, Garner H, Itan Y, Volpi S, Lafaille FG, Trouillet C, Schmolke M, Albrecht RA, et al. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency. Science. 2015;348:448–453. doi: 10.1126/science.aaa1578. [This study provides clear evidence of genetic susceptibility to severe influenza with whole exome sequencing-mediated discovery of IRF7 mutations in a child with life-threatening influenza.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Izaguirre A, Barnes BJ, Amrute S, Yeow W-S, Megjugorac N, Dai J, Feng D, Chung E, Pitha PM, Fitzgerald-Bocarsly P. Comparative analysis of IRF and IFN-alpha expression in human plasmacytoid and monocyte-derived dendritic cells. Journal of Leukocyte Biology. 2003;74:1125–1138. doi: 10.1189/jlb.0603255. [DOI] [PubMed] [Google Scholar]
- 132.Kerkmann M, Rothenfusser S, Hornung V, Towarowski A, Wagner M, Sarris A, Giese T, Endres S, Hartmann G. Activation with CpG-A and CpG-B Oligonucleotides Reveals Two Distinct Regulatory Pathways of Type I IFN Synthesis in Human Plasmacytoid Dendritic Cells. The Journal of Immunology. 2003;170:4465–4474. doi: 10.4049/jimmunol.170.9.4465. [DOI] [PubMed] [Google Scholar]
- 133.Honda K, Yanai H, Negishi H, Asagiri M, Sato M, Mizutani T, Shimada N, Ohba Y, Takaoka A, Yoshida N, 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]
- 134.Spann KM, Loh Z, Lynch JP, Ullah A, Zhang V, Baturcam E, Werder RB, Khajornjiraphan N, Rudd P, Loo Y-M, et al. IRF-3, IRF-7, and IPS-1 Promote Host Defense against Acute Human Metapneumovirus Infection in Neonatal Mice. The American Journal of Pathology. 2014;184:1795–1806. doi: 10.1016/j.ajpath.2014.02.026. [DOI] [PubMed] [Google Scholar]
- 135.Crotta S, Davidson S, Mahlakoiv T, Desmet CJ, Buckwalter MR, Albert ML, Staeheli P, Wack A. 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]
- 136.Chen H-W, King K, Tu J, Sanchez M, Luster AD, Shresta S. The Roles of IRF-3 and IRF-7 in Innate Antiviral Immunity against Dengue Virus. The Journal of Immunology. 2013;191:4194–4201. doi: 10.4049/jimmunol.1300799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Li W, Hofer MJ, Noçon AL, Manders P, Campbell IL. Interferon regulatory factor 7 (IRF7) is required for the optimal initial control but not subsequent clearance of lymphocytic choriomeningitis virus infection in mice. Virology. 2013;439:152–162. doi: 10.1016/j.virol.2013.02.015. [DOI] [PubMed] [Google Scholar]
- 138.Schilte C, Buckwalter MR, Laird ME, Diamond MS, Schwartz O, Albert ML. Cutting Edge: Independent Roles for IRF-3 and IRF-7 in Hematopoietic and Nonhematopoietic Cells during Host Response to Chikungunya Infection. The Journal of Immunology. 2012;188:2967–2971. doi: 10.4049/jimmunol.1103185. [DOI] [PubMed] [Google Scholar]
- 139.Levy DE, Marie I, Smith E, Prakash A. Enhancement and diversification of IFN induction by IRF-7-mediated positive feedback. J Interferon Cytokine Res. 2002;22:87–93. doi: 10.1089/107999002753452692. [DOI] [PubMed] [Google Scholar]
- 140.Marie I, Durbin JE, Levy DE. Differential viral induction of distinct interferon-alpha genes by positive feedback through interferon regulatory factor-7. EMBO J. 1998;17:6660–6669. doi: 10.1093/emboj/17.22.6660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jewell NA, Cline T, Mertz SE, Smirnov SV, Flaño E, Schindler C, Grieves JL, Durbin RK, Kotenko SV, Durbin JE. Lambda Interferon Is the Predominant Interferon Induced by Influenza A Virus Infection In Vivo. Journal of Virology. 2010;84:11515–11522. doi: 10.1128/JVI.01703-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, Taya C, Taniguchi T. Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature. 2005;434:1035–1040. doi: 10.1038/nature03547. [DOI] [PubMed] [Google Scholar]
- 143.Honda K, Taniguchi T. IRFs: master regulators of signalling by Toll-like receptors and cytosolic pattern-recognition receptors. Nature reviews. Immunology. 2006;6:644–658. doi: 10.1038/nri1900. [DOI] [PubMed] [Google Scholar]
- 144.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]
- 145.Colina R, Costa-Mattioli M, Dowling RJ, Jaramillo M, Tai LH, Breitbach CJ, Martineau Y, Larsson O, Rong L, Svitkin YV, 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]
- 146•.Lee MN, Ye C, Villani A-C, Raj T, Li W, Eisenhaure TM, Imboywa SH, Chipendo PI, Ran FA, Slowikowski K, et al. Common Genetic Variants Modulate Pathogen-Sensing Responses in Human Dendritic Cells. Science. 2014;343 doi: 10.1126/science.1246980. [This study finely mapped SNPs associated with the variation in antiviral response in human dendritic cells infected with influenza and detected a common variant in IRF7 associated with the IAV-induced upregulation of several IFN genes.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Casanova JL, Abel L, Quintana-Murci L. Immunology taught by human genetics. Cold Spring Harb Symp Quant Biol. 2013;78:157–172. doi: 10.1101/sqb.2013.78.019968. [DOI] [PubMed] [Google Scholar]
